You are on page 1of 1143

Barkovich_FM.

indd i 5/25/2011 11:31:48 AM


Executive Editor: Charles W. Mitchell
Product Manager: Ryan Shaw
Vendor Manager: Alicia Jackson
Senior Manufacturing Manager: Benjamin Rivera
Senior Marketing Manager: Angela Panetta
Design Coordinator: Stephen Druding
Production Service: SPi Global

© 2012 by LIPPINCOTT WILLIAMS & WILKINS, a WOLTERS KLUWER business


Two Commerce Square
2001 Market Street
Philadelphia, PA 19103 USA
LWW.com

All rights reserved. This book is protected by copyright. No part of this book may be reproduced in
any form by any means, including photocopying, or utilized by any information storage and retrieval
system without written permission from the copyright owner, except for brief quotations embodied in
critical articles and reviews. Materials appearing in this book prepared by individuals as part of their
official duties as U.S. government employees are not covered by the above-mentioned copyright.

Printed in China

Library of Congress Cataloging-in-Publication Data


Barkovich, A. James, 1952- author.
Pediatric neuroimaging / A. James Barkovich, Charles Raybaud, charles.raybaud@sickkids. — Fifth
Edition.
p. ; cm.
Includes bibliographical references and index.
ISBN 978-1-60547-714-5 (alk. paper)
1. Nervous system—Magnetic resonance imaging. 2. Nervous system—Ultrasonic imaging.
3. Pediatric diagnostic imaging. 4. Pediatric neurology—Diagnosis. I. Raybaud, C. (Charles), author.
II. Title.
[DNLM: 1. Central Nervous System Diseases—diagnosis. 2. Child. 3. Infant. 4. Magnetic
Resonance Imaging. 5. Tomography, X-Ray Computed. WS 340]
RJ488.5.M33B37 2012
618.92’8047548—dc22
2010049611

Care has been taken to confirm the accuracy of the information presented and to describe gener-
ally accepted practices. However, the authors, editors, and publisher are not responsible for errors or
omissions or for any consequences from application of the information in this book and make no
warranty, expressed or implied, with respect to the currency, completeness, or accuracy of the con-
tents of the publication. Application of the information in a particular situation remains the profes-
sional responsibility of the practitioner.
The authors, editors, and publisher have exerted every effort to ensure that drug selection and
dosage set forth in this text are in accordance with current recommendations and practice at the time
of publication. However, in view of ongoing research, changes in government regulations, and the
constant flow of information relating to drug therapy and drug reactions, the reader is urged to check
the package insert for each drug for any change in indications and dosage and for added warnings
and precautions. This is particularly important when the recommended agent is a new or infrequently
employed drug.
Some drugs and medical devices presented in the publication have Food and Drug Administra-
tion (FDA) clearance for limited use in restricted research settings. It is the responsibility of the health
care provider to ascertain the FDA status of each drug or device planned for use in their clinical
practice.

To purchase additional copies of this book, call our customer service department at (800) 638-3030 or
fax orders to (301) 223-2320. International customers should call (301) 223-2300.

Visit Lippincott Williams & Wilkins on the Internet: at LWW.com. Lippincott Williams & Wilkins
customer service representatives are available from 8:30 am to 6 pm, EST.
10 9 8 7 6 5 4 3 2 1

Barkovich_FM.indd ii 5/25/2011 11:31:48 AM


To our families, without whose inspiration none of
this would have been possible.

Barkovich_FM.indd iii 5/25/2011 11:31:48 AM


Barkovich_FM.indd iv 5/25/2011 11:31:48 AM
P R E FAC E TO T H E F I R S T E D I T I O N

ew techniques for pediatric brain and spine diagnoses modalities by far for imaging the pediatric brain. In those areas where
N have rapidly developed over the past ten years. Computed
tomography, ultrasound, and magnetic resonance imaging
ultrasound and plain film radiology are important adjuncts or are of
primary importance in diagnosis, they have been included. Specifi-
have opened a new window to the pediatric central nervous system. cally, this includes the diagnosis of intracranial pathology in prema-
Through the use of these imaging modalities, an increased under- ture infants.
standing of the pathological processes that occur in the pediatric Readers will note that this is not an encyclopedic work on diseases
brain has emerged. However, in spite of the wealth of new concepts of the pediatric central nervous system. Those disease processes that
that have evolved from these new resources, there has been a nota- are well covered in other texts, or are extremely uncommon, are de-
ble lack of textbooks on the subject, particularly in dealing with CT emphasized here. Instead, I have attempted to cover subjects that are
and MR. In this book, I attempt, at lease in part, to fill the gap of encountered in everyday practice. Furthermore, I have emphasized
knowledge that exists in pediatric neuroimaging. concepts that are crucial to proper imaging techniques and image inter-
This book strongly emphasizes CT and MR in pediatric neurodi- pretation. Embryology, normal development, and pathophysiology are
agnosis. The reasons for this are twofold. First, there are a number explained. Once these basic concepts are understood, interpretation of
of good textbooks available that focus on plain film and sonographic images is greatly facilitated. Finally, an attempt was made to present the
evaluation of the pediatric central nervous system. Second, and more information in a concise and straightforward manner that will make
important, I feel that CT and MR, particularly MR, are the best reading this book an enjoyable learning experience.

Barkovich_FM.indd v 5/25/2011 11:31:48 AM


Barkovich_FM.indd vi 5/25/2011 11:31:48 AM
P R E FAC E

he rapid evolution of Pediatric Neuroimaging continues to injury in preterm and encephalopathic term neonates (including birth
T accelerate. New imaging techniques continue to emerge and
improve to the point where they become useful in the imag-
asphyxia), as well as traumatic brain injury (accidental and inflicted,
both of which continue to be topics of hot debate). We have tried to
ing evaluation of children. New diseases continue to be described, give a fair and balanced view of controversial issues.
along with new categories of diseases and new ways to classify them. In Chapter 5 on malformations has been extensively updated with
addition, the diseases are better understood, as a result of advances in newly described disorders, as well as new information, theories, genet-
genetics, molecular biology, biochemistry, and imaging. It has become ics, and classifications. The organization of the chapter has changed; it
difficult to keep up with all of it; thus, for the first time, it no lon- is now organized according to the part of the brain primarily involved
ger seemed feasible for a single author to update the entire book. As by the disease process: (dorsal forebrain [cerebral cortex and com-
a result, the fifth edition of Pediatric Neuroimaging is a collaborative missures], ventral forebrain [base of the brain], midbrain/hindbrain,
effort. Charles Raybaud and James Barkovich have written much of it craniocervical junction, brain coverings) in order to help the reader
and edited the work of seven other authors (Christopher Hess, Pratik locate the disorder within the text; we believe that this will be useful to
Mukherjee, Zoltan Patay, Erin Schwartz, Gilbert Vezina, Gary Hedlund, readers. Chapters 6 (Phakomatoses) and 7 (Brain Tumors) have been
and Phil Meyers), who have graced this edition with information from updated with new genetic information, new classifications, many new
their areas of expertise. The result is a more complete book, which images, and several new disorders being described.
shares the experience and opinions of dedicated pediatric neuroradi- Chapter 8 on hydrocephalus has been significantly modified and
ologists with many decades of experience in many different locations. upgraded to include new theories of hydrocephalus that better explain
Contributions by many authors increase the risk of an uneven text, the effects upon the ventricular system and the underlying brain.
with much overlap, differences in style, and the potential for contra- Chapters 9 (Spine Anomalies), 10 (Spine Tumors), and 12 (Disorders
dictions. The authors, indeed, have different styles, but the book was of Cerebral Blood Vessels) have all been carefully updated with new
edited to minimize those differences and, at the same time, keep the data, new theories, and new images that facilitate the diagnosis and
book readable so that it can be used as a tool for learning, as well as understanding of these disorders. Chapter 11 (Infection) has been
a reference text. In all chapters, the readers will find extensive use of greatly expanded and improved by adding many new disorders and
tables to help to organize the disorders and images to illustrate them. images, in order to expand our understanding of viruses, disorders
Chapter 1 on imaging techniques and Chapter 2 on normal devel- that are prevalent outside of North America and Europe, and newly
opmental features seen by imaging have been updated with attention to described diseases.
new information on limiting exposure to ionizing radiation while dis- Despite the many changes in this edition, we hope the reader will
cussing both more traditional and new imaging techniques (diffusion notice that the philosophy of the book remains the same. A large num-
tractography, arterial spin labeling, and volumetrics) and protocols for ber of disorders are discussed and illustrated, as it is much easier to
MRI. Chapter 2 also has a new section on resting state functional MRI, recognize a disorder by seeing imaging than by reading about imag-
potentially a very interesting and useful technique. ing features. The cause of the disorder, the main clinical features, and
Readers will note that, after Chapter 2, all new techniques have been the underlying cause/pathophysiology are discussed whenever possible
integrated into the discussion of specific disease entities, which is still because it is easier to remember disorders when the genetic or embryo-
organized by category of disease. Thus, fetal images, perfusion studies, logic or destructive cause is understood, rather than trying to match
diffusion imaging, PET images, or proton spectroscopy are illustrated imaging characteristics to a disease name.
when they are useful for diagnosis. For malformations, hydrocepha- For the convenience of the reader, some topics are discussed
lus, and brain injury, in particular, fetal imaging is extremely useful more than once in the text. The purpose of this is to avoid forcing
for early detection of disease. These fetal images show all of the same the reader to page back through the book, trying to find the previ-
features of these disorders that are identified in infants and children ous mention of a disorder. For example, Chiari II malformations are
and can be identified using the same search patterns; there is no reason discussed in Chapter 5, under disorders of the craniocervical junction,
to put them in a separate section. as a brain malformation and also in Chapter 9 under myelomenin-
Chapter 3 on metabolic diseases has been expanded to include many goceles because they are almost always associated. Within Chapter 5,
newly described disorders and groups of disorders. It also includes a new disorders secondary to abnormal pial basement membrane formation
section on genetic disorders with extensive cerebellar involvement (par- are discussed in both the dorsal forebrain section and the midbrain/
ticularly cerebellar hypoplasia and cerebellar atrophy) as the major imag- hindbrain section because both regions are variably involved in the
ing finding. In addition, new information on diffusivity, spectroscopy, pathologic process, such that they might present as a forebrain or a
and clinical/genetic manifestations has been added. We have retained the hindbrain malformation.
two section approach, with the initial section having a brief, diagnosis- We hope that this new edition of Pediatric Neuroimaging will serve
oriented discussion and the longer second section giving a more detailed as a textbook for residents, fellows, and practicing physicians who are
explanation of the disorders and their imaging manifestations. interested in diseases of the pediatric brain and spine while, at the same
Chapter 4 on destructive lesions of the brain and spine includes time, serving as a reference book for clinicians seeing patients with
new information on imaging of pediatric stroke, imaging of brain these diseases in their daily work.

vii

Barkovich_FM.indd vii 5/25/2011 11:31:48 AM


Barkovich_FM.indd viii 5/25/2011 11:31:49 AM
CONTENTS

Preface to the First Edition v


Preface vii
List of Disorders xi
Contributors xvii

1. Techniques and Methods in Pediatric Neuroimaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1


CHRISTOPHER P. HESS AND A. JAMES BARKOVICH

2. Normal Development of the Neonatal and Infant Brain, Skull, and Spine . . . . . . . . . . . . . . . . . 20
A. JAMES BARKOVICH AND PRATIK MUKHERJEE

3. Metabolic, Toxic, and Inflammatory Brain Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81


A. JAMES BARKOVICH AND ZOLTAN PATAY

4. Brain and Spine Injuries in Infancy and Childhood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 240


ERIN SIMON SCHWARTZ AND A. JAMES BARKOVICH

5. Congenital Malformations of the Brain and Skull . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367


A. JAMES BARKOVICH AND CHARLES A. RAYBAUD

6. The Phakomatoses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 569


GILBERT VEZINA AND A. JAMES BARKOVICH

7. Intracranial, Orbital, and Neck Masses of Childhood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 637


CHARLES RAYBAUD AND A. JAMES BARKOVICH

8. Hydrocephalus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 808
A. RAYBAUD AND A. JAMES BARKOVICH

9. Congenital Anomalies of the Spine. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 857


ERIN SIMON SCHWARTZ AND A. JAMES BARKOVICH

10. Neoplasms of the Spine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 923


A. JAMES BARKOVICH

11. Infections of the Developing and Mature Nervous System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 954


GARY HEDLUND, JAMES F. BALE, JR, AND A. JAMES BARKOVICH

12. Anomalies of Cerebral Vasculature: Diagnostic and Endovascular Considerations . . . . . . . . 1051


PHILIP M. MEYERS, VAN V. HALBACH, AND A. JAMES BARKOVICH

Index 1109

ix

Barkovich_FM.indd ix 5/25/2011 11:31:49 AM


Barkovich_FM.indd x 5/25/2011 11:31:49 AM
LIST OF DISORDERS

CHAPTER 3: METABOLIC, TOXIC, AND INFLAMMATORY 4. White matter diseases with nonspecific patterns
BRAIN DISORDERS a. Nonketotic hyperglycinemia (glycine encephalopathy)
IV.B. Metabolic Disorders Primarily Affecting White b. Dihydropyrimidine dehydrogenase deficiency
Matter c. 3-Hydroxy-3-methylglutaryl-coenzyme A lyase deficiency
1. White matter diseases initially affecting periventricular cere- d. Congenital white matter hypoplasia/familial spastic
bral white matter paraplegia
a. Metachromatic leukodystrophy 5 Idiopathic inflammatory, autoimmune, infectious, and toxic
b. Globoid cell leukodystrophy (Krabbe disease) disorders affecting white matter
c. Classic X-linked adrenal leukodystrophy/adrenomy- a. Multiple sclerosis
eloneuropathy/acyl-CoA-oxidase deficiency b. Neuromyelitis optica (Devic disease)
d. Leukoencephalopathy with vanishing white matter c. Acute disseminated encephalomyelitis (ADEM)
e. Giant axonal neuropathy d. Acute hemorrhagic encephalomyelitis
f. Phenylketonuria e. Collagen vascular diseases/systemic lupus erythematosus
g. Maple syrup urine disease f. Osmotic myelinolysis in childhood
h. Hyperhomocysteinemia (formerly known as homocysti- g. Toxins
nuria) h. Lead encephalopathy
i. Cystathionine beta synthase deficiency i. Solvent abuse
j. 5, 10 methylenetetrahydrofolate reductase deficiency j. Progressive multifocal leukoencephalitis
(MTHFRD) IV.C. Metabolic Disorders Primarily Involving
k. Errors affecting cobalamin (vitamin B12) metabolism Gray Matter
l. Biotinidase deficiency 1. Gray matter disorders primarily involving the deep gray
m. Methionine adenosyltransferase deficiency cerebral nuclei
n. Oculocerebrorenal syndrome (Lowe syndrome) a. Pantothenate kinase-associated neuropathy (neurode-
o. Merosin-deficient congenital muscular dystrophy generation with brain iron accumulation 1, formerly
(MDC1A) Hallervorden-Spatz disease)
p. Mucolipidosis type IV b. Juvenile Huntington disease
q. Autosomal recessive spastic paraplegia with thin corpus c. Isovaleric acidemia
callosum d. Succinic semialdehyde dehydrogenase deficiency
r. Sjögren-Larsson syndrome e. Creatine deficiency syndromes
s. Brain injury from radiation and chemotherapy f. Wernicke encephalopathy
2. White matter disorders with dysmyelination initially affect- g. Extrapontine myelinolysis
ing subcortical cerebral white matter h. Hemolytic-uremic syndrome
a. Megalencephalic leukoencephalopathy with subcortical i. Sydenham chorea
cysts (MLC) j. Chronic liver disease
b. Cystic leukoencephalopathy without megalencephaly 2. Gray matter disorders primarily involving cortex
c. Aicardi-Goutières syndrome a. Neuronal ceroid lipofuscinosis
d. Cockayne syndrome b. Aspartylglucosaminuria
e. Galactosemia c. Anti-N-methyl-D-aspartate receptor (NMDAR) encepha-
3. White matter disorders due to hypomyelination litis in children and adolescents
(hypomyelinating leukodystrophies) d. Infantile neuroaxonal dystrophy
a. Pelizaeus-Merzbacher disease e. Niemann-Pick disease
b. Pelizaeus-Merzbacher-like disease f. Rett syndrome
c. Leukodystrophies with trichothiodystrophy g. Toxins
d. 18q-Syndrome and other chromosome 18 mutations h. Progressive cerebral poliodystrophy (Alpers disease)
e. Sialuria IV.D. Metabolic Disorders that Affect both Gray and White
f. Hypomyelination with congenital cataracts Matter
g. Fucosidosis 1. Canavan disease (aspartoacylase deficiency, spongiform
h. Hypomyelination with atrophy of the basal ganglia and leukodystrophy)
cerebellum (HABC) 2. Fibrinoid leukodystrophy (Alexander disease)

xi

Barkovich_FM.indd xi 5/25/2011 11:31:49 AM


xii List of Disorders

3. Mucopolysaccharidoses 17. Cerebrotendinous xanthomatosis


a. Multiple sulfatase deficiency 18. Wolfram syndrome
4. Peroxisomal disorders 19. Marinesco-Sjögren syndrome
a. Peroxisomal biogenesis disorders 20. X-linked nonprogressive congenital cerebellar hypoplasia
b. Acyl-CoA-oxidase deficiency 21. Höyeraal-Hreidarsson syndrome
c. Rhizomelic chondrodysplasia punctata 22. Revesz syndrome
d. Nonrhizomelic chondrodysplasia punctata
5. Wilson disease CHAPTER 4: BRAIN AND SPINE INJURIES IN INFANCY AND
6. Mitochondrial disorders (respiratory chain disorders) CHILDHOOD
a. Mitochondrial encephalomyopathy with lactic acidosis II. Basic Patterns of Brain Destruction
and strokelike episodes (MELAS) 1. Porencephaly
b. Kearns-Sayre syndrome/progressive external ophthal- 2. Hydranencephaly
moplegia 3. Encephalomalacia
c. Mitochondrial neurogastrointestinal encephalomyopathy
d. Disorders causing Leigh syndrome III. Hypoxic-ischemic Brain Injury
e. Alpers disease 1. Localized infarctions
f. Trichopoliodystrophy (Menkes disease) a. Manifestations and causes of infarctions in children
g. Glutaric aciduria type I (glutaryl-CoA-dehydrogenase b. Choice of radiologic study in pediatric stroke
deficiency) c. Arterial infarctions
h. Glutaric aciduria type II (multiple acyl-CoA-dehydrogenase Presumed perinatal ischemic stroke
deficiency) Transient cerebral arteriopathy
i. Neonatal lactic acidosis, complex I/IV deficiency, and fetal d. Infarction secondary to venous occlusion
cerebral disruption 2. Diffuse ischemic or inflammatory brain injury
j. Friedreich ataxia a. Patterns of diffuse hypoxic-ischemic brain injury
k. Ethylmalonic encephalopathy b. Injury in the premature infant
l. Nonspecific mitochondrial disorders Periventricular and intraventricular hemorrhage
7. Pyruvate dehydrogenase deficiency Cerebellar hemorrhage
8. Disorders of the urea cycle and ammonia Venous infarctions
9. Methylmalonic and propionic acidemias White matter injury
10. GM1 and GM2 (Tay-Sachs and Sandhoff diseases) gangliosi- Cerebellar injury
doses Imaging findings in premature neonates
11. Fucosidosis Profound hypotension in premature neonates
12. l-2-Hydroxyglutaric aciduria c. Injury in the term infant
13. Acute necrotizing encephalitis Parasagittal/watershed injury
14. Hypomyelination with atrophy of the basal ganglia and Profound hypotension
cerebellum Parenchymal and intraventricular hemorrhage in the
15. 3-Methylglutaconic aciduria and Barth syndrome term neonate
16. Molybdenum cofactor deficiency d. Injury in older infants and children
17. Isolated sulfite oxidase deficiency e. Ischemia secondary to venous occlusion
18. Toxin ingestions IV. CNS Injury in Multiple Pregnancies
IV.E. Metabolic Disorders Primarily Involving the Cerebellum V. Neonatal Hypoglycemia
1. Friedreich ataxia
VI. Bilirubin Encephalopathy (Kernicterus)
2. Ataxia-Telangiectasia
3. Late onset GM2 gangliosidosis VII. Brain Injury Associated with Congenital Heart Disease
4. Ataxia with oculomotor apraxia, types 1 and 2 VIII. Hypernatremic Dehydration
5. Autosomal recessive spastic ataxia (of Charlevoix-
IX. CNS Trauma in Infancy and Childhood
Saguenay)
1. Birth trauma
6. Mitochondrial disorders causing cerebellar atrophy
a. Spinal cord injury
a. Coenzyme Q10 deficiency
b. Nerve root and brachial plexus injuries
b. SANDO syndrome
c. Head trauma
c. Infantile onset spinocerebellar ataxia
2. Postnatal trauma
7. Infantile olivopontocerebellar atrophy
a. Spinal trauma
8. Pontocerebellar hypoplasia
Injuries to young children
9. Congenital disorders of glycosylation
Adolescent injuries
10. Mevalonic kinase deficiency (Mevalonic aciduria)
Torticollis/rotational deformities of C-1 and C-2
11. Progressive encephalopathy with edema, hypsarrhythmia,
Back pain in children
and optic atrophy (PEHO)
b. Head trauma
12. Dentatorubral and pallidoluysian atrophy
Extraparenchymal hematomas
13. Neuronal ceroid lipofuscinoses
Subarachnoid hemorrhage
14. Langerhans cell histiocytosis
Injury to the brain parenchyma
15. Hypomyelination with atrophy of the basal ganglia and
Sequelae of trauma
cerebellum (HABC)
16. Spinocerebellar ataxias X. Nonaccidental Trauma (Child Abuse)

Barkovich_FM.indd xii 5/25/2011 11:31:49 AM


List of Disorders xiii

CHAPTER 5: CONGENITAL MALFORMATIONS OF THE BRAIN f. Cerebellar duplication


AND SKULL g. Pontine tegmental cap dysplasia
II. Anomalies of Dorsal Prosencephalon Development 4. Combined hypoplasia and atrophy in putative prenatal onset
1. Anomalies of the cerebral commissures—corpus callosum, degenerative disorders
anterior commissure, hippocampal commissure, septum a. Pontocerebellar hypoplasias
pellucidum b. Congenital disorders of glycosylation and other metabolic
2. Malformations of cortical development disorders
a. Malformations secondary to abnormal cell proliferation c. Cerebellar hemisphere hypoplasia
and apoptosis V. Anomalies of the Craniocervical Junction
Microcephaly 1. Chiari I
Focal cortical dysplasia type II 2. Chiari II
Hemimegalencephaly 3. Chiari III
b. Malformations secondary to abnormal cell migration VI. Anomalies of the Mesenchyme (Meninges and Skull)
Lissencephalies 1. Cephaloceles and other calvarial and skull base defects
Malformations due to defects in the pial basement 2. Intracranial lipomas
membrane (Walker-Warburg, muscle-eye-brain, Fukuy- 3. Arachnoid cysts
ama congenital muscular dystrophy, bilateral frontopa- 4. Craniosynostosis syndromes
rietal polymicrogyria phenotypes) a. Nonsyndromic synostosis
Heterotopia b. Synostosis syndromes
c. Malformations secondary to abnormal late migration and Apert syndrome
cortical organization Saethre-Chotzen syndrome
Polymicrogyria Pfeiffer syndrome
Schizencephaly Crouzon syndrome
Focal cortical dysplasia types I, III
VII. Specific Chromosomal Anomalies
III. Anomalies of Ventral Prosencephalon Development 1. Down syndrome
1. Holoprosencephalies 2. Trisomy 18
a. Alobar holoprosencephaly 3. Trisomy 13
b. Semilobar holoprosencephaly 4. Fragile X syndrome
c. Lobar holoprosencephaly
2. Arrhinia/arrhinencephaly CHAPTER 6: THE PHAKOMATOSES
3. Septooptic dysplasia
4. Isolated absence of septum pellucidum I. Neurofibromatosis Type I
5. Anomalies of the hypothalamic-pituitary axis II. Neurofibromatosis Type II
a. Pituitary absence, hypoplasia, and duplication
III. Other Forms of Neurofibromatosis
b. Pituitary dwarfism
c. Kallmann syndrome (hypogonadotropic hypogonadism) IV. Tuberous Sclerosis
d. Hypothalamic dysgenesis V. Sturge-Weber Disease
6. Anomalies of the eyes
VI. Von Hippel-Lindau Disease
a. Ocular malformations
b. Anophthalmia VII. Ataxia-Telangiectasia
c. Microphthalmic malformations VIII. Neurocutaneous Melanosis
d. Macrophthalmia
e. Other congenital ocular anomalies IX. Incontinentia Pigmenti

IV. Anomalies of Midbrain-Hindbrain Development X. Hypomelanosis of Ito


1. Defects of AP and DV patterning XI. Basal Cell Nevus Syndrome
2. Malformations associated with later generalized develop- XII. Cutaneous Hemangioma-Vascular Complex Syndrome
mental disorders that significantly affect the brainstem and
cerebellum XIII. Chediak-Higashi Syndrome
a. Cerebellar aplasia/hypoplasia XIV. Progressive Facial Hemiatrophy (Parry-Romberg
b. Rhombencephalosynapsis Syndrome)
c. Dandy-Walker continuum
XV. Epidermal Nevus Syndrome
d. Cerebral malformations (including microcephaly) with
cerebellar anomalies XVI. Encephalocraniocutaneous Lipomatosis
e. Joubert syndrome and related disorders (molar tooth XVII. Other Overgrowth Syndromes
malformations)
3. Localized brain malformations that significantly affect the BS CHAPTER 7: INTRACRANIAL, ORBITAL, AND NECK MASSES
and CBL: isolated cerebellar and brain stem malformations OF CHILDHOOD
a. Horizontal gaze palsy with progressive scoliosis
I. Introduction
b. Congenital fibrosis of extraocular muscles
1. Introduction and severity of brain tumors in children
c. Midbrain clefts
2. Changing concepts of the biology of brain tumors
d. Cerebellar nodular heterotopia with overlying dysgenesis
3. Clinical features of brain tumors
e. Cerebellar foliation disorders

Barkovich_FM.indd xiii 5/25/2011 11:31:49 AM


xiv List of Disorders

II. Techniques of Imaging Pediatric Brain Tumors c. Infantile myofibromatosis


III. Imaging Characteristics Used to Identify Brain d. Leukemia and lymphoma
Tumors e. Langerhans cell histiocytosis
f. Extramedullary hematopoiesis
IV. Posterior Fossa Tumors g. Calvarial tumors
1. Intraparenchymal tumors
a. Medulloblastoma VI. Brain Tumors in the First Year of Life
b. Atypical teratoid-rhabdoid tumor VII. Radiation Induced Tumors of the Central Nervous
c. Cerebellar astrocytomas System
d. Ependymomas VIII. Tumors of the Head and Neck
e. Brain stem tumors 1. Ocular tumors
f. Hemangioblastoma a. Retinoblastoma
2. Extraparenchymal tumors b. Medulloepithelioma
a. Schwannomas 2. Extraocular orbital masses
b. Dysembryoplastic tumors (epidermoids, dermoids, and a. Vascular masses (hemangiomas, venolymphatic
enteric cysts) malformations, varices)
c. Teratomas b. Orbital cysts
d. Miscellaneous tumors of posterior fossa/skull base c. Leukemia
(meningiomas, langerhans cell histiocytosis, chordoma, d. Metastatic neuroblastoma
chondrosarcoma, neuroblastoma, aneurysmal bone cyst, e. Langerhans cell histiocytosis
Ewing sarcoma) f. Plexiform neurofibroma
V. Supratentorial Tumors g. Dermoids and epidermoids
1. Tumors of the cerebral hemispheres 3. Masses of the face and neck
a. Hemispheric astrocytomas a. Thyroglossal duct cyst
b. Neuronal and mixed neuronal-glial tumors b. Branchial cleft cyst
(gangliogliomas, desmoplastic infantile ganglioglioma/ c. Thymic cyst
desmoplastic astrocytoma of infancy, dysembryoplastic d. Venolymphatic malformations
neuroepithelial tumor (DNET) e. Hemangiomas
c. Supratentorial ependymoma f. Neurofibromas/schwannomas
d. Tumors with neuroblastic or glioblastic elements g. Teratoma
(primitive neuroectodermal tumor (PNET), h. Cervical lymphadenitis
medulloepithelioma) i. Fibromatosis colli
e. Atypical teratoid/rhabdoid tumor j. Rhabdomyosarcoma
f. Astroblastoma k. Nasopharyngeal carcinoma
g. Oligodendroglioma l. Juvenile angiofibroma
h. Neuroglial hamartomas m. Melanotic neuroectodermal tumors of infancy
i. Plasma cell granulomas (inflammatory n. Fibrous dysplasia of the skull base
pseudotumors)
j. Lymphoproliferative disorders CHAPTER 8: HYDROCEPHALUS
k. Germ cell tumors I. Embryology and Physiology of CSF Kinetics and
l. Meningioangiomatosis Dynamics
2. Sellar and suprasellar tumors 1. Choroid plexuses and CSF formation
a. Chiasmatic/hypothalamic astrocytomas 2. CSF circulation
b. Craniopharyngioma 3. CSF absorption
c. Hypothalamic hamartomas
d. Langerhans cell histiocytosis II. Mechanisms of Hydrocephalus
e. Pediatric pituitary tumors III. Classification of Hydrocephalus
f. Rathke cleft cysts 1. Obstructive hydrocephalus
g. Lymphocytic hypophysitis 2. Communicating hydrocephalus
h. Suprasellar germ cell tumors
IV. Clinical Aspects if Hydrocephalus
3. Pineal region masses
a. Germ cell tumors V. Radiologic Diagnosis of Hydrocephalus
b. Pineal parenchymal tumors (pineocytoma, 1. The tools
pineoblastoma) 2. Fetal diagnosis of hydrocephalus
c. Pineal region gliomas 3. Postnatal diagnosis of hydrocephalus and distortions of the
d. Pineal cysts brain from hydrocephalus
4. Extraparenchymal tumors VI. Specific Categories of Hydrocephalus
a. Choroid plexus tumors 1. Hydrocephalus resulting from excessive formation of CSF
b. Tumors of the meninges (meningiomas, plasma cell (choroid plexus papillomas)
granulomas, meningeal fibromas and myofibromas, and 2. Hydrocephalus secondary to intraventricular obstruction of
meningeal sarcomas) CSF flow

Barkovich_FM.indd xiv 5/25/2011 11:31:49 AM


List of Disorders xv

3. Hydrocephalus secondary to extraventricular obstruction CHAPTER 11: INFECTIONS OF THE DEVELOPING AND
of CSF MATURE NERVOUS SYSTEM
4. Communicating hydrocephalus I. Congenital Infections
VII. Benign Enlargement of Subarachnoid Spaces in 1. Cytomegalovirus
Infants 2. Toxoplasmosis
VIII. Treatment of Hydrocephalus and the Resulting 3. Congenital/neonatal herpes simplex encephalitis
Complications 4. Rubella
1. Radiologic assessment of third ventriculostomies 5. Congenital syphilis
2. Shunt malfunctions 6. Lymphocytic choriomeningitis virus
3. Shunt infections 7. Neonatal parechovirus
4. Subdural hematoma formation 8. Congenital varicella
5. Slit ventricle syndrome 9. Human immunodeficiency virus (perinatal transmission)
6. Imaging of intracranial hypotension 10. Disorders mimicking congenital infections
II. Meningitis and Complications
CHAPTER 9: CONGENITAL ANOMALIES OF THE SPINE 1. Neonatal meningitis
I. Normal and Abnormal Embryogenesis of the Spine: an 2. Meningitis in infants, children, and adolescents
Overview 3. Pathophysiology of meningitis
4. Imaging manifestations
II. Clinical Manifestations of Spinal Anomalies
III. Empyema Secondary to Sinusitis and Otomastoiditis
III. Terminology
IV. Bacterial, Spirochetal, and Rickettsial Infections
IV. Imaging Techniques 1. Bacterial cerebritis
V. Abnormalities of Neurulation 2. Brain abscess
1. Disorders resulting from nondisjunction: myelocele, 3. Cat-scratch disease
myelomeningocele, dorsal dermal sinuses, cervical 4. Lyme disease
myelocystocele 5. Rocky Mountain spotted fever
2. Disorders resulting from premature disjunction—spinal V. Viral Infections
lipomas 1. General concepts
VI. Anomalies of the Caudal Cell Mass 2. Herpesvirus family—herpes simplex, varicella zoster,
1. Normal conus medullaris and filum terminale Epstein-Barr, human herpesvirus-6
2. The terminal ventricle 3. Nonpolio enteroviruses
3. Fibrolipomas of the filum terminale/tight filum terminale 4. Arthropod-borne viruses—eastern equine encephalitis,
4. Syndrome of caudal regression western equine encephalitis, Venezuelan equine encepha-
5. Terminal myelocystocele litis, Japanese encephalitis, St. Louis encephalitis, West
6. Anterior sacral meningocele Nile, dengue virus, LaCrosse encephalitis virus, California
7. Sacrococcygeal teratoma encephalitis viruses, Colorado tick fever virus
5. Influenza-associated encephalitis/encephalopathy
VII. Anomalies of Development of the Notochord
6. Acute cerebellitis
1. The split notochord syndrome
7. Rabies
2. Split cord malformation (diastematomyelia and
8. Chronic viral infections—AIDS encephalopathy, progressive
diplomyelia)
multifocal leukoencephalopathy, Rasmussen encephalitis,
VIII. Malformations of Unknown Origin postrubella panencephalitis, subacute sclerosing encephalitis,
1. Segmental spinal dysgenesis variant Creutzfeldt-Jakob disease
2. Dorsal meningocele
VI. Fungal Infections
3. Lateral meningocele
1. Neonatal candidiasis
IX. Congenital Tumors of the Spine 2. Aspergillosis
X. Syringohydromyelia 3. Coccidioidosis
4. Cryptococcus
CHAPTER 10: NEOPLASMS OF THE SPINE VII. Parasitic Infections
II. General Imaging Characteristics of Spinal Tumors 1. Neurocysticercosis
2. Visceral larva migrans
III. Intramedullary Tumors: Clinical Presentation, Pathology,
3. Cerebral malaria
Imaging Characteristics
VIII. Sarcoidosis
IV. Extramedullary Tumors
1. CSF dissemination of intracranial neoplasms IX. Infections of the Spine
2. Tumors of the spinal column 1. Discitis/osteomyelitis
3. Meningeal tumors 2. Spinal empyemas
4. Tumors of nerve roots and nerve root sheaths X. Emerging Infections of the CNS
5. Extraspinal tumors invading the epidural space 1. Nipah virus
V. Congenital Spinal Tumors 2. Dengue virus
3. Chikungunya virus

Barkovich_FM.indd xv 5/25/2011 11:31:49 AM


xvi List of Disorders

CHAPTER 12: ANOMALIES OF CEREBRAL VASCULATURE: VI. Arteriovenous Fistulas


DIAGNOSTIC AND ENDOVASCULAR CONSIDERATIONS 1. Dural arteriovenous fistulas
II. Technical Considerations in Pediatric Neuroangiography 2. Direct carotid cavernous fistulas
and Intervention 3. Vertebral fistulas

III. Intracerebral Hemorrhage in Children VII. Intracranial Aneurysms


1. Saccular aneurysms
IV. Intracerebral Vascular Malformations 2. Mycotic aneurysms
1. Arteriovenous malformations 3. Traumatic aneurysms
2. Vein of galen malformations
3. Cavernous malformations VIII. Spinal Cord Arteriovenous Malformations
4. Venous malformations 1. Intramedullary arteriovenous malformations
5. Capillary telangiectasias 2. Perimedullary arteriovenous fistulas
3. Epidural fistulas
V. Extradural Vascular Malformations and Neoplasms
Requiring Endovascular Treatment IX. Cerebral Vasculopathy in Childhood
1. Hemangioma 1. Moyamoya syndrome
2. Venous malformation 2. Sickle cell disease
3. Facial AVM 3. Hereditary hemorrhagic telangiectasia
4. Epistaxis and juvenile angiofibroma X. Arterial Dissection in Childhood

Barkovich_FM.indd xvi 5/25/2011 11:31:49 AM


C O N T R I B U TO R S

A. James Barkovich, MD Pratik Mukherjee, MD, PhD


Professor Associate Professor in Residence
Radiology, Neurology, Pediatrics, and Neurosurgery Department of Radiology & Biomedical Imaging
University of California at San Francisco Chief of Functional Neuroimaging
Chief University of California at San Francisco
Pediatric Neuroradiology San Francisco, California
UCSF Medical Center/Benioff Children’s Hospital
San Francisco, California Zoltán Patay MD, PhD
Professor of Radiology
Van V. Halbach, MD Department of Radiology
Clinical Professor of Radiology and Neurological Surgery University of Tennessee Health Science Center
University of California at San Francisco College of Medicine
San Francisco, California Chief
Section of Neuroimaging
Gary L. Hedlund, DO Department of Radiological Sciences
Adjunct Professor St. Jude Children’s Research Hospital
Department of Radiology Memphis, Tennessee
University of Utah
Director Charles A. Raybaud MD, FRCPC
Pediatric Neuroradiology Professor of Radiology
Department of Medical Imaging University of Toronto
Primary Children’s Medical Centre Division Head of Neuroradiology
Salt Lake City, Utah Hospital for Sick Children
Toronto, Canada
Christopher P. Hess, MD, PhD
Assistant Professor in Residence Erin Simon Schwartz, MD
Department of Radiology & Biomedical Imaging Associate Professor
University of California, San Francisco Department of Radiology
Chief University of Pennsylvania School of Medicine
Neuroradiology Clinical Director
Department of Radiology Magnetoencephalography
San Francisco Veterans Administration Medical Center Department of Radiology
San Francisco, California The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Philip M. Meyers, MD
Associate Professor L. Gilbert Vezina, MD
Radiology and Neurological Surgery Professor of Radiology and Pediatrics
Columbia University, College of Physicians & Surgeons George Washington University
Director Director of Neuroradiology
Neuroendovascular Services Children’s National Medical Center
Radiology and Neurological Surgery Washington, D.C.
New York Presbyterian Hospitals—Columbia
New York, New York

xvii

Barkovich_FM.indd xvii 5/25/2011 11:31:49 AM


Barkovich_FM.indd xviii 5/25/2011 11:31:49 AM
CHAPTER

Techniques and Methods


1 in Pediatric Neuroimaging
CHRISTOPHER P. HESS AND A. JAMES BARKOVICH

Sedation Special MR techniques


MR scanning without sedation MR and CT angiography
Sedation techniques CSF flow imaging
Monitoring Magnetic resonance neurography
Special problems in the imaging of premature infants Magnetization transfer
Contrast agents Perfusion imaging
Iodinated contrast agents Diffusion tensor imaging
Paramagnetic contrast agents Proton MR spectroscopy
Ultrasound indications and techniques Automated morphometric analysis and tissue
CT scanning indications and techniques segmentation
Routine MR imaging sequences Imaging of brain function
Brain Summary
Spine
Fetal MRI

odern imaging modalities have greatly advanced both the


SEDATION
M understanding and the diagnosis of pathology of the pediatric
central nervous system. In order to maximize the information The most important factor for obtaining high-quality MR images in
gained from these studies, high-quality images must be obtained. The children is making sure that they remain still while the images are
use of high-frequency transducers and acquisition of images via the acquired. When children do not remain still, motion artifacts will
anterior and posterolateral fontanelles allows outstanding sonographic obscure important diagnostic information. In general, sedation is not
visualization of the brain and intracranial blood vessels in neonates and necessary for CT when modern scanners are being used; indeed, seda-
young infants (1,2). Helical (spiral) computed x-ray tomography (CT) tion was required in only 1.4% of 219 children on a multisection helical
scanners obtain images in fractions of a second, permitting high-quality CT scanner (3). Younger children can usually be adequately restrained,
vascular imaging and facilitating rapid, high-quality three-dimensional and older children will hold still long enough for a good CT examina-
(3D) and two-dimensional (2D) reformations. The effects of physio- tion to be obtained. The technologist should watch the patient during
logic and bulk motion during scanning are minimized and high-quality the CT exam and initiate scanning while the patient is not moving;
images can be rapidly obtained (but one must always remember that rapid reconstruction times allow acquisition of new images at targeted
higher image quality comes with a price of increasing radiation dose). levels that are motion degraded on the initial scans. In those difficult
Although the use of fractional k-space acquisitions, echo-planar imag- cases where sedation is required, the protocols outlined in this section
ing (EPI) and other fast-scan techniques, and PROPELLER help reduce are applicable. In general, however, the use of helical acquisition has
sensitivity to patient motion in magnetic resonance imaging (MRI), made sedation rare for CT in most institutions (3,4).
artifacts remain an issue for certain applications, especially when seda-
tion is inadequate. Moreover, optimal imaging parameters for the
MR Scanning Without Sedation
pediatric patient (especially the neonate and young infant) differ from
those of the adult because standard adult imaging sequences do not Although sedation is necessary for most MR studies of children younger
consider the changing chemical composition of the developing brain. than 8 years, fast MR techniques such as EPI, half-Fourier acquisition
Finally, the increasing use of 3 tesla (3 T) scanners requires alterations single-shot turbo spin echo (HASTE, also called single-shot fast spin
of imaging techniques and parameters to compensate for differences in echo or ssFSE by manufacturers) and rapid acquisition with relaxation
T1 relaxation times and magnetic susceptibility that result from higher enhancement (RARE, also called fast spin echo, FSE, or turbo spin
field strength. The purpose of this chapter is to outline techniques for echo, TSE) (5,6) allow high-quality images of the entire brain to be
safely and effectively obtaining high-quality imaging studies in pediat- obtained within a sufficiently short examination time (<1 minute) to
ric patients. In addition, the techniques that we currently use in the CT avoid significant motion artifacts. Alternatively, the periodically rotated
and MR evaluation of infants and children at the University of Califor- overlapping parallel lines with enhanced reconstruction (PROPELLER
nia, San Francisco, are summarized. These techniques are referred to or PROP) acquisition and reconstruction scheme is relatively tolerant
throughout the remainder of this book. of patient motion (7,8). EPI and HASTE/ssFSE are often adequate for

Barkovich_Chap01.indd 1 5/5/2011 11:43:32 PM


2 Pediatric Neuroimaging

diagnosis of ventriculomegaly/shunt failure or acute infarcts but are 6. Method of continuous patient observation (window, camera)
not, at this time, of adequate quality to allow diagnosis of subtle mal- 7. Resuscitation equipment, including oxygen delivery and suction,
formations, leukodystrophies, or tumor progression. PROPELLER will that is checked and maintained on a scheduled basis
allow identification of malformations and tumors but does not have 8. Uniform system of record-keeping and charting (with continuous
the contrast resolution to detect subtle white matter abnormalities. assessment and recording of vital signs)
If sedation can be avoided, it should be, as complication rates are 9. Location and protocol for recovery and discharge
generally in the range of 0.4% to 1% even in the best of hands (9). The 10. Program for continuous quality improvement that tracks compli-
incidence of complications seems to be significantly higher if the child cations and morbidity
has a history of respiratory illness or if more than one agent is used
for sedation (9). Therefore, it is important to remember that sedation In addition, the 2008 update of the AAP guidelines requires the fol-
is not universally necessary for obtaining standard MR sequences of lowing (14); please read all of these guidelines before embarking on
pediatric patients. Neonates can often be scanned without sedation if pediatric MRI sedation.
they are fed immediately prior to the study, kept warm, and earmuffs/ 1. No administration of sedating medication without the safety net
earplugs are used to reduce the noise; we have found the use of 24% of medical supervision
oral sucrose (Sweet Ease) and nonnutritive sucking (i.e., a sucrose paci- 2. Careful presedation evaluation for underlying medical or surgical
fier) immediately prior to the scan to be useful by promoting a calm conditions that would place the child at increased risk from sedat-
state and reducing stress. Mathur et al. report considerable success in ing medications
scanning unsedated neonates (10). We have had success with this, as 3. Appropriate fasting for elective procedures and a balance between
well, as described in the section below on Special Issues in Scanning of depth of sedation and risk for those who are unable to fast because
Premature Neonates. Scanning neonates requires an armamentarium of the urgent nature of the procedure
of MR compatible life-support and monitoring equipment, trained 4. A focused airway examination for large tonsils or anatomic airway
transport personnel, at least 1 hour of preparation time before the baby abnormalities that might increase the potential for airway obstruc-
is transported to the scanner, and a similar amount of time after the tion
scan (10). 5. A clear understanding of the pharmacokinetic and pharmacody-
With adequate personnel and equipment, it can be achieved with namic effects of the medications used for sedation as well as an
a high success rate. The success rate diminishes with the maturity of appreciation for drug interactions
the baby, however. As they mature, infants become increasingly aware 6. Appropriate training and skills in airway management to allow res-
of their surroundings and sedation becomes increasingly necessary in cue of the patient
order to obtain diagnostic scans; by the age of 2 to 3 months, sedation 7. Age- and size-appropriate equipment for airway management and
is nearly always necessary. Fewer older children need sedation if some venous access
sort of MR-compatible audiovisual system is available. We find that 8. Appropriate medications and reversal agents
most children aged 8 years and older as well as some 6- and 7-year-olds 9. Sufficient numbers of people to carry out the procedure and moni-
can hold still for an MR study if they are adequately enraptured by a tor the patient
movie. Harned and Strain (11) found a 25% reduction in the number 10. Appropriate physiologic monitoring during and after the proce-
of sedations for children aged 3 to 10 years and a 50% reduction in dure
sedations of children more than 10 years old when they began to use an 11. A properly equipped and staffed recovery area
audiovisual system in their MR scanner. They also found a 17% reduc- 12. Recovery to presedation level of consciousness before discharge
tion in the MR room time for patients who were examined without from medical supervision
sedation. Hallowell et al. (12) prepared children (aged 3.7–17 years) 13. Appropriate discharge instructions
in a practice MRI unit (without real magnets); of the 78% who per-
formed adequately during preparation and underwent an MR scan, Although the most common complications from sedation are related
they were able to obtain diagnostic noncontrast images in 96%. How- to respiratory compromise, special attention must be given to monitor-
ever, contrast-enhanced images were not obtained and motion artifact ing body temperature in the neonatal and the young pediatric popula-
was seen in more than 60% of studies. tion. Temperature monitoring equipment that is approved for use in
the MR suite is becoming more readily available. A growing number of
commercial MR-approved neonatal isolation transport units and other
Sedation Techniques warming devices have also become more readily available for use dur-
ing MR scans.
When sedation is necessary, the practitioner should be familiar with the
A number of different drugs can be and have been used safely for
guidelines published by the American Academy of Pediatrics (13,14), the
pediatric sedation (17–27). Specific anesthetic regimens vary region-
American Society of Anesthesiologists (15), and the American College of
ally and institutionally, and a comprehensive discussion of appropriate
Radiology (16). These guidelines require the following provisions:
medications for sedation during imaging is beyond the scope of this
1. Preprocedural medical history, physical examination, and physical text. Some regimens that have been used safely by physicians creden-
status evaluation tialed in pediatric sedation in our practice are listed below for reference.
2. Informed consent according to local, state, and institutional The reader should consult with local anesthesiologists and/or pediatri-
requirements cians before employing any anesthesia protocol, whether listed here or not.
3. Fasting guidelines appropriate for age to prevent pulmonary Moreover, close contact should be maintained with an anesthesiologist or
aspiration experienced pediatrician for consultation in difficult cases.
4. Uniform training and credentialing for sedation providers Sedation can be administered by many routes: oral (PO), rectal
5. Baseline, intraprocedural, and postprocedural monitoring with (PR), intramuscular (IM), or intravenous (IV). PO, PR, and IM routes
devices appropriately sized for children and compatible with the have the important advantage that they do not require special skills
magnetic field in the case of MRI for administration. However, the absorption of many drugs from the

Barkovich_Chap01.indd 2 5/5/2011 11:43:32 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 3

muscle and gastrointestinal tract can be erratic. Thus, sedatives and is used for reversal of respiratory depression from opioids and given
anesthetics administered in large doses by these routes can be difficult intravenously in increments of 0.005 to 0.01 mg at 2 to 3 minute inter-
to reverse in the event a complication arises during sedation. The rec- vals. It is important to keep in mind that the half-life of both reversal
ommendation of the American Academy of Pediatrics is that infants agents is less than most of the drugs that they are used to reverse, and
should be given nothing by mouth for at least 4 hours prior to deep that patients should thus be monitored following the procedure in the
sedation; older children for at least 6 hours (28). This accelerates bioab- event that additional doses of these agents are necessary.
sorption in the case of PO and PR medications but, more importantly, The most popular drug used for IV sedation is pentobarbital
decreases the risk of pulmonary aspiration during the examination. (21,22), which is administered intravenously in the following manner.
Sodium pentobarbital (Nembutal) is a useful short-acting bar- Approximately 2.5 mg/kg is given over 30 to 40 seconds while closely
biturate for enteral or parenteral sedation in pediatric patients (20), observing the patient. If the patient does not fall asleep within 60 sec-
although it should be used with care in patients who have hepatic or onds, a second dose of 1.0 mg/kg is administered. If the patient remains
metabolic disease, as the drug may alter the metabolism of other medi- awake, a third and fourth administration of 1.0 mg/kg can be given
cations. The usual dose of sodium pentobarbital is 6 mg/kg for the first up to a total dose of 6 mg/kg. Repeated doses of 1 to 1.25 mg/kg are
15 kg of body weight, followed by 5 mg for each additional kg up to then administered, as needed, to maintain sedation during the exam.
a total dose of no more than 100 mg (as a single IM injection). The Small motions of the arms and legs are usually an indication that the
drug should be administered 35 to 45 minutes before the anticipated level of sedation is diminishing. At UCSF, our pediatric anesthesiolo-
imaging time. gists have recommended that pentobarbital be used in conjunction
Historically and in a previous edition of this book, the safest drug with morphine sulfate, as the two drugs have complimentary actions.
for sedating children was considered to be chloral hydrate. Others have This regimen consists of an initial dose of 1 to 2 mg/kg pentobarbital,
also reported on the safety and efficacy of chloral hydrate, especially in followed by 0.05 mg/kg morphine, followed by another dose of pen-
infants under the age of 18 months (22) and even up to age 4 years (29). tobarbital, another of morphine, etc., until the patient is adequately
However, because questions have been raised concerning the potential sedated. Additional doses are given, as needed, if the level of sedation
carcinogenicity of this drug (30), its use has been restricted in some appears to be diminishing.
countries. In the United States, the American Academy of Pediatrics IV sedation using propofol (Diprivan), an ultrashort-acting sed-
has judged the evidence insufficient (31), and many physicians con- ative-hypnotic, has been successfully used instead of pentobarbital
tinue to use chloral hydrate without incident. Usually administered in some studies (26). Propofol acts rapidly and, more importantly, is
orally, known side effects include prolonged somnolence (>4 hours), rapidly metabolized so that the sedated children rapidly recover after
unsteadiness, and hyperactivity in some children after waking (32). imaging. The disadvantage of this drug is the significantly higher rate
Such symptoms may require the child to be kept at the imaging center of respiratory events compared to, for example, pentobarbital, requir-
and observed for a prolonged time period. In addition, one study sug- ing significantly more airway manipulations to relieve obstruction
gested that the complication rate, particularly for episodes of oxygen (34). Therefore, in the United States, it has been recommended that
desaturation, is lower with oral pentobarbital than with chloral hydrate only anesthesiologists administer propofol (15). As a result, those wish-
and recommended that oral pentobarbital be used as an equally effec- ing to take advantage of the rapid induction and recovery of pediatric
tive and safer drug than chloral hydrate in infants (33). If used, chloral patients sedated with propofol need to arrange to have pediatric seda-
hydrate is administered orally in a dose of 50 to 75 mg/kg for the first tion performed by anesthesiologists or nurse anesthetists. This is cur-
10 kg of body weight, then 50 mg/kg for each kg of body weight above rently the situation at UCSF. The reader should be aware that the use
10 kg. If the child is still awake after 20 minutes, supplementary doses of propofol with 100% oxygen can cause artifacts in fluid attenuation
can be given up to a total dose of 2000 mg. Chloral hydrate is not cur- inversion recovery (FLAIR) images, with the cerebrospinal fluid (CSF)
rently used for infant sedation at UCSF. appearing abnormally hyperintense in the sulci and cisterns (35,36).
After enteric or intramuscular sedatives have been administered, If CSF hyperintensity is seen in the cisterns on FLAIR sequences in a
the patient and parent should be transferred to a dark, quiet room. The sedated child, the agent used for sedation and the percent of supplemen-
noise and activity of a busy waiting room will otherwise keep the child tal oxygen must be researched before the scan is interpreted as abnormal.
awake. Sleep deprivation is also helpful. If the imaging study is per- Using lower concentrations of supplemental oxygen (50%–60% works
formed in the morning, keep the child awake several hours past nor- well) eliminates this artifact (37,38).
mal bedtime the night preceding the exam and awaken the child earlier
than usual on the morning of the exam. If the imaging is performed in
the afternoon, deprive the child of his or her normal naps. Do not allow
MONITORING
the child to fall asleep during travel to the imaging facility. Finally, per- The American Academy of Pediatrics and the American Society of
mit a parent to enter the scanner with the patient, if desired, in order Anesthesiologists recommend that the following parameters be moni-
to reassure the child that security is nearby. The second body will not tored in all sedated infants and children: heart and respiratory rates,
create artifact, as long as the parent has been screened adequately for blood pressure, and arterial oxygen saturation (13–15). Monitoring a
prosthetics or other metallic objects. patient in the CT suite is relatively simple. A patient who is undergo-
IV sedation offers a number of advantages over other routes of ing MRI is more difficult to monitor because of the safety issues and
administration. Once IV access is obtained, sedation can be rapidly because biomonitoring devices (a) may not work properly in the mag-
achieved. Dosage can be titrated more easily, and supplemental doses netic environment and (b) may cause distortion of the magnetic field,
are easily administered without disturbing the patient. Finally, and per- especially within the narrow confines of the bore of the magnet. Moni-
haps most importantly, the effects of many drugs (e.g., opiates and ben- toring must be performed using equipment composed of diamagnetic
zodiazepines) can be rapidly reversed. Reversal agents should be readily metals (e.g., aluminum) and/or plastics; several MRI-compatible mon-
available for all cases in which sedation is administered. Flumazenil itoring devices are commercially available and are the best option for
(Romazicon), a benzodiazepine receptor agonist, is administered in IV patient safety and optimal imaging. However, in the absence of MRI-
doses of 0.01 mg/kg (up to 0.2 mg) over 15 seconds and repeated up to compatible monitoring equipment, other techniques may be used.
a maximum of four additional times. Naloxone (Narcan), in contrast, A plastic stethoscope with very long tubing may be taped to the

Barkovich_Chap01.indd 3 5/5/2011 11:43:32 PM


4 Pediatric Neuroimaging

patient’s chest so that heart rate can be monitored from outside the If MR imaging of premature infants is uncommonly performed at an
bore of the magnet. Electrocardiogram leads, if necessary, may be run institution and an MR-compatible incubator is not cost-effective, the
underneath the patient and as far away as possible from the body part infant may be wrapped in an air bag that is warmed to body tempera-
being imaged. A CO2-sensitive apnea monitor connected by long, small ture or in prewarmed towels. Alternatively, chemical “blankets” contain-
caliber tubing to the patient in the scanner provides visual display of ing mixtures of chemicals that maintain a temperature of 37°C when
respiration and audio and visual alarms during apnea episodes with- mixed can be used to maintain body heat. A stockinet hat may be used
out affecting image quality. Disposable, pediatric size nasal cannulae to prevent heat loss from the head. Earmuffs reduce noise exposure and
are available. Fortunately, pediatric-sized MR-compatible monitoring further reduce heat loss. Monitoring of the vital signs in these infants is
equipment is now available from many manufacturers. If the practi- critical. The child should be disturbed as little as possible.
tioner is performing MR examinations on a large number of sedated
children, this equipment is well worth the investment. CONTRAST AGENTS
Although some lower field strength magnets and some shielding
configurations will allow life support equipment to be in close proxim- Iodinated Contrast Agents
ity to the patient, most high field MR suites require significant modifi-
CT scans in children should initially be performed without IV contrast.
cations to achieve intense, close-in electronic monitoring. As a general
If a noncontrast scan reveals an abnormality and an MR cannot be
rule, CT should be used for the identification of life-threatening con-
obtained in a timely fashion, IV contrast should be given. If the noncon-
ditions in highly unstable patients. More and more commonly, how-
trast CT scan is normal, very little information is gained by administering
ever, unstable patients are also being imaged with MR. MR-compatible
contrast unless a vascular lesion is suspected (49). Nonionic, iso-osmolar
pediatric ventilators are commercially available; in their absence,
(Visipaque) or low-osmolar (Omnipaque) contrast media should be
respirator-dependent patients must be manually ventilated. When
used because they are safer and less uncomfortable for the patient. The
MRI is necessary in patients with significant respiratory or hemody-
exact type of iodinated contrast is not important, as long as the concen-
namic instability, the physician or nurse may sometimes have to crawl
tration of iodine is approximately 300 mg/mL. The recommended dose
into the bore of the magnet and observe the child from this extremely
is 3 mL/kg of body weight up to a total dose of 120 mL. The child should
uncomfortable position throughout the examination.
be scanned as soon as possible after the contrast has been administered.
Adverse reactions to iodinated contrasts are rare in the pediatric popula-
SPECIAL PROBLEMS IN THE IMAGING OF tion. They are most rare in the youngest patients (50). Acute reactions are
PREMATURE INFANTS most common in children weighing 24 to 40 kg. Asthma and previous
reactions to contrast medium are risk factors for acute reactions (50).
Premature infants present the special problems of small size and
inability to maintain constant body temperature. In general, prema-
ture infants should be imaged initially with ultrasound in the neona-
Paramagnetic Contrast Agents
tal intensive care unit. Cranial ultrasound is the initial examination of MRI can be performed in most cases without paramagnetic MR con-
choice in these patients because it is inexpensive and portable (exams trast agents, although the administration of gadolinium chelates may
can be performed without moving infant from the neonatal intensive improve the identification and evaluation of primary and metastatic
care unit). Moreover, transfontanelle ultrasonography with high fre- brain tumors (especially extraparenchymal tumors), infections (abscess,
quency transducers is excellent for the detection of edema, blood or empyema, cerebritis, and meningitis), neoplasms of the spinal cord and
infarction in deep white matter of the brain, the location of most cen- spinal canal, and some neurocutaneous disorders (neurocutaneous
tral nervous system pathology in the premature infant (see Chapter 4), melanosis, neurofibromatosis type II, Sturge-Weber syndrome) (51,52)
and for development or progression of hydrocephalus. However, MR and permits the assessment of cerebral perfusion using dynamic sus-
has an increasing role in the evaluation of the premature infant, as it ceptibility contrast MRI (DSC-MRI) methods (53,54). Administration
can detect abnormalities that are not visible by sonography (39,40) and of contrast to infants or children with developmental delay is unlikely
these abnormalities are of prognostic significance (41–44). to be of any diagnostic benefit unless a space-occupying lesion is iden-
When an MR examination is necessary, special precautions must be tified on a noncontrast study or a cutaneous lesion suggesting one of
observed to ensure the safety of the neonate. It is best to enlist neona- the neurocutaneous disorders mentioned above is present. Similarly,
tologists or neonatal nurses to assist in the transport of the patient and no advantage is gained by administering paramagnetic contrast agents
monitoring during the imaging study, as they are most experienced in to children with malformations of the brain or spine, with the excep-
maintaining homeostasis in the neonate. This is especially the case for tion of dermal sinus tracts (see Chapter 9).
preterm infants, who have traditionally been managed with mechanical Once widely administered in the setting of impaired renal func-
ventilation but more recently are being extubated at an earlier time point tion, gadolinium chelates are now known to be associated with neph-
and ventilated using nasal continuous positive airway pressure (45). At rogenic systemic fibrosis (NSF) in patients with renal insufficiency
UCSF, where MR imaging of prematurely born neonates is commonly (55,56). This rare but serious systemic disorder causes fibrosis of the
performed, we use a prototype MR-compatible incubator with forced air skin and other tissues, thereby leading to considerable morbidity or
heating and an infrared video system (46). Small windows in the walls even death. To the date this chapter was revised; there have been nine
of the incubator allow monitoring equipment to be utilized while the reported cases of NSF in children after gadolinium injection (57,58).
patient is in the incubator. The child is not disturbed during the entire All of these occurred in patients with low glomerular filtration rate
trip to and from the scanner (including the scan) except on the very rare (GFR < 30), although there are a few case reports of NSF in adults
occasions when problems occur. A similar system is now commercially with slightly higher GFR between 40 and 60. A proinflammatory state
available and reportedly works well, providing excellent images while (e.g., systemic infection, limb or major tissue injury, recent surgery or
allowing safety and close monitoring for the infant (47,48). We have thrombosis) may increase the risk of developing this complication.
found that when the baby is minimally disturbed in the incubator, we can With the recognition of the relationship between gadolinium and NSF,
perform the MR scan without sedation in as many as 70% of premature there is no longer a justification for administering contrast to every
neonates; this should be attempted before sedation is administered. sedated patient “just in case,” that is, in order to avoid resedating the

Barkovich_Chap01.indd 4 5/5/2011 11:43:32 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 5

child for a repeated contrast-enhanced scan. When gadolinium is subtle changes in echogenicity. Finally, major arteries and veins should
necessary, screening for NSF risk factors is essential, and the relative be assessed by Doppler techniques, looking for peak systolic velocities,
risks and benefits in patients with renal failure should be carefully con- end diastolic velocities, and resistive indices.
sidered in consultation with referring physicians.
At high and repeated doses, IV gadolinium is teratogenic in animal
studies (59). Although no similar effects have been observed in human CT SCANNING INDICATIONS AND
studies of teratogenicity, gadolinium is known to cross the placenta
and may be excreted in the amniotic fluid, where the dwell time of
TECHNIQUES
the agent may exceed several days. There are no indications for which Radiation dose is always a consideration in pediatric radiology, as
maternal paramagnetic contrast agent administration is necessary for younger patients have a greater chance of developing radiation-
fetal MRI. associated diseases (62–64) and, possibly, developmental impairment
From a diagnostic efficacy standpoint, no significant difference has (65). The increasing utilization of CT over the past decade among the
been demonstrated among the different paramagnetic contrasts that entire US population, including pediatric patients, and the variability
are commercially available. All are given intravenously; the standard in scan parameters for CT (66) have resulted in an increased focus
dose is 0.1 mmol/kg (corresponding to a volume of 0.2 mL/kg for on the need to consider alternative modalities first (67). When CT is
nearly all commercial preparations). After infusion of contrast, only necessary, one must also consider whether dose reduction techniques
T1-weighted spin-echo, three-dimensional Fourier transform (3DFT) (68,69) should be used, recognizing that the diagnostic quality of the
gradient echo, (i.e., spoiled gradient recalled [SPGR], or magnetiza- imaging study may suffer when image contrast-to-noise is reduced by
tion prepared rapid acquisition gradient echo [MPRAGE]), FLAIR, or dose reduction (70,71). Institutional approaches based on systematic
dynamic contrast-enhanced MRI perfusion sequences should be consideration of clinical indications and prior history of exposure
obtained; fat suppression may be useful if meningeal disease is sus- to ionizing radiation may help increase the consistency with which
pected (60) or if suspected pathology lies in the orbits or neck, where fat specific parameters are used for pediatric CT scanning and to reduce
may obscure the enhancing lesion. T2-weighted images have no value overall radiation exposure (72).
in this situation. It is likely that higher doses of contrast (0.3 mmol/ The indications for the study and scan parameters should be
kg) or the use of magnetization transfer pulses to suppress white mat- weighed together. For example, when a child has sustained acute head
ter signal allows a higher sensitivity for detection of enhancing lesions, trauma, the concern is mainly for fractures, pneumocephalus, or a
particularly CSF-borne metastases from brain tumors (4); however, space-occupying hematoma that may alter emergent management.
higher doses likely increase the incidence of NSF and, therefore, should The goal in this scenario is rapid diagnosis and not necessarily high-
be used only when the benefits of added contrast, such as identification resolution evaluation of the brain parenchyma; therefore, low-dose CT
of a condition that might alter therapy, outweigh the potential risks. is adequate for most cases. If indicated, an MR can be acquired when
the patient is more stable to better evaluate the extent of any brain
injury. In contradistinction, in the acutely encephalopathic child with
ULTRASOUND INDICATIONS AND new neurological signs or symptoms, our approach is to get an MR
on a timely basis. If MR is contraindicated or unavailable, we obtain
TECHNIQUES a standard-dose CT scan of good quality in order to avoid repeating
Ultrasound is nearly always the first study of choice in neonates, as the scan or necessitating another type of scan. When arterial or venous
it is noninvasive, inexpensive, and portable (can be performed at the thrombosis is suspected, we will usually obtain a magnetic resonance
bedside) and produces no ionizing radiation. Sedation is not neces- angiography (MRA) or magnetic resonance venograhy (MRV). If
sary. Ultrasound technique has markedly improved in recent years questions arise due to saturation effects resulting from complex flow,
with the manufacture of high-frequency transducers, the introduction contrast-enhanced MRA or MRV will almost always answer them. Very
of high-bandwidth tissue harmonic imaging techniques, and the use of rarely, CT angiography (CTA) may be necessary, as it is less affected by
multiple acoustic windows, to the point where much information can artifacts and can be performed rapidly and without sedation. However,
be gathered about most regions of the brain from a good ultrasound the scan should be limited to specific regions of concern, and exposure
study. Indeed, measurements of brain structures with ultrasound and of the eyes, thyroid, and breasts should be minimized.
MRI are nearly identical (61), with the small differences (mainly cor- When patients have conditions that will require a number of imag-
tical thickness and interhemispheric fissure size) most likely due to ing studies over many years, the imaging technique of choice requires
inability to differentiate the cortex from the overlying leptomeninges considerable thought. For example, patients with hydrocephalus will
on ultrasound. The peripheral aspects of the brain may also be difficult often have multiple, possibly dozens, of scans during childhood and
to visualize with ultrasound, particularly if the fontanelles are small, the accumulated radiation dose can become significant (73). If the pur-
reducing the size of the acoustic window. pose of the scan is only to check ventricular size after placement or
Meticulous technique on the part of well-trained sonographers revision of a ventricular catheter in a child with known hydrocephalus,
must be used to optimize the study. Imaging should always be per- a high-dose technique is not necessary; instead, one should consider a
formed using multiple transducers functioning at variable frequencies. CT technique using lower kilovoltage or lower amperage. By reducing
Vector, curved, and linear array transducers should all be used. Resolu- CT dose from 220 to 80 mA, one study showed a reduction in radia-
tion and depth penetration can be optimized by adjusting the frequen- tion dose of 63% while maintaining diagnostically acceptable images
cies (between 8 and 17 MHz) and the focal zone of the ultrasound (74); as noted above, however, the reduction of signal-to-noise that
beam. When the brain is thus analyzed via the anterior and posterior accompanies the dose reduction severely limits assessment of brain
fontanelles and the temporal, mastoid, and occipital synchondroses, all parenchyma. Alternatively, a fast MR sequence, such as HASTE/ssFSE,
regions of the brain (central and peripheral) can be seen well. Abnor- or a PROPELLER sequence, may be used, thereby eliminating ioniz-
malities will be shown best if images are acquired in sagittal, paras- ing radiation altogether. In the age of magnetically adjustable pressure
agittal, coronal, and axial planes. The radiologist should review both valves, however, the use of MR requires a second visit to the neurosur-
static and real-time images; the latter allows a better appreciation of geon after the scan to readjust the valve; thus, the decision is complex.

Barkovich_Chap01.indd 5 5/5/2011 11:43:32 PM


6 Pediatric Neuroimaging

If, after careful consideration, CT is chosen as the imaging of the cerebral hemispheres, especially around the sylvian fissures.
technique of choice, the study should be performed with the lowest When acquired as the first sequences in the examination, this sequence
possible dose that allows a diagnostic quality scan. Some recent reviews can also be used as a localizer for additional axial or coronal sequences.
of methods for calculating and reducing dose for patients of all ages Using a standard quadrature head coil at 1.5 T, suitable parameters
elegantly describe how to accomplish this (68,69). These methods for T1-weighted imaging are a repetition time (TR) of 500 to 600
should be familiar to all physicians involved in imaging. The techniques milliseconds, echo time (TE) as short as possible (20 milliseconds or
for scanning older children with CT are identical to those for scanning less), 3 to 4 mm slice thickness (1 mm gap), acquisition matrix 192 × 256,
adults. Axial images are obtained using ≤5 mm slice thickness; an eye and one signal average. Using these parameters, it is possible to cover
shield may be used and the plane of section should be chosen to mini- most of the brain in the sagittal plane. If the option of 192 acquisitions
mize exposure to the eyes. As CT scanning has become more rapid, in the phase encoding direction is not available, 128 phase encodings
little time penalty is paid for relatively thin slice profiles and significant (at the expense of decreased spatial resolution) or 256 phase encodings
additional information may be acquired, especially in the small heads (at the expense of increased imaging time and diminished signal-to-
of infants and young children. However, a price is paid in that the sig- noise ratio) can be used. Using a short TR (<500 milliseconds) will save
nal-to-noise diminishes with decreasing slice thickness, and this must imaging time while still allowing assessment of the midline structures,
be compensated with increased kilovoltage or milliamperes per second, but a price is paid in that signal-to-noise decreases and multislicing may
which increases dose. To mitigate that issue, the thinner sections can not allow the lateral portions of the brain to be covered unless fractional
be reformatted as thicker (4–5 mm) sections, giving good signal-to- k-space acquisitions or a rectangular field of view is also employed. If a
noise and the ability to look at the thinner sections (e.g., when look- phased-array head coil and parallel imaging are available, particularly
ing for fractures) if desired (68). We typically acquire sequential scans at 3 T, a 3D radiofrequency or gradient-spoiled gradient-echo volu-
rather than using spiral acquisition for head studies, as signal-to-noise metric sequence (SPGR, MPRAGE, 3D-FLASH, CE-FFE-T1, T1-FAST)
is improved, artifacts are avoided, and there is little time penalty (75). should be obtained in the sagittal plane (employing the largest, cran-
If coronal images are needed, strong consideration should be given to iocaudal dimension as the readout axis for quickest acquisition) and
reformatting axial images; this saves the added radiation of an extra high-quality reformations in the coronal and axial (and, if necessary,
sequence. In such cases, spiral acquisition may be desirable. A low pitch true sagittal) planes can be easily obtained. This has become our most
(<1) should be used if higher resolution is needed; reducing milliam- useful sequence for the diagnosis of structural abnormalities. For this
pere settings can ameliorate any increase in radiation dose. If spiral sequence, we use TR/TE = 35 milliseconds/minimum, flip angle 35°,
CT is not available, the higher resolution may be obtained via the use 22 × 22 cm FOV, 1 mm partition size, and 256 × 192 acquisition matrix
of direct coronal images; the unsedated patient may be examined in with no signal averaging. If imaging at 3 T is not available or thin volu-
the prone or supine positions, but coronal scans of sedated patients metric images are not desired, FLAIR T1 acquisition using TR/TE =
are best performed in the supine position to avoid compromising the 600/20 milliseconds and inversion time (TI) of 750 milliseconds also
airway. Ideally, the plane of scanning should be perpendicular to the gives excellent T1-weighted images.
planum sphenoidale. After the sagittal T1 images have been obtained, axial T2-weighted
Another special situation in pediatric CT imaging in which tech- images should be acquired in all patients. For patients older than
niques can be modified is in the scanning of patients with craniofacial 24 months, only these T2-weighted images are necessary for the most
anomalies or craniosynostosis. These patients should be scanned using basic imaging protocol. If a volumetric sequence has not been acquired
≤2.5 mm slice thickness. Thicker slices allow too much averaging and and reformatted axially in infants with developmental delay or sei-
obscure detail. Reconstruction algorithms that give high-detail bone zures, we also obtain an axial T1-weighted sequence (inversion recov-
resolution should be used to evaluate the cranium; they give better ery, spin echo). For patients less than 18 months of age, both T1- and
images with a lower radiation dose. If no MR scan will be obtained, T2-weighted images are necessary for accurate interpretation. Brain
discuss with the referring surgeon the use of a soft tissue algorithm to maturation is evaluated best by T1-weighted images from birth to
assess the underlying brain for abnormality; the unnecessary data can 6 months of age. However, from 6 to 8 months of age until approxi-
then be discarded during the process of reconstructing 3D reforma- mately 24 months of age (at which time the brain is essentially mature
tions. Software that allows 3D surface- and volume-rendered recon- by MR standards), T2-weighted images are more useful for assessing
structions of the bones of the face and skull is available from most myelination and brain maturity. During the process of white matter
manufacturers; this has significant value in assessing suture patency maturation, the cerebral cortex and subcortical white matter of the
and in planning reconstructive surgery (see Chapter 5). brain become isointense for a variable period of time on MR images;
Because the newborn brain has a very high water content, proper this isointensity obscures structural detail (see Chapter 2). Therefore,
windowing of the CT scan is essential for optimal analysis of brain during the first 8 months of life (while the white matter is matur-
abnormalities. In general, CT images of the newborn brain should be ing on T1-weighted images), T2-weighted images are necessary to
reviewed and filmed with a window of 60 and level of 20. Use of normal see the details of the gyral and sulcal patterns. Similarly, as the white
adult brain windows will result in pathology being missed. matter matures on T2-weighted images between 8 and 24 months
of age, T1-weighted images are essential for evaluation of structural
abnormalities.
ROUTINE MR IMAGING SEQUENCES The optimal imaging parameters may vary from one scanner to
another and at different field strengths. 3DFT spoiled gradient-echo
Brain
techniques that use radiofrequency or gradient pulses to “spoil” residual
A sagittal, T1-weighted sequence should be performed on all patients. transverse magnetization have the ability to acquire very thin (1 mm or
This sequence allows critical assessment of the midline structures that less) contiguous images through the brain in a relatively short imaging
are frequently abnormal in congenital brain malformations. Sagittal time. These techniques are particularly useful in looking for small, subtle
images are optimal for evaluating the corpus callosum, pituitary gland, cortical malformations (see Chapter 5) and for subtle areas of T1 short-
hypothalamus, and cerebellar vermis, common locations of pediatric ening (injured brain) in neonates and young infants (see Chapter 4).
brain tumors. They are also excellent for assessing the lateral convexities As discussed earlier, they also have the important advantage that they

Barkovich_Chap01.indd 6 5/5/2011 11:43:32 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 7

can be reformatted in any plane (sagittal, axial, coronal, or any obliq- Low-field scanners (≤1.0 T) do not produce adequately T1-weighted
uity), allowing acquisition of only one T1-weighted sequence. We images with conventional spin-echo technique. The reasons for the
routinely obtain volumetric gradient-echo sequences in all neonates, poorer images are twofold. First, T1 relaxation times increase with
all patients with developmental delay, and patients being imaged for increasing static magnetic field strength; therefore, images obtained
epilepsy. For brain tumors, neuronavigation techniques are used at with identical parameters will be more T1-weighted at higher field
our institution, and these require volumetric techniques as well, for strengths than at lower field strengths. Second, lower field strength
T1-weighted images. As visualization of enhancement is important scanners, in general, are unable to achieve TEs as short as high-field
in these patients, we use slightly different parameters for postgado- scanners. The combination results in unsatisfactory T1-weighted spin-
linium imaging, with TR/TE = 34 milliseconds/minimum, flip angle echo images in infants scanned on most midfield and low-field scan-
70°, and partition size of 3 mm. If volumetric images cannot or are ners. Inversion recovery or 3DFT GE images should be used instead.
not acquired, T1 FLAIR or spin-echo sequences can be used. Although The parameters for these sequences are similar to those used at 1.5 T,
T1 FLAIR gives better T1 information than conventional spin-echo although the flip angle and TR in particular may require fine-tuning in
sequences, an SE 600/minimum sequence (similar to that outlined for order to maximize gray-white contrast.
the sagittal images) can be routinely used for T1-weighted images on Some authors have advocated the use of T2 FLAIR sequences to
1.5 T scanners. Spin echo is the sequence of choice when paramagnetic look for regions of abnormal T2 prolongation. In our experience and
contrast is to be administered. Use of this imaging sequence produces that of others (81), FLAIR images are not sensitive to cerebral pathol-
excellent images; moreover, the imaging time is shorter than with most ogy in neonates or infants but are useful in older children in whom
inversion recovery sequences, resulting in less motion artifact. Spin- myelination is complete or nearly complete. We do not use FLAIR as a
echo T1 images do not work as well at 3 T due to the prolongation primary sequence in infants, but we do sometimes use it as a second-
of T1 relaxation times, however. Therefore, T1 FLAIR (usually using ary sequence for lesion characterization. In children beyond the age of
short TEs in the range of 20 milliseconds and TIs of 750 milliseconds) 2 years, when myelination is nearly complete, FLAIR becomes a part of
sequences are used; these give excellent T1 weighting but do not show our routine protocol because of its high sensitivity for subtle supraten-
contrast enhancement as well as spin-echo images or spoiled gradient- torial lesions, particularly in the cerebral cortex and periventricular
echo images. white matter. Sargent and Poskitt found that FLAIR is complementary
For T2-weighted sequences, dual spin-echo sequences with a 4 to to T2-weighted infants and children (82). They found fast spin-echo
5 mm slice thickness (2–2.5 mm gap), TR of 2500 to 3000 milliseconds, FLAIR to have better CSF nulling and better gray matter–white matter
TE of 30 to 60 milliseconds (first echo) and 70 to 120 milliseconds (sec- differentiation than echo-planar FLAIR. Our standard parameters for
ond echo), 192 × 256 acquisition matrix, and 0.75 to 1 excitation are 2D FLAIR imaging are TR/TE = 10,000/140 milliseconds, TI = 2200
recommended. In infants less than 12 months old, heavily T2-weighted milliseconds, with 3 mm contiguous slices and 192 phase encoding
sequences are highly recommended, as the water content of the brain steps. More recently, we have started to use single slab 3D FSE FLAIR
in young children is considerably higher than in older children and sequences (CUBE, SPACE, VISTA, depending on the manufacturer),
adults (76,77). We routinely use TR = 3000 milliseconds and TE = 60, which combine parallel imaging and variable flip angle excitation to
120 milliseconds (first and second echoes) for this patient group. convert slowly decaying longitudinal magnetization back to transverse
Others use a dual-echo short tau inversion recovery (STIR) sequence magnetization. These have the benefit of very high spatial resolution
with TR = 5400 milliseconds, TI = 130 milliseconds, and TE = 30, of 1 to 2 mm and relatively short imaging times of 7 to 8 minutes,
128 milliseconds (52). Although RARE (also called fast spin echo [FSE] and like volumetric gradient-echo T1 images can be reformatted in
or turbo spin echo [TSE]) images are used by some groups for infants any plane. However, longer imaging times render these images more
and children and seem to show myelination with fair accuracy (78), we susceptible to artifacts from patient motion, and in our experience the
and others (52) are not completely satisfied with the contrast between tissue contrast of these sequences does not always compare favorably
gray and white matter of commercially available RARE sequences, espe- to 2D FLAIR sequences.
cially with long echo train lengths. Contrast between gray and white A significant drawback of T2-weighted and FLAIR sequences is
matter, and between normal and pathologic tissues, is less in infants longer imaging times when compared to T1-weighted imaging. Some
than in older children and adults; therefore, imaging parameters and extremely fast MR techniques can be used in selected cases without seda-
sequences must be adjusted to maximize contrast. Moreover, although tion. These include the previously discussed single-shot RARE (called
missing a single small white matter lesion in an adult is relatively ssFSE or HASTE by manufacturers) (5,6) and periodically rotated
unimportant, the same type of abnormality may be the only clue to the overlapping parallel lines with enhanced reconstruction (called PRO-
correct diagnosis in a child (79). Therefore, we continue to use conven- PELLER or PROP) (7,8). These techniques are currently useful only for
tional spin-echo technique in infants with nonspecific developmental gross assessments such as ventricular size in patients with hydrocepha-
delay. However, we do use RARE sequences to supplement our standard lus or follow-up of extraparenchymal fluid collections. PROPELLER
images; they are ideal for obtaining T2-weighted images in a second has the advantage of better contrast to noise, more flexible contrast,
plane for infants with focal neurologic signs or symptoms and for eval- and the ability to compensate for motion by retrospectively correcting
uation of congenital anomalies or tumors (80). In addition, children the acquisitions (called “blades”), but has the disadvantage of slightly
who are imaged without sedation can benefit from the shorter acquisi- longer acquisition times (7); in our experience, tissue contrast is not as
tion time of RARE sequences. Typical parameters include TR = 2500 good in PROPELLER sequences as in conventional spin-echo or STIR
to 3000 milliseconds (up to 4000 milliseconds in infants <12 months sequences. For half-Fourier single-shot RARE images, we use TR/TE =
old), effective TE = 17 to 100 milliseconds, and echo train length = 8. 20,000/90 milliseconds, 0.5 excitations, 24 cm field of view, 256 × 256
We also use 3D RARE sequences to assess for subtle cortical anomalies acquisition matrix, and 4 mm slice thickness. For the PROPELLER
in infants at times when subcortical white matter is isointense to cortex FSE sequence, we use TR/TE = 4000/83 milliseconds, 2 excitations,
on our volumetric T1-weighted images (usually between ages 3 and 8 24 cm field of view, 224 × 224 acquisition matrix, and 4 to 5 mm slice
months). For this technique, we use TR = 4000 milliseconds, TE = 85 thickness.
milliseconds, echo train length = 16, partition size = 1.5 mm, and a 192 After sagittal and axial images have been obtained, coronal images
× 256 acquisition matrix. through the brain may be helpful in certain patients for further

Barkovich_Chap01.indd 7 5/5/2011 11:43:33 PM


8 Pediatric Neuroimaging

elucidation of pathology. Coronal images are particularly helpful for invasive and more difficult. A normal ultrasound is sufficient to exclude
imaging of the cerebellum, the temporal lobes, and the skull base. They most developmental spine pathology in neonates and young infants,
are also very valuable in the imaging of tumors, where the relationship but becomes less useful in older infants and children as the posterior
of the mass to the surrounding brain and dura is of great importance elements of the spinal column ossify (see Chapter 9). Moreover, it is
to the neurosurgeon. Our primary coronal sequence is the reformat- still not clear that sonography can consistently detect a fatty filum if
ted T1-weighted 3DFT volumetric spoiled gradient-echo sequence the conus medullaris is at a normal level (89). Moreover, even if sonog-
described above. For seizure and tumor protocols, and between the raphy detects a developmental spine anomaly, additional pathology is
ages of 3 and 8 months when gray-white differentiation is especially picked up by MR in 20% of cases (89).
difficult on T1-weighted images, 3D RARE sequences are added to In MR imaging of the spine, different techniques are used depend-
obtain supplementary T2-weighted coronal images. ing upon the clinical situation. When scanning a child with suspected
T2*-weighted gradient-echo images and susceptibility weighted or confirmed spinal dysraphism, sagittal T1-weighted images (TR/TE =
images (SWIs) are sometimes useful in pediatric imaging, particularly 600 ms/min) should be obtained initially using 3 mm slice thickness.
when looking for areas of old hemorrhage, as in suspected trauma or Axial images should then be obtained through any areas of abnormal-
vascular malformations (83–85). Beyond infancy, these images may ity or suspected abnormality. We acquire both T1-weighted and RARE
sometimes also provide the only clue on MRI that calcification is pres- T2-weighted axial images (note, however, that use of RARE sequences
ent, for example, within a brain tumor or as the result of congenital at 3 T in infants may result in high specific absorption rates, which
infection. Whereas short TR, short TE (<15 milliseconds) GE acqui- slow acquisition). The T1-weighted images show the spinal cord with-
sition results in primarily T1 weighting, short-TR, long TE (25–100 out the associated artifacts from circulating CSF that are so common
milliseconds) GE acquisition results in T2* weighting. The T2*- on T2-weighted images of the spine and will nicely show lipomas; the
weighted GE images are particularly useful in the evaluation for hem- T2-weighted RARE images nicely show the cauda equina, the filum ter-
orrhage because they are quite sensitive to the magnetic susceptibility minale, adhesions (such as meningocele manqué, see Chapter 9), and
changes and local heterogeneity of magnetic fields caused by the blood any bony or fibrous spurs. Another approach to the problem of spine
breakdown products hemosiderin and ferritin. Images show marked imaging is to perform 3D spoiled GE or RARE volumetric imaging
signal loss in the regions of the blood products (see Chapters 4 and (90). Weinberger et al. suggested using a TR of 500 milliseconds, TEeff
12). SWI, which uses the magnitude and phase images from volumet- of 21 milliseconds, ETL= 8, echo spacing = 21 milliseconds, 256 × 256
ric long-TE GE acquisition, shows even more dramatic loss of signal in plane matrix, 20 to 32 cm FOV, 1.0 mm sagittal partition thickness,
intensity in regions with calcium or blood products than do conven- receive bandwidth of 16 kHz, and one signal average. They typically
tional magnitude-only T2*-weighted GE images (86); however, paren- obtained 32 partitions in each slab (minus 4 peripheral slices). One
chymal contrast is reduced, and acquisition time is much longer, up to potential problem with 3D acquisition in the spine, however, is the
8 minutes. Recommended technique is TR/TE = 57/40 milliseconds, addition of a second phase-encoding direction that can result in signal
flip angle 20°, Nz = 32 slices, slice thickness = 2 mm, and matrix size being mismapped onto the spine from moving protons in the chest or
of 256 × 512. New versions that use echo-planar acquisition are being abdomen.
developed that should shorten the acquisition time (83). Patients with scoliosis present a challenge for any imaging modal-
We routinely obtain diffusion-weighted images in nearly every ity. Initially, a coronal T1-or T2-weighted image should be obtained
patient that we scan. The technique is a single shot using TR/TE = to assess vertebral anomalies and look for a split cord malformation
8000 milliseconds/minimum, BW = 166.7 kHz, FOV = 36 cm × 27 cm, (Chapter 9). Oblique sagittal (parallel to a straight segment of spine)
slice thickness = 3 to 5 mm. We use b = 600 s/mm2 in prematurely and oblique axial (perpendicular to a straight segment of spine, see
born babies scanned before term, b = 700 s/mm2 in neonates born at Chapter 9) planes can then be defined to give optimal information. If
term, and b = 1000 s/mm2 in infants and children older than 3 months a split spinal cord is seen, thin-section axial T2- or T2*-weighted GE
adjusted age. Because these images are acquired rapidly, there is very images should be obtained through the entirety of the split to look for
little time penalty incurred by measuring diffusion in six or more direc- a fibrocartilagenous or calcified spur. Alternatively, a 3D spoiled GE or
tions to improve the numerical accuracy of the average diffusivity (Dav RARE sequence, as in the prior paragraph, can be obtained; each seg-
or apparent diffusion coefficient [ADC]) and to allow calculation of ment of the spine can be displayed quite well after postprocessing.
diffusion tensors, which consequently permit analysis of white matter Patients who present with myelopathy but no suspicion of dysra-
pathways (diffusion tensor imaging, see below) (87,88). phism should have sagittal 3-mm RARE T2-weighted images (TR =
3500 milliseconds, TEeff = 102 milliseconds) through the entire spinal
cord, in addition to the T1-weighted sagittal and axial images. Axial GE
Spine
or RARE (TSE, FSE) images can then localize any abnormal region of
MR is the study of choice for imaging the pediatric spine in the setting T2 prolongation within the cord. We prefer GE (TR = 600 milliseconds,
of neurological dysfunction. CT is a useful exam in the setting of acute TE = 25 milliseconds) images in the cervical and upper- to midthoracic
trauma without neurological deficits and can be useful for evaluating spine, where rapid CSF flow causes considerable artifact on axial FSE
vertebral body or craniocervical junction anomalies. CT myelography images. Axial RARE images are usually better in the low thoracic and
may be necessary when the spine has been instrumented for scoliosis, lumbar spine. In either case, the use of flow-compensating readout gra-
as the resulting metallic artifact sometimes renders MR useless (the dients can help reduce these artifacts. T1-weighted images after infusion
use of titanium instrumentation will markedly reduce the susceptibil- of IV paramagnetic contrast are often very helpful for evaluating intrin-
ity effects, but it is not yet widely used). Optimally, multidetector spiral sic spinal cord pathology, particularly when neoplasm is suspected.
scanning should be performed to increase speed while still allowing Standard 2D spin-echo images are most often used for the postcontrast
adequate coverage and multiplanar reformations (75). Best results are scans; however, when patient motion is not a problem, 3DFT gradient-
obtained by acquiring the study with relatively low (<1) pitch values, echo postcontrast images with multiplanar reformations may be slightly
narrow (0.75 mm) collimation, and reconstructing 3-mm thick slices more sensitive to small foci of CSF tumor spread (6).
(75). However, when neurological deficits are present, CT is only use- To look at bone injury (pars interarticularis stress reaction or
ful after the injection of intrathecal contrast and is, therefore, more edema from a fracture) or tumor involving the vertebrae, sagittal

Barkovich_Chap01.indd 8 5/5/2011 11:43:33 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 9

3 mm STIR sequences are very useful. In our experience, they are more When evaluating the fetal brain, the optimal slice thickness is
sensitive than fat-suppressed RARE or contrast-enhanced T1 images 3 mm with no interslice gap; this slice thickness allows adequate signal-
and well worth the slightly increased imaging time. We use TR = 5000 to-noise without excessive volume averaging in the small fetal brain.
milliseconds, TI = 150 milliseconds, and TE = 58 milliseconds with For the fetal spine, we have found a slice thickness of 2 mm to be ideal
ETL = 8 and 2 excitations. for identifying small structures such as the spinal cord, bony spurs, and
The portions of the spine that should be imaged depend not only myelomeningoceles. Images are acquired during free maternal breath-
on the clinical indication for the study but also on coil geometry and ing, although we have found respiratory triggering useful in decreasing
total imaging time. At many institutions, a dedicated spine coil can motion. Field of view is typically small, although should be adjusted for
be useful for cases in which the entire spinal axis requires imaging increased fetal and/or maternal size, and when aliasing artifacts obscure
(e.g., drop metastases or scoliosis) and eliminates the need to switch important structures. Our routine imaging parameters are TEeff = 90
coils in the middle of the examination. In premature or young infants, milliseconds, TR = 4000 milliseconds, bandwidth = 25 kHz, matrix =
an adult head coil may provide sufficient coverage of the entire spine. 192 × 160, field of view = 24 cm, and number of excitations = 0.5.
To reduce the total time necessary for the examination and the associ- Images are acquired in an interleaved manner in order to reduce the
ated motion artifacts that come with long imaging times, automated possibility of signal loss resulting from crosstalk between slices. On
techniques for breath-hold, high-resolution gradient-echo survey of some scanners, a pause of 1 second between acquisitions of images may
the entire pediatric spine have been proposed (91). be needed to allow sequential scanning in an interleaved manner.
Some manufacturers have developed interactive scanning pro-
grams that can be applied to fetal MRI. These programs allow adjust-
Fetal MRI
ment of scanning parameters, such as slice angle and orientation, in
MRI is rapidly becoming the study of choice when fetal sonography “real time” (95,97). Such programs allow the technologist to adjust the
shows abnormalities, both in order to verify the presence of a suspected angle of a prescribed image without having to exit and reprogram a
abnormality and to specify the severity and extent of the abnormality new sequence; they are invaluable when the fetus is active (moving)
(92–96). With adequate attention to detail, excellent images of the fetal during the scan. This ability is particularly critical when trying to
brain and spine can be obtained. The study begins before the gravid acquire high-quality images of midline structures. Moreover, in cases
mother arrives at the MR scanner, as it is important that she does not of aliasing artifact, the field of view can be increased and/or the phase
take any oral nourishment for 4 hours prior to the MR examination. encoding and frequency encoding directions can be changed during
The lack of food reduces the amount of intestinal peristalsis, which image acquisition. Overall, image quality is improved, and scan time is
reduces the amount of misregistration artifact that is registered over reduced when this program is utilized.
the fetus. It also seems to reduce the amount of fetal motion. Upon Whenever possible, T1-weighed images are acquired to supplement
arriving at the MR site, the mother is asked to lie supine in the scan- the ssFSE (HASTE) T2-weighted images, but these are generally of lim-
ner and an 8-channel phased-array torso coil is applied for optimal ited value, especially in fetuses less than 27 gestational weeks, as the
imaging. If the mother cannot tolerate the supine position (because of signal-to-noise is invariably poor. Fast, multiplanar, spoiled gradient-
back pain or compression of the inferior vena cava, not usually a prob- echo techniques are primarily used, mainly to detect hemorrhage or
lem until after 30 weeks), the exam can be performed with her in the calcification; both are seen as hyperintense compared to the fetal brain.
left lateral decubitus position (lying on her left side). In our practice, T1-weighted images cannot be programmed as single-shot images in a
excellent studies are typically produced without using maternal or fetal rapid manner and require longer acquisition times (18 seconds). Scan-
(via the umbilical cord) sedation (93). ning parameters include TR = 120 milliseconds, TE = minimum, flip
Once the mother is positioned comfortably, a series of ultrafast angle = 70°, field of view = 24 cm, matrix = 256 × 160, number of exci-
T2-weighted ssFSE (HASTE) images are acquired, complemented tations = 1, slice thickness = 5 mm, interslice gap = 1 mm, and bandwidth
by echo-planar gradient-echo images and, whenever possible, by = 31.25 kHz, which yields eight slices in the axial plane. If possible,
T1-weighted images. The ultrafast T2-weighted images are the most the images are acquired while the mother holds her breath. However,
important diagnostically, as each image can be acquired in less than there is usually some maternal motion during the breath-holding,
1 second, reducing the likelihood of fetal motion during image acqui- and, as a result, this sequence is more susceptible to both maternal
sition. An initial localizer is obtained in three orthogonal planes (from the breath-hold) and fetal (from the duration of the sequence)
with respect to the mother, using 6- to 8-mm thick ssFSE (HASTE) motions. Other techniques for obtaining T1-weighted images have
T2-weighted slices with 1 to 2 mm gap and a large (40 cm) field of view. been explored, but all are subject to the limitations resulting from the
The localizer is useful for visualizing the position of the fetus, deter- long T1 relaxation time of the fetal brain (poor tissue contrast and
mining the location of the placenta within the uterus, and determining either long acquisition time or low signal-to-noise). Gradient-echo
which side of the brain is located on the side of the heart (left) and echo-planar T2*-weighted images are routinely acquired and can be
which is on the side of the liver (right). It is usually then easy to figure useful in detecting hemorrhage.
out which side of the brain is located posteriorly (closer to the maternal Advanced MR techniques such as diffusion-weighted imaging,
spine) and which side is located anteriorly. Importantly, the localizer spectroscopy, and parallel imaging are being successfully applied
is also used to ensure that maximal signal is obtained from the area to fetal MRI, although their development is still in the early stages
of interest. In certain cases, such as imaging of twins and fetuses with (98,99). Diffusion-weighted imaging provides quantitative informa-
myelomeningoceles, the coil may need to be repositioned in the middle tion about water motion and tissue microstructure. Single-shot echo-
of the examination (e.g. when switching from one twin to the other, or planar diffusion-weighted images are acquired in 18 seconds during
from the fetal brain to the spine). The initial diagnostic ssFSE (HASTE) a single maternal breath hold (98,99). Scanning parameters include
T2-weighted images of the fetal brain are prescribed from this local- TR = 4500 milliseconds, TE = minimum, field of view = 32 cm, matrix =
izer. Each subsequent image set is prescribed from the preceding set by 128 × 128, slice thickness = 5 mm, interslice gap = 2 mm, and band-
determining a plane orthogonal to that set. In this manner, axial, sagit- width = 167 kHz. Gradients are applied in three orthogonal direc-
tal, and coronal image sets of the fetal brain are obtained. We routinely tions using a b-value of 0 and 600 s/mm2. Because of the relatively
obtain at least two image sets of good quality in each plane. long scan time, images are susceptible to both fetal and maternal

Barkovich_Chap01.indd 9 5/5/2011 11:43:33 PM


10 Pediatric Neuroimaging

motion. With increasing gestational age and engagement of fetal head those in adults, however, and gadolinium-enhanced MRA may have
in the pelvis, the amount of motion is decreased and the quality of the added value in children with underlying vasculopathy or in whom the
studies improves. Diffusion-tensor imaging (DTI) is currently only normal neck circulation is distorted by tumors or infections. Difficult
applicable when the fetal head is already engaged in the pelvis (100), peripheral venous access and differences in hemodynamic parameters
because otherwise too much fetal motion occurs during the acquisi- require that bolus injection protocols be modified for safe application
tion time. Similar troubles, in addition to the large size of the voxel to pediatric studies, however. Newer techniques for balancing spatial
relative to the fetal brain size, plague fetal MR spectroscopy. There- and temporal resolution, such as time-resolved imaging of contrast
fore, proton MRS is limited to application during the latter half of kinetics (TRICKS), may prove useful for arterial-venous separation
the third trimester, when the fetal head is relatively large and engaged (109).
in the pelvis. Hopefully, the advent of faster sequences and improved Phase-contrast (PC) techniques are useful for imaging slow flow
coil design will allow these techniques to be more widely used in the within vessels, analyzing flow direction, quantifying flow parameters,
near future. and reducing saturation effects from slow flow (101,110). In pediat-
ric neuroimaging, multislice 2D PC is most often used to determine
flow direction, as it is significantly quicker than 3D PC imaging. 2D PC
SPECIAL MR TECHNIQUES studies are performed using a GE 27/4.5 sequence, a 192 × 256 acquisi-
MR and CT Angiography tion matrix, slice thickness 5 mm, flip angle 20°, and 15 acquisitions.
The image is obtained using a presaturation band technique. Encod-
For studies of the intracranial vasculature, 3D Fourier transform time ing velocity (Venc), the velocity at which there is a phase shift of 180°
of flight (3D TOF) studies using multiple overlapping time of flight between acquisitions, is typically set at 80 cm/s for arterial studies and
acquisitions (MOTSA) are the most useful (101,102). For 3D TOF at 30 cm/s for venous studies.
studies, a GE 45/min (TR/TE) sequence is performed with flip angle Studies of the dural venous sinuses are best achieved using the
20°, partition thickness of 0.9 mm, 128 partitions, and 1 acquisition. A TRICKS technique described above. 2D TOF venograms (111) can still
superior saturation band is applied to eliminate, as much as possible, be used but are less reliable, as complex or in-plane flow can give equiv-
contamination of the images by flowing blood from venous struc- ocal results. 2D TOF studies are acquired in the coronal plane to avoid
tures. Flow compensation gradients are applied in the read and phase- saturation effects in the transverse sinuses secondary to in-plane flow.
encoding directions. After data acquisition is completed and each of Contiguous 1.5-mm GE 45/4.9 images are obtained using a flip angle
the partitions is reconstructed, separate MR angiograms of each major of 60°, 256 × 160 matrix, and 1 excitation. Note that shorter TEs result
intracranial arterial circulation (right internal carotid, left internal in less in-plane saturation of protons and, therefore, in fewer appar-
carotid, basilar) are created by means of drawing a region of inter- ent “flow gaps” within the sinuses (112). Walking saturation pulses are
est around each and creating maximum-intensity projections (MIPs) applied posteriorly for coronal acquisition, and an inferior saturation
using the technique described by Laub and Kaiser (103). Creating a slab is applied to saturate arterial inflow. As with arteriograms, regions
separate MIP for each circulation is critical in order to avoid overlap of complex flow and dephasing can sometimes be better assessed with
of vessels and to reduce artifacts. The reader should remember that the postcontrast studies, although it is important to recognize that many
process of creating the MIP generates a number of artifacts (104,105) and smaller veins will be visualized after contrast administration and that
that, therefore, the individual partition images should always be scruti- these smaller veins may obscure some details. Recently, we have begun
nized in every MRA or MRV. Sometimes, multiplanar reformations of to use contrast-enhanced 3D venography using a 3D gradient-echo
the partition images are more useful than the MIP images for assessing technique (TR = 1.74 milliseconds, TE = 0.64 milliseconds, flip angle
vascular pathology. 15°, 128 × 128 matrix, 25 cm FOV, 64 sagittal 2-mm sections with zero
For studies of the cervical carotid and vertebral arteries, 2D Fou- filling of k-space in the partition direction) (113,114) as our routine.
rier transform time of flight (2D TOF) images are fast and efficient The first dataset is discarded, and magnitude subtraction of the sub-
(101,106,107) and are usually quite adequate in children because the sequent volumes from the second dataset is performed to remove
vessels are typically straight and stenoses (which make flow more com- background signal intensity. This technique requires a rapid injection
plex and give rise to artifacts with 2D TOF) are uncommon. Contrast- of paramagnetic contrast and timing of the image acquisition to the
enhanced MRA or 3D TOF MOTSA sequences can be used in the neck entrance of the contrast into the superior sagittal sinus. The advantages
for higher resolution in specific areas. 2D TOF is also useful in place of of the contrast-enhanced, dynamic 3D data set are that no artifactual
3D TOF for studies of intracranial vessels when complicated by exces- flow gaps are present and that specific portions of the venous sinuses
sive patient motion, although it is primarily used for imaging of vessels can be carefully analyzed by reconstructing 1 to 2 cm slabs of data. For
in the neck and for venography. We perform 2D TOF studies using a GE example, midline vessels can be carefully evaluated by reconstructing
4.5/min sequence. Fifty-five consecutive axial MR images are obtained a sagittal slab of the central 2 cm and the transverse sinuses by recon-
with a section thickness of 1.5 mm, flip angle 60°, and bipolar gradients structing axial slabs.
for flow compensation. Walking superior saturation pulses are applied CTA has become a useful tool with the advent of helical, multidetec-
cephalad to the axial images to eliminate signal intensity from venous tor CT scanners (75). However, it should be reserved for cases in which
blood returning via the jugular veins. MIP images of the right carotid/ Doppler sonography and MRA are not diagnostic, as the radiation dose
right vertebral circulation and separate MIP images of the left carotid is high. A high-quality CTA requires contrast administration in a tight
vertebral circulation are reconstructed in multiple planes. bolus of iodinated contrast through a large gauge IV catheter (usu-
In adults, 3D TOF MRA with bolus injection of contrast allows ally with a power injector) and properly timed acquisition of images
production of high-resolution MRA images with minimization of through the area of interest. In order to acquire images while contrast
saturation and dephasing artifacts (108). Although there have now is in the arteries but not yet in the veins, a test bolus is administered and
been a number of studies describing high-quality depiction of the images are obtained through a single level until the bolus arrives. The
great vessels in the chest and abdomen using this technique in chil- time required for this bolus to arrive determines when image acqui-
dren, we are not aware of any work on its utility for imaging the pedi- sition begins after the bolus injection is begun. Alternatively, when
atric cervical vessels. It is expected that the results would be similar to obtained as a part of the examination, perfusion CT can be used to

Barkovich_Chap01.indd 10 5/5/2011 11:43:33 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 11

estimate the optimal timing of the bolus. Helical acquisition CTA on Magnetization Transfer
our scanners consists of 16 sections at 0.75 mm collimation, 0.5 sec-
ond rotation time, and pitch of 1.5 using a small (typically 10–15 cm, The technique of magnetization transfer contrast (MTC) is useful
depending upon patient size) field of view (75). 2D and 3D reforma- in assessing myelination and demyelination (121,122). MTC imag-
tions can then be constructed to optimally show the blood vessels of ing relies on detecting differences in relaxation properties of free and
interest. Both CTA and MRA are accurate and easy; both show the ves- bound water (122–124). The technique is based on the fact that water
sels of the neck, the dural venous sinuses, and circle of Willis quite well. protons bound to macromolecules (such as those composing myelin)
However, considering the considerable dose of ionizing radiation from have very short T1 and T2 relaxation times; thus, bound protons do
CTA, MRA is probably a better initial study in children, even when not normally contribute to the signal obtained by MR imaging. How-
sedation is necessary. At UCSF, we only perform CTA if the necessary ever, a constant exchange of water molecules is occurring between the
diagnostic information cannot be obtained by other methods. free water molecules and the bound water molecules. Those protons
in water molecules that are bound to macromolecules at the time of
the initial radiofrequency pulse and separate from the macromolecule
CSF Flow Imaging before the readout gradient is applied will have considerably shorter
CSF flow can be visualized and quantitatively analyzed using PC stud- T1 and T2 relaxation times than the rest of the free water proton mol-
ies (115,116). MR studies of CSF flow are most useful when looking ecules. The measured relaxation times of the entire population of water
for interruptions of normal flow patterns. In particular, we use these protons will be shortened by this process, which is known as magne-
studies in patients with Chiari I and II malformations (see Chapter 5) tization transfer. The amount of magnetization transfer will depend
when they are being considered for foramen magnum decompression upon the quantity of macromolecular components available to bind
surgery. We use PC sagittal studies because the CSF flow can be quanti- within the free water molecules and the rate of exchange between the
fied (116,117). For studies of patients with Chiari malformations, the two pools of water molecules.
images are centered at the foramen magnum. Our typical parameters If an RF pulse is applied slightly (5–10 kHz) off the peak resonance
include Venc of 5 cm/s, TR = 27 milliseconds, TE = 11.7 milliseconds, frequency of free water, it will saturate the bound water protons, which
flip angle 30°, 22 cm field of view, a 128 × 256 acquisition matrix, and have a much broader absorption peak, with minimal effect upon the
2 excitations. Peripheral cardiac gating is used. free water protons. Application of the off-resonance RF pulse, there-
If only qualitative information is desired about CSF flow, other fore, will negate any contribution of bound water protons to the overall
flow-sensitive sequences can be used. Steady-state free procession T1 and T2 relaxation times and to the MR image is negated. Subtrac-
sequences (CISS or FISP or SSFP) using 3D Fourier transformation tion of the image obtained with the off-resonance RF saturation pulse
can be very useful to show rapid CSF flow that disturbs the steady will show the contribution from the bound protons, that is, the amount
state and, therefore, appears as low signal intensity. We use a TR of of magnetization transfer. The amount of magnetization transfer can
30 milliseconds, TE of 14 milliseconds, flip angle 40°, and 200 phase be assessed qualitatively or quantitatively (125).
encoding steps. Using this technique, 128 partitions and an 18 cm Magnetization transfer images can be obtained by acquiring
field of view yield a partition size of 1.4 mm. Because of the 3D acqui- two sets of gradient-echo scans with gradient spoilers. Data are best
sition, the images can be formatted in any plane. RARE and FLAIR acquired using a TR of 300 milliseconds, TE of 7 milliseconds, flip
sequences are also motion sensitive and can be used to qualitatively angle 20°, 3 mm partition thickness, 12 cm field of view, and 3DFT
evaluate CSF flow. reconstruction techniques. A RF saturation band applied to the sec-
ond set at 5 kHz off the resonance frequency of free water will satu-
rate the pool of bound protons. Subtraction of the second data set
Magnetic Resonance Neurography from the first results in a magnetization transfer image. The amount
RARE and STIR sequences can be used to highlight nerve fascicles as of magnetization transfer in the different regions of the brain, which
they course through the brachial plexus, arms, legs, or neck and are should reflect the state of myelination, can be calculated from region
particularly useful for evaluating these structures in cases of trauma, of interest measurements from those regions of brain (MTR =
inflammatory or radiation injuries. Either sequence should use long [Mo − Ms]/Mo, where MTR is the magnetization transfer ratio, Mo is
TR (4000–5000 milliseconds) and long TEeff (100–110 milliseconds) the magnitude of signal without saturation by an off frequency radiof-
and flow saturation bands on all sides of the imaging volume (to sup- requency pulse, and Ms is the magnitude of tissue signal with the satu-
press high signal from incoming flow). If RARE sequences are used, ration pulse on).
fat suppression must be applied as well. In general, STIR sequences
work better for the neck and brachial plexus, as fat suppression pulses
Perfusion Imaging
are often affected by bulk susceptibility in this region, resulting in het-
erogeneous fat saturation and water saturation; STIR alleviates these Perfusion imaging in infants and young children is difficult. Limita-
problems. If fat-suppression pulses are used, for the neck or brachial tions include difficulty in gaining proximal IV access, the use of small
plexus, water bags, oil bags, or commercially available neck pads should IV catheters that limit injection rates and prolong injection times,
be used to help reduce such artifacts. Alternatively, chemically selec- the necessity of using low doses of contrast, and a high incidence of
tive radiofrequency excitation pulses or modified Dixon techniques motion artifacts. If these limitations can be overcome, the potential
(IDEAL, iterative decomposition of water and fat with echo asymmetry exists for gaining useful information, especially in older children, using
and least squares estimation) may help achieve more homogeneous fat this method.
suppression (118). For all sequences, we acquire sections of 3- to 4-mm With the widespread availability of helical CT scanners, CT per-
slice thickness with no interslice gap. The acquisition matrix should be fusion imaging has become feasible and is now commonly used in
no less than 256 × 256; if possible, a matrix size of 512 × 512 should adults (126,127). However, because of the limitations mentioned in
be used. The resultant images highlight nerve bundles as longitudinal the previous paragraph and the associated radiation, CT perfusion has
bands of high signal intensity. These sequences are excellent for search- not gained widespread acceptance for use among young children. The
ing for intrinsic pathology in peripheral nerves (119,120). few studies employing this technique in children have focused on the

Barkovich_Chap01.indd 11 5/5/2011 11:43:33 PM


12 Pediatric Neuroimaging

variation in perfusion parameters with age (128) or on the use of the intensity as a function of tissue perfusion is small; measurements are,
CT perfusion in the emergency setting (129). therefore, easily impaired by head motion or instability of instrumen-
Several approaches exist for performing dynamic MR imaging tation. Spin tagging is therefore better performed using echo-planar
after administration of a contrast agent (130–133). One method is techniques, which reduce instrumental drift and head motion artifacts
the use of fast gradient-echo sequences with inversion prepulses. This (141), making the results more reproducible. Probably the best of the
sequence allows strongly T1-weighted sequences to be acquired in as current ASL techniques is called quantitative signal targeting by alter-
little as 1 second. Enhancement with paramagnetic contrast is mani- nating radiofrequency pulses labeling of arterial regions (QUASAR)
fested as an increase in brain signal intensity of the images. Alterna- (142). This sequence is capable of acquiring images at multiple TIs after
tively, T2*-weighted pulse sequences can be used. If EPI is available, it labeling of the arterial water protons with a high temporal resolution;
should be used, as it allows acquisition of images at multiple levels with it features a bolus saturation scheme for clear definition of the arterial
a single bolus administration of contrast. Gradient-echo images are blood bolus (142). The readout is performed by using conventional
used instead of spin echo because they are more sensitive to medium multisection single-shot gradient-echo EPI with a small flip angle.
to large vessels, and, therefore, greater signal drop is seen during the Typical parameters for QUASAR are four sections of 7 mm thickness;
first pass of a contrast agent. Although more prone to susceptibility ascending section order; 2 mm section gap; 64 × 64 imaging matrix;
artifact, gradient-echo techniques are more sensitive to small changes 24 cm field of view; 30° flip angle; TR of 4000 milliseconds; TE of
in blood volume. Therefore, the majority of the literature for tumor 23 milliseconds; first TI of 50 milliseconds, DTI of 200 milliseconds;
imaging favors using a gradient-echo sequence to estimate cerebral bolus length (start time for bolus saturation) of 1050 milliseconds;
blood volume (susceptibility weighted perfusion imaging). If echo- stop time for bolus saturation of 2250 milliseconds; 18 acquisition
planar techniques are not available, standard gradient-echo techniques time points (i.e., TIs); single-shot EPI; sensitivity encoding factor of
using sufficiently long TEs will work, as well. During first pass of the 3.0; inversion slab width of 15 cm; and inversion gap of 3 cm (143).
paramagnetic contrast agent through the brain, the shortened T2* Another approach to ASL is the technique of continuous ASL,
results in a loss of signal intensity (134). Region of interest intensity in which arterial spins are continuously labeled and a steady-state
measurements are acquired from the images sequentially to generate condition is measured in the cerebral parenchyma. This method has
a curve (called the DR2* curve) in which relaxivity (DR2*) is plotted suffered from unwanted effects of magnetization transfer and micro-
versus time (see Chapter 7). From the DR2* curve, mean transit time, vascular “noise” (138). A method has been introduced that assesses
relative blood volume, and relative blood flow can be deduced. Our changes in magnetization of multiple sections during continuous
experience and that of others (133) indicates that superior contrast ASL. This technique appears to overcome the magnetization transfer
to noise is obtained using the T2*-weighted sequence, so we use that effects and allows a quantitative brain perfusion study to be acquired
method to evaluate for brain ischemia and to assess tumors (see sec- noninvasively in 10 to 15 minutes (144). Although this duration may
tion at the beginning of Chapter 7). The DR2* curve can be used to dif- still be a little long for the study of a child, it seems to be a big step
ferentiate low-grade from high-grade gliomas, recurrent tumor from forward in the development of a noncontrast, quantitative MR perfu-
treatment-related injury, intraparenchymal from extraparenchymal sion technique.
tumors, and abscesses and demyelinating plaques from intraparenchy-
mal tumors (53,135,136).
Diffusion Tensor Imaging
The patients are imaged during infusion of paramagnetic con-
trast, given intravenously as a bolus injection. For EPI, we use a single- The availability of very strong, rapidly acting gradients on MR scanners
shot GE technique with TR/TE = 1250/54 milliseconds, bandwidth = has permitted the measurement of the net motion of water molecules
178 MHz, FOV = 26 × 26 cm, 4 mm slice thickness, acquisition matrix within the brain. These motions arise predominantly from diffusion,
of 256 × 128, and 1 excitation. This sequence allows acquisition of but also from the flow of blood in capillaries and probably active trans-
70 sets of images from 7 slice locations in 2 minutes. The imaging port of water with macromolecules within cells. The combination of
sequence for standard gradient-echo sequences utilizes TR/TE = 35/25 these motions causes a loss of signal when strong gradients are applied;
milliseconds, flip angle 10°, 1 excitation, 10 mm slice thickness, and this loss of signal can be measured and is called the ADC or average
256 × 64 acquisition matrix. This sequence allows acquisition of images diffusivity (Dav) (145). The Dav can be measured in any direction by
every 2 seconds, acquired over 30 seconds. As the cerebral circulation acquiring images in the presence of very large field gradients in that
time (including arterial, capillary, and venous phases) varies from 7 to direction; acquisition of a series of images with different strengths
9 seconds, 30 seconds provides adequate “buffer” capacity for the tim- of applied gradients allows quantification of the Dav (145). Although
ing. Data analysis can provide local blood volume, approximate local diffusion imaging is theoretically possible using standard spin-echo
blood flow, and perfusion delay measurements (131,133). Although and gradient-echo techniques, the high level of susceptibility to even
most of our experience with pediatric perfusion imaging has been in slight motion makes these techniques practically impossible. Therefore,
older children, the method can be also be safely used when specifically diffusion imaging is nearly always performed using EPI techniques.
tailored for infants (54,137). At UCSF, we use a single-shot technique with TR of 10,000 millisec-
Perfusion imaging can also be performed using blood water as an onds, minimum TE, with a 256 × 128 acquisition matrix, bandwidth
endogenous tracer. This technique is referred to as the spin tagging, of 166.7 MHz, 36 × 27 cm FOV, 3 to 5 mm slice thickness, and 1 exci-
arterial spin labeling (ASL), or black blood method (130,138,139); it is tation. Use of strong gradients (with appropriate “b” value [145]) is
entirely noninvasive and is theoretically quantitative. In spin tagging, critical to achieving good diffusion images. We utilize different b values
the spins of incoming protons are inverted at the level of the carotid depending on the age of the patient. For premature neonates, we use
arteries. A series of images through a section in the brain downstream b = 600 s/mm2; for term neonates, we use b = 700 s/mm2; and for older
from the site of inversion are subsequently obtained. A change in net infants and children, we use b = 1000 s/mm2. Diffusion gradients must
magnetization of the tissue in the plane being imaged will occur due to be applied in a minimum of three orthogonal planes in order to obtain
thin inflow of protons with inverted spins; this change in magnetiza- high sensitivity to changes in proton diffusion (in a minimum of six
tion is a function of perfusion of the tissue in that plane. Although this planes for DTI, see below). It is important to be consistent in the use of
technique works with spin-echo techniques (140), the change in signal diffusion gradients, especially if quantifying the ADC. Work has shown

Barkovich_Chap01.indd 12 5/5/2011 11:43:33 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 13

that the magnitude of the Dav varies with the b value utilized when the in children (87,166) and to help us better understand white matter
number of acquired directions is small (200). In addition, absolute Dav anomalies associated with developmental brain disorders (see Chapter
measurements should be interpreted with the knowledge that numeri- 2, Section “Diffusion Tensor Imaging and Tractography”) (167–170).
cal values may vary between 4% and 9% depending on scanner vendor, Finally, diffusion characteristics can be used to differentiate different
field strength, and sequence parameters (147). layers of the developing cerebral mantle in fetuses and prematurely
The work of Basser et al. (148,149) in the mid-1990s allowed born neonates (171). For example, the developing cerebral cortex had
widespread use of a more sophisticated method of diffusion imaging, different diffusion characteristics than the subplate (a transient layer
known as diffusion tensor imaging (DTI). DTI is really a mathematical beneath the cortex that is important in the establishment of permanent
description (the tensor) of water motion in a voxel. To determine the axonal connections within the developing brain), and both have differ-
diffusion tensor, it is necessary to acquire diffusion-weighted images in ent characteristics than the intermediate zone (the layer of developing
a minimum of six different planes, along with an image acquired with- axons and glia between the subplate and the germinal matrices) (171).
out diffusion weighting (i.e., b = 0). Once the diffusion tensor is calcu- The ability to distinguish these layers on imaging studies may be cru-
lated for each voxel, it is possible to calculate the Dav and the degree of cial to unraveling the cause of developmental difficulties in premature
anisotropy (asymmetrical water diffusion) of the motion of the water neonates.
molecules in that voxel (150). As it is difficult to display tensor data with A second potentially important aspect of diffusion imaging in
images, the concept of diffusion ellipsoids is often used. An ellipsoid is pediatric neuroradiology is that water appears to diffuse more and
a 3D representation of the distance and direction traveled in space by more randomly (i.e., there is greater diffusivity and less anisotropy)
the average water molecule in a voxel during a give time Td. The axes of in patients with developmental delay of unknown cause than in devel-
the ellipsoid are directed along the three orthogonal eigenvectors (n1, opmentally normal patients (172). The full significance of this finding
n2, and n3, the principal diffusion orientations) and the lengths of the has not been elucidated but the implication is that, not surprisingly,
vectors are scaled by the corresponding eigenvalues (l1, the primary abnormally formed or abnormally organized white matter does not
eigenvalue [sometimes also called the parallel eigenvalue, a measure function as well as normal white matter in transmitting action poten-
of longitudinal diffusivity], l2, the intermediate or median eigenvalue, tials through the brain.
and l3, the minor eigenvalue [note that (l2 + l3)/2 is sometimes called The third aspect of diffusion imaging that is of value in pediatric
the perpendicular eigenvalue, a measure of transverse diffusivity]). By neuroimaging is the ability to detect reduced diffusion in the acute
using the concept of the ellipsoid and its eigenvalues, it is possible to phase after a traumatic, metabolic, or toxic injury to the brain. For
discuss diffusion anisotropy relatively simply. Diffusion anisotropy is reasons that are not entirely clear, the injury results in an acute
generally expressed as relative anisotropy (RA), fractional anisotropy reduction in water motion (and hence the Dav) in affected regions
(FA) or the coefficient of variation of the eigenvalues (As). The relative of the brain. For this function, the “trace,” which is the average of
strengths and weaknesses of these parameters are still being worked out. the Dav in three perpendicular directions, is the most useful mea-
However, FA appears to be more sensitive to small differences among surement (note that trace is least sensitive to anisotropic differences
white matter tracts when anisotropy is low (as in neonates and infants) in Dav). An abrupt reduction in Dav is indicative of acute cerebral
(151) and has higher signal-to-noise ratio than RA (152); therefore, we injury. This aspect of diffusion imaging is discussed extensively in
use FA in studies of neonates and infants. Although most radiologists Chapters 3 and 4.
and MR scientists currently use RA, FA, or As to describe anisotropy,
it is important to realize that combining the individual eigenvalues to
Proton MR Spectroscopy
calculate these values leads to an overall loss of information and that, if
possible, it is optimal to calculate and analyze the individual eigenvalues Proton MR spectroscopy is useful in the assessment of encephalopathic
after acquiring DTI data (151). The details of DTI are beyond the scope neonates (173,174), in detection and diagnosis of some inborn errors
of this book. Interested readers are referred to the excellent review by of metabolism (175,176), in developmental delay (177), and in pre-
Le Bihan et al. (150), the reviews of Mukherjee et al. (153,154), or the therapeutic and posttherapeutic assessment of intracranial tumors
series of excellent reviews in the November to December, 2002, issue of (178,179). Single voxel spectra are obtained from volumes of approxi-
NMR in Biomedicine (155–159). mately 6 cm3, but 2D and 3D spectroscopy packages allow acquisition
Three features of DTI are of interest in pediatric neuroimaging. of spectra from voxels sized 1 cm3 (or smaller on 3 T scanners). The
First, diffusion characteristics can be used to assess brain maturation voxel is prescribed from MR images obtained in an imaging sequence
(151) because both the rate of water diffusion (Dav) and the direction during the same examination. A stimulated echo sequence (180)
of water diffusion (diffusion anisotropy) vary with maturation in both is combined with chemical shift selective pulses for localization and
immature white matter and in immature cerebral cortex (160,161). water suppression, yielding only a signal from the region of interest
In premature neonates, water molecules in the cerebral cortex move at the spatial intersection of three section selective pulses. Most mod-
more radially (perpendicular to the surface of the cortex) than hori- ern MR scanners now have “push button” spectroscopy sequences that
zontally (parallel to the surface of the cortex) (161); this anisotropy automatically perform shimming and water suppression. One hun-
disappears with maturation (see Chapter 2, Section “Diffusion Tensor dred twenty-eight averages are obtained to achieve adequate signal-
Imaging and Tractography” and Figure 2-35). In white matter, the rate to-noise. Short TE spectra (TR/TE = 2.0 seconds/20–30 milliseconds,
of diffusion parallel to axon fascicles is faster than the diffusion per- TM = 10.7 milliseconds) require use of the stimulated echo acquisition
pendicular to them (162,163). This anisotropy increases as the axons mode (STEAM) technique; they allow assessment of the peaks of many
myelinate, although, interestingly, anisotropy is present even before metabolites and are most useful for assessing patients with inborn errors
myelination (162,163). Measurement of both the Dav and the anisot- of metabolism (175). Long TE spectra (TR/TE = 2.0 seconds/272–288
ropy may be useful in assessing brain maturation (151,164,165) and milliseconds) are typically acquired using a point resolved spectro-
brain injury (81). In addition, tensor information in conjunction with scopic (PRESS) technique (which gives better signal-to-noise ratio than
connectivity algorithms allows axonal tracts to be mapped in the brain STEAM) and are more useful in injury states, in which optimal evalu-
(155); although currently a research tool, this diffusion “tractography” ation and quantification of lactate and N-acetylaspartate (NAA) are
has tremendous potential to identify the causes of neurologic defects needed (173,182), and in tumors, in which quantification of choline,

Barkovich_Chap01.indd 13 5/5/2011 11:43:33 PM


14 Pediatric Neuroimaging

creatine, and NAA is needed (183). Spectra are routinely acquired with reduces the local concentration of deoxyhemoglobin. The reduction
a standard imaging quadrature head coil, although signal-to-noise and of local deoxyhemoglobin results in an increase in local signal inten-
resolution can be improved by the use of specially designed pediatric sity. Subtracting the image acquired during the resting state from that
coils (46) or surface coils. obtained during the activated state results in an image that (indirectly)
When using 2D and 3D spectroscopic imaging techniques, mul- shows local brain activity. This technique, known as blood oxygen
tiple spectra from a variety of locations can be acquired during a single level–dependent (BOLD) imaging (194), is the primary technique used
acquisition. The location of the smaller voxels within a larger acquisi- for functional MR. Because the sensitivity to changes in the oxyhemo-
tion area can be modified retrospectively, allowing a great deal of flex- globin/deoxyhemoglobin ratio is increased at higher magnetic field
ibility in the regions sampled. The 2D spectroscopy is nearly as fast as strength, BOLD imaging is likely to become more widely used as more
single voxel acquisition, but the 3D currently increases the acquisition 3 and 4 T MR scanners become available.
time considerably (184,185). The use of higher field strength MR scan- fMRI in older children is performed in a manner identical to that in
ners with echo-planar and spiral acquisition techniques promises to adults (194–196). At 1.5 T, we use a single-shot spin-echo echo-planar
allow faster acquisition in the near future, which will likely make 3D sequence with TR/TE = 2000/60 milliseconds, acquisition matrix of
spectroscopy, using both long and short TEs, clinically viable sequences 256 × 128, 10 mm slice thickness, and one acquisition. We choose spin-
in the near future (186,187). Proton spectroscopy can currently be used echo echo-planar because it has greater sensitivity to signal arising from
to evaluate brain maturation (Chapter 2), to assess disorders of metabo- capillaries and venules, whereas gradient-echo echo-planar images are
lism (Chapter 3), to determine the severity of brain injury (Chapter 4), more sensitive to larger veins and have the potential for greater spatial
and to evaluate intracranial masses (Chapters 3 and 7). mismapping. In addition, spin-echo sequences have fewer susceptibility-
related artifacts (however, susceptibility is advantageous for BOLD
Automated Morphometric Analysis imaging techniques). Use of echo-planar sequences is essential in stud-
ies of children, as motion artifacts are typically more of a problem in
and Tissue Segmentation
children than in adults (197). Patient cooperation becomes a problem
The more widespread use of volumetric T1-weighted GE sequences in younger children, particularly those below the age of 8 years and
and the increasing availability of off-line, computationally intense those with developmental delay or mental retardation (198). In this
advanced processing tools have recently enabled the systematic study group, conditioning of the patients to the experimental situation by
of brain morphology using automated techniques. Although not yet going through the necessary tasks in a mock MR scanner can save con-
implemented clinically, techniques for analyzing cortical folding pat- siderable time when the actual experiment is performed (199). Pedi-
terns have shown promise for studying brain development and in the atric fMRI shows particular promise in the evaluation of disorders of
evaluation of disorders such as autism (188) and schizophrenia (189). language acquisition (200). In infants and young children, resting state
In the neonatal brain, twins and newborns with intrauterine growth fMRI shows particular promise as a means of assessing normal and
restriction show alterations in brain gyration and sulcation compared abnormal brain function. An introduction to this topic is presented in
to normal singletons that appear to correlate well with outcomes Chapter 2, Section “Functional MR Imaging”.
(190,191). Beyond assessment of folding, automated tissue segmenta-
tion allows the targeted study of cortical gray matter development in
the maturing neonatal (192) and even the fetal brain (193). More work
SUMMARY
will be necessary before these tools can be applied to study the individ- To summarize, the many differences between the pediatric and adult
ual patient, but in the future, automated evaluation of morphometry nervous systems require that different strategies be used for diagnos-
may serve as a useful adjunct to subjective interpretation. tic neuroimaging. Sedation is often crucial in acquiring diagnostic
images in children. MR-compatible life support equipment must be
suitable for pediatric patients if unstable patients are to be imaged
Imaging of Brain Function
in the hospital setting. The choice between neurosonography, CT,
A number of different techniques can be used to map the spatiotem- and MRI depends upon the age and clinical status of the patient, the
poral distribution of neuronal activity during specific cognitive states. indications for the study, and a consideration of radiation dose in the
These include magnetoencephalography (which, when mapped onto short and long term. The need for iodinated or paramagnetic contrast
an MR scan is referred to as magnetic source imaging), electroencepha- media should be determined, and quantities of contrast agents must
lographic mapping, positron emission tomography, and blood oxygen be adjusted. Imaging protocols must be modified to compensate for
level–dependent functional MR imaging (fMRI). Among these, fMRI the different water content, changes in amount of myelin, changes in
has several advantages. fMRI combines relatively high temporal and metabolite levels, and the different size of the pediatric brain. If these
spatial resolution without ionizing radiation or externally adminis- factors are considered and proper modifications are made to imaging
tered contrast agents. It also allows rapid and flexible experimental protocols, then functional and anatomic neuroimaging techniques can
protocols. Finally, fMRI allows functional and structural images to add valuable diagnostic information and greatly improve the medical
be acquired during the same examination; these images can easily be care of children with neurological disorders.
coregistered to produce maps that show the precise relationship of
function to anatomy. Functional MR data can be based upon local
cerebral blood volume, local cerebral blood flow (perfusion, see above), REFERENCES
or blood oxygenation. As the blood oxygenation–based methods are
1. Enriquez G, Correa F, Aso C, et al. Mastoid fontanelle approach for sono-
the most widely used, they are briefly discussed.
graphic imaging of the neonatal brain. Pediatr Radiol 2006;36:532–540.
The capillary and venous blood in the vascular network around 2. Luna JA, Goldstein R. Sonographic visualization of neonatal posterior
the normal resting cerebral cortex has a relatively high concentra- fossa abnormalities through the posterolateral fontanelle. Am J Roentgenol
tion of deoxyhemoglobin. When the cortex is activated, the increased 2000;174:561–567.
metabolic demand results in a nearly instantaneous increase in local 3. Pappas JN, Donnelly LF, Frush DP. Reduced frequency of sedation of young
blood flow that raises the local concentration of oxyhemoglobin and children with multisection helical CT. Radiology 2000;215:897–899.

Barkovich_Chap01.indd 14 5/5/2011 11:43:33 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 15

4. Zimmerman RA, Bilaniuk L, Harris C, Phillips P. Use of 0.3 mmol/kg 28. American Academy of Pediatrics Committee on Drugs. Guidelines for
gadoteridol in the evaluation of pediatric central nervous system diseases. monitoring and management of pediatric patients during and after seda-
Radiology 1993;189P:222. tion for diagnositc and therapeutic procedures. Pediatrics 1992;89:1110–
5. Patel M, Klufas R, Alberico R, Edelman R. Half-fourier acquisition single- 1115.
shot turbo spin-echo (HASTE) MR: comparison with fast spin-echo MR in 29. Greenberg SB, Faerber EN, Aspinall CL, Adams RC. High-dose chloral
diseases of the brain. Am J Neuroradiol 1997;18:1635–1640. hydrate sedation for children undergoing MR imaging: safety and efficacy
6. Sugahara T, Korogi Y, Hirai T, et al. Comparison of HASTE and segmented- in relation to age. Am J Roentgenol 1993;161:639–641.
HASTE sequences with a T2-weighted fast spin-echo sequence in the 30. Smith MT. Chloral hydrate warning [Letter]. Science 1990;249:359.
screening evaluation of the brain. Am J Roentgenol 1997;169:1401–1410. 31. American Academy of Pediatrics Committee on Drugs. American Acad-
7. Forbes K, Pipe J, Karis J, Farthing V, Heiserman J. Brain imaging in the emy of Pediatrics Committee on Drugs and Committee on Environ-
unsedated pediatric patient: comparison of periodically rotated overlap- mental Health: use of chloral hydrate for sedation in children. Pediatrics
ping parallel lines with enhanced reconstruction and single-shot fast spin- 1993;92(3):471–473.
echo sequences. Am J Neuroradiol 2003;24:794–798. 32. Kao SC, Adamson SD, Tatman LH, Berbaum KS. A survey of post-discharge
8. Pipe J. Motion correction with PROPELLER MRI: application to head side effects of conscious sedation using chloral hydrate in pediatric CT and
motion and free-breathing cardiac imaging. Magn Reson Med 1999;42: MR imaging. Pediatr Radiol 1999;29:287–290.
963–969. 33. Mason KP, Sanborn P, Zurakowski D, et al. Superiority of Pentobarbital
9. Sanborn PA, Michna E, Zurakowski D, et al. Adverse Cardiovascular and versus Chloral Hydrate for Sedation in Infants during Imaging. Radiology
Respiratory Events during Sedation of Pediatric Patients for Imaging Feb 2004;230(2):537–542.
Examinations. Radiology 2005;237(1):288–294. 34. Zgleszewski SE, Zurakowski D, Fontaine PJ, D’Angelo M, Mason KP. Is
10. Mathur AM, Neil JJ, McKinstry RC, Inder TE. Transport, monitoring, propofol a safe alternative to pentobarbital for sedation during pediatric
and successful brain MR imaging in unsedated neonates. Pediatr Radiol diagnostic CT? Radiology 2008;247:528–534.
2008;38:260–264. 35. Filippi C, Ulug A, Lin D, Heier L, Zimmerman R. Hyperintense signal
11. Harned II RK, Strain JD. MRI compatible audio/visual system: impact on abnormality in subarachnoid spaces and basal cisterns on MR images of
pediatric sedation. Pediatr Radiol 2001;31:247–250. children anesthetized with propofol: new fluid-attenuated inversion recov-
12. Hallowell LM, Stewart SE, de Amorim e Silva CT, Ditchfield MR. Reviewing ery finding. Am J Neuroradiol 2001;22:394–399.
the process of preparing children for MRI. Pediatr Radiol 2008;38:271–279. 36. Deliganis A, Fisher D, Lam A, Maravilla K. Cerebrospinal fluid signal inten-
13. American Academy of Pediatrics Committee on Drugs. Guidelines for sity increase on FLAIR MR images in patients under general anesthesia: the
monitoring and management of pediatric patients during and after seda- roles of supplemental O2. Radiology 2001;218:152–156.
tion for diagnostic and therapeutic procedures: addendum. Pediatrics 37. Frigon C, Jardine D, Weinberger E, Heckbert S, Shaw D. Fraction of inspired
2002;110:836–838. oxygen in relation to cerebrospinal fluid hyperintensity on FLAIR MR
14. American Academy of Pediatrics, American Academy of Pediatric Dentistry, imaging of the brain in children and young adults undergoing anesthesia.
Cote CJ, Wilson S, Work Group on Sedation. Guidelines for monitoring and Am J Roentgenol 2002;179:791–796.
management of pediatric patients during and after sedation for diagnostic 38. Braga F, da Rocha A, Hernandez Filho G, Arikawa R, Ribeiro I, Fonseca R.
and therapeutic procedures: an update. Pediatrics 2008;118:2587–2602. Relationship between the concentration of supplemental oxygen and signal
15. American Society of Anesthesiologists TFoSaAbN-A. Practice guide- intensity of CSF depicted by fluid-attenuated inversion recovery imaging.
lines for sedation and analgesia by non-anesthesiologists. Anesthesiology Am J Neuroradiol 2003;24:1863–1868.
2002;96:1004–1017. 39. Inder T, Anderson N, Spencer C, Wells S, Volpe J. White matter injury in
16. Kanal E, Barkovich AJ, Bell C, et al. ACR guidance document for safe MR the premature infant: a comparison between serial cranial sonographic and
practices: 2007. Am J Roentgenol 2007;188(6):1447–1474. MR findings at term. Am J Neuroradiol 2003;24:805–809.
17. Heard CMB, Joshi P, Johnson K. Dexmedetomidine for pediatric MRI seda- 40. Miller S, Cozzio C, Goldstein R, et al. Comparing the diagnosis of
tion: a review of a series of cases. Pediatric Anesthesia 2007;17(9):888–892. white matter injury in premature newborns with serial MR imaging
18. Glasier CM, Stark JE, Brown R, James CA, Allison JW. Rectal thiopental and transfontanel ultrasonography findings. Am J Neuroradiol 2003;24:
sodium for sedation of pediatric patients undergoing MR and other imag- 1661–1669.
ing studies. Am J Neuroradiol 1995;16:111–114. 41. Miller SP, Ferriero DM, Leonard C, et al. Early brain injury in premature
19. Volle E, Park W, Kaufmann HJ. MRI examination and monitoring of pedi- newborns detected with magnetic resonance imaging is associated with
atric patients under sedation. Pediatr Radiol 1996;26:280–281. adverse early neurodevelopmental outcome. J Pediatr 2005;147(5):609.
20. Thompson JR, Schneider S, Ashwal S, Holden BS, Hinshaw DB, Jr, Hasso 42. Woodward LJ, Anderson PJ, Austin NC, Howard K, Inder TE. Neonatal
AN. The choice of sedation for computed tomography in children: a pro- MRI to predict neurodevelopmental outcomes in preterm infants. N Eng J
spective evaluation. Radiology 1982;143:475–479. Med 2006;355:685–694.
21. Strain JD, Campbell JB, Harvey LA, Foley LC. IV nembutal: safe sedation 43. Drobyshevsky A, Bregman J, Storey P, et al. Serial Diffusion Tensor Imag-
for children undergoing CT. Am J Roentgenol 1988;151:975–979. ing Detects White Matter Changes That Correlate with Motor Outcome in
22. Hubbard AM, Markowitz RI, Kimmel B, Kroger M, Bartko MB. Sedation Premature Infants. Dev Neurosci 2007;29:289–301.
for pediatric patients undergoing CT and MRI. J Comput Assist Tomogr 44. Berman JI, Glass HC, Miller SP, et al. Quantitative fiber tracking analysis of
1992;16:3–6. the optic radiation correlated with visual performance in premature new-
23. Holshouser BA, Hinshaw DB, Jr, Shellock FG. Sedation, anesthesia, and borns. Am J Neuroradiol 2009;30:120–124.
physiologic monitoring during MRI. In: Hasso AN, Stark D, eds. Spine and 45. Merchant N, Groves A, Larkman DJ, et al. A patient care system for early 3.0
Body MRI: Categorical course syllabus. Boston: American Roentgen Ray Tesla magnetic resonance imaging of very low birth weight infants. Early
Society; 1991. Hum Dev 2009;85(12):779–783.
24. Keeter S, Benator RM, Weinberg SM, Hartenberg MA. Sedation in pediatric 46. Dumoulin CL, Rohling KW, Piel JE, et al. Magnetic resonance imaging
CT: national survey of current practice. Radiology 1990;175:745–752. compatible neonate incubator. Concept Magn Reson (Magn Reson Engineer-
25. Burckhart GJ, White TJ, III, Siegle RL, Jabour JT, Ramey DR. Rectal thio- ing) 2002;15(2):117–128.
pental versus an intramuscular cocktail for sedating children before com- 47. Bluml S, Friedlich P, Erberich S, Wood JC, Seri I, Nelson MD, Jr. MR
puterized tomography. Am J Hosp Pharm 1980;37:222–224. Imaging of newborns by using an MR-compatible incubator with inte-
26. Bloomfield EL, Masaryk TJ, Caplin A, et al. Intravenous sedation for MR grated radiofrequency coils: initial experience. Radiology 2004;231(2):
imaging of the brain and spine in children: pentoparbital versus Propofol. 594–601.
Radiology 1993;186:93–97. 48. Whitby EH, Griffiths PD, Lonneker-Lammers T, et al. Ultrafast magnetic
27. Berger PE, Kuhn JP, Brusehaber J. Techniques for computed tomography in resonance imaging of the neonate in a magnetic resonance-compatible
infants and children. Radiol Clin North Am 1981;19:399–408. incubator with a built-in coil. Pediatrics 2004;113(2):e150–152.

Barkovich_Chap01.indd 15 5/5/2011 11:43:33 PM


16 Pediatric Neuroimaging

49. Branson HM, Doria AS, Moineddin R, Shroff MM. The brain in children: 73. Holmedal LJ, Friberg EG, Børretzen I, Olerud H, Lægreid L, Rosendahl
is contrast enhancement really needed after obtaining normal unenhanced K. Radiation doses to children with shunt-treated hydrocephalus. Pediatr
CT results? Radiology 2007;244(3):838–844. Radiol 2007;37:1209–1215.
50. Mikkonen R, Kontkanen T, Kivisaari L. Late and acute adverse reactions to 74. Udayasankar UK, Braithwaite K, Arvaniti M, et al. Follow-up evaluation of
iohexol in a pediatric population. Pediatr Radiol 1995;25:350–352. children with ventriculoperitoneal shunt: reduction of radiation and effect
51. Ge HL, Hirsch WL, Wolf GL, Rubin RA, Hackett RK. Diagnostic role of on image quality. Am J Neuroradiol 2008;29:802–806.
gadolinium-DTPA in pediatric neuroradiology: a retrospective review of 75. Flohr TG, Schaller S, Stierstorfer K, Bruder H, Ohnesorge BM, Schoepf UJ.
655 cases. Neuroradiology 1992;34:122–125. Multi-dector row CT systems and image reconstruction techniques. Radi-
52. Saunders D, Thompson C, Gunny R, Jones R, Cox T, Chong W. Magnetic ology 2005;235:756–773.
resonance imaging protocols for paediatric neuroradiology. Pediatr Radiol 76. Barkovich AJ, Kjos BO, Jackson DE, Jr, Norman D. Normal matura-
2007;37(8):789–797. tion of the neonatal and infant brain: MR imaging at 1.5 T. Radiology
53. Cha S, Knopp E, Johnson G, Wetzel S, Litt A, Zagzag D. Intracranial mass 1988;166:173–180.
lesions: dynamic contrast-enhanced susceptibility-weighted echo-planar 77. Holland BA, Haas DK, Norman D, Brant-Zawadzki M, Newton TH. MRI of
perfusion MR imaging. Radiology 2002;223:11–29. normal brain maturation. AJNR 1986;7:201–208.
54. Wintermark P, Moessinger AC, Gudinchet F, Meuli R. Perfusion-weighted 78. Shaw DWW, Weinberger E, Astley SJ, Tsuruda JS. Quantitative compari-
magnetic resonance imaging patterns of hypoxic-ischemic encephalopathy son of conventional spin echo and fast spin echo during brain myelination.
in term neonates. J Magn Reson Imaging 2008;28(4):1019–1025. J Comput Assist Tomogr. 1997;21:867–871.
55. Collidge TA, Thomson PC, Mark PB, et al. Gadolinium-enhanced MR 79. Kjos BO, Umansky R, Barkovich AJ. MR of the brain in children with
imaging and nephrogenic systemic fibrosis: retrospective study of a renal developmental retardation of unknown cause. Am J Neuroradiol 1990;11:
replacement therapy cohort. Radiology 2007:2451070353. 1035–1040.
56. Sadowski EA, Bennett LK, Chan MR, et al. Nephrogenic systemic fibro- 80. Tice HM, Jones KM, Mulkern RV, et al. Fast spin-echo imaging if intracra-
sis: risk factors and incidence estimation. Radiology 2007;243(1): nial neoplasms. J Comput Assist Tomogr 1993;17:425–431.
148–157. 81. McKinstry R, Miller J, Snyder A, et al. A prospective, longitudinal diffu-
57. Penfield JG. Nephrogenic systemic fibrosis and the use of gadolinium- sion tensor imaging study of brain injury in newborns. Neurology 2002;59:
based contrast agents. Pediatr Nephrol 2008;23(12):2121–2129. 824–833.
58. Dharnidharka VR, Wesson SK, Fennell RS. Gadolinium and nephrogenic 82. Sargent MA, Poskitt KJ. Fast fluid-attenuated inversion recovery (FLAIR)
fibrosing dermopathy in pediatric patients. Pediatr Nephrol 2007;22(9): magnetic resonance imaging of the brain: a comparison of multi-shot
1395. echo planar and fast spin-echo techniques. Pediatr Radiol 1997;27:
59. Okuda Y, Sagami F, Tirone P, Morisetti A, Bussi S, Masters RE. [Reproduc- 545–549.
tive and developmental toxicity study of gadobenate dimeglumine formula- 83. Tong KA, Ashwal S, Obenaus A, Nickerson JP, Kido D, Haacke EM. Suscep-
tion (E7155) (3)—Study of embryo-fetal toxicity in rabbits by intravenous tibility-weighted MR imaging: a review of clinical applications in children.
administration]. J Toxicol Sci 1999;24(Suppl 1):79–87. Am J Neuroradiol January 1, 2008 2008;29(1):9–17.
60. Galassi W, Phuttharak W, Hesselink JR, Healy JF, Dietrich RB, Imbesi SG. 84. Tong K, Ashwal S, Holshouser B, et al. Diffuse axonal injury in children:
Intracranial meningeal disease: comparison of contrast-enhanced MR clinical correlation with hemorrhagic lesions. Ann Neurol 2004;56:36–50.
imaging with fluid-attenuated inversion recovery and fat-suppressed T1 85. Tong K, Ashwal S, Holshouser B, et al. Hemorrhagic shearing lesions in chil-
weighted sequences. Am J Neuroradiol 2005;26:553–559. dren and adolescents with posttraumatic diffuse axonal injury: improved
61. Leijser LM, Srinivasan L, Rutherford MA, Counsell SJ, Allsop JM, Cowan F. detection and initial results. Radiology 2003;227:332–339.
Structural linear measurements in the newborn brain: accuracy of cranial 86. Sehgal V, Delproposto Z, Haacke EM, et al. Clinical applications of neu-
ultrasound compared to MRI. Pediatr Radiol 2007;37:640–648. roimaging with susceptibility-weighted imaging. J Magn Reson Imaging
62. Hall E. Lessons we have learned from our children: cancer risks from diag- 2005;22(4):439–450.
nostic radiology. Pediatr Radiol 2002;32:700–706. 87. Ito R, Mori S, Melhem E. Diffusion tensor brain imaging and tractography.
63. National Academy of Sciences. Health risks from exposure to low lwvels of Neuroimaging Clin N Am 2002;12:1–19.
inonizing radiation: BEIR VII. Washington, DC: National Academies Press; 88. Mori S, van Zijl PC. Fiber tracking: principles and strategies - a technical
2005. review. NMR Biomed Nov-Dec 2002;15(7–8):468–480.
64. Berrington de Gonzalez A, Mahesh M, Kim KP, et al. Projected cancer risks 89. Rohrschneider WK, Forsting M, Darge K, Tröger J. Diagnostic value of spi-
from computed tomographic scans performed in the United States in 2007. nal US: comparative study with MR imaging in pediatric patients. Radiol-
Arch Intern Med 2009;169(22):2071–2077. ogy 1996;200:383–388.
65. Hall P, Adami HO, Trichopoulos D, et al. Effect of low doses of ionising 90. Weinberger E, Murakami J, Shaw D, White K, Radvilas M, Yean C. Three-
radiation in infancy on cognitive function in adulthood: Swedish popula- dimensional fast spin echo T1 weighted imaging of the pediatric spine.
tion based cohort study. BMJ 2004;328(7430):19. J Comput Assist Tomogr 1995;19:721–725.
66. King MA, Kanal KM, Relyea-Chew A, Bittles M, Vavilala MS, Hollingworth 91. Weiss KL, Sun D, Weiss JL. Pediatric MR imaging with automated
W. Radiation exposure from pediatric head CT: a bi-institutional study. spine survey iterative scan technique (ASSIST). Am J Neuroradiol Apr
Pediatr Radiol 2009;39(10):1059–1065. 2009;30(4):821–824.
67. Robertson R, Robson C, Zurakowski D, Antiles S, Strauss K, Mulkern R. 92. Chen SC, Simon EM, Haselgrove JC, et al. Fetal posterior fossa volume:
CT versus MR in neonatal brain imaging at term. Pediatr Radiol 2003;33: assessment with MR imaging. Radiology March 1, 2006 2006;238(3):
442–449. 997–1003.
68. Smith AB, Dillon WP, Gould R, Wintermark M. Radiation Dose-Re- 93. Glenn OA, Barkovich AJ. Magnetic resonance imaging of the fetal brain
duction Strategies for Neuroradiology CT Protocols. Am J Neuroradiol and spine: an increasingly important tool in prenatal diagnosis, Part 1. Am
2007;28(9):1628–1632. J Neuroradiol September 1, 2006 2006;27(8):1604–1611.
69. Smith AB, Dillon WP, Lau BC, et al. Radiation dose reduction strategy for 94. Glenn OA, Barkovich J. Magnetic resonance imaging of the fetal brain and
CT protocols: successful implementation in neuroradiology section. Radi- spine: an increasingly important tool in prenatal diagnosis: Part 2. Am J
ology 2008;247:499–506. Neuroradiol October 1, 2006 2006;27(9):1807–1814.
70. Frush D. Pediatric CT: practical approach to diminish the radiation dose. 95. Levine D. Obstetric MRI. J Magn Reson Imaging 2006;24(1):1–15.
Pediatr Radiol 2002;32:714–717. 96. Tang PH, Bartha AI, Norton ME, Barkovich AJ, Sherr EH, Glenn OA. Agene-
71. Huda W. Dose and image quality in CT. Pediatr Radiol 2002;32:709–713. sis of the corpus callosum: an MR imaging analysis of associated abnormal-
72. Singh S, Kalra MK, Moore MA, et al. Dose reduction and compliance with ities in the fetus. Am J Neuroradiol February 1, 2009 2009;30(2):257–263.
pediatric CT protocols adapted to patient size, clinical indication, and 97. Busse RF, Riederer SJ, Fletcher JG, Bharucha AE, Brandt KR. Interactive fast
number of prior studies. Radiology 2009;252(1):200–208. spin-echo imaging. Magn Reson Med 2000;44(3):339–348.

Barkovich_Chap01.indd 16 5/5/2011 11:43:33 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 17

98. Schneider MM, Berman JI, Baumer FM, et al. Normative apparent 123. Wolff SD, Balaban RS. Magnetization transfer contrast (MTC) and tissue
diffusion coefficient values in the developing fetal brain. Am J Neuroradiol water proton relaxation in vivo. Magn Reson Med 1989;11:135–144.
2009;30(9):1799–1803. 124. Wolff SD, Eng J, Balaban RS. Magnetization transfer contrast: method
99. Glenn O. MR imaging of the fetal brain. Pediatr Radiol 2010;40(1):68–81. for improving contrast in gradient-recalled-echo images. Radiology
100. Kasprian GJ, Brugger PC, Lindner C, Stuhr F, Prayer D. Fetal motor and 1991;179:133–137.
sensory tractis visualized by in vivo diffusion tensor imaging. Paper pre- 125. Eng J, Ceckler TL, Balaban RS. Quantitative 1 H magnetization transfer
sented at: American Society of Neuroradiology; 2007; Chicago. imaging in Vivo. Mag Res Med 1991;17:304–314.
101. Edelman RR, Mattle HP, Atkinson DJ, Hoogewoud HM. MR angiography. 126. Eastwood JD, Lev MH, Azhari T, et al. CT perfusion scanning with decon-
Am J Roentgenol 1990;154:937–946. volution analysis: pilot study in patients with acute middle cerebral artery
102. Ruggieri PM, Gerhard AL, Masaryk TJ, Modic MT. Intracranial circula- stroke. Radiology 2002;222(1):227–236.
tion: pulse-sequence considerations in three-dimensional (volume) MR 127. Hoeffner EG, Case I, Jain R, et al. Cerebral perfusion CT: technique and
angiography. Radiology 1989;171:785–791. clinical applications. Radiology 2004;231:632–644.
103. Laub GA, Kaiser WA. MR angiography with gradient motion refocusing. J 128. Wintermark M, Lepori D, Cotting J, et al. Brain perfusion in children: evo-
Comp Assist Tomogr 1989;12:377–382. lution with age assessed by quantitative perfusion computed tomography.
104. Anderson CM, Saloner D, Tsuruda JS, Shapeero LG, Lee RE. Artifacts in Pediatrics 2004;113(6):1642–1652.
maximum-intensity-projection display of MR angiograms. Am J Roent- 129. Wintermark M, Cotting J, Roulet E, et al. Acute brain perfusion disorders
genol 1990;154:623–629. in children assessed by quantitative perfusion computed tomography in
105. Tsuruda JS, Saloner D, Norman D. Artifacts associated with MR neuroan- the emergency setting. Pediatr Emerg Care 2005;21(3):149–160.
giography. Am J Neuroradiol 1992;13:1411–1422. 130. Detre JA, Leigh JS, Williams DS, Koretsky AP. Perfusion imaging. Magn
106. Heiserman JE, Drayer BP, Fram EK, et al. Carotid artery stenosis: clini- Reson Med 1992;23:37–45.
cal efficacy of two-dimensional time-of-flight angiography. Radiology 131. Zigun JR, Frank JA, Barrios FA, et al. Measurement of brain activity with
1992;182:761–768. bolus administration of contrast agent and gradient-echo MR imaging.
107. Keller PJ, Drayer BP, Fram EK, Williams KD, Dumoulin CL, Souza SP. MR Radiology 1993;186:353–356.
angiography with two-dimensional acquisition and three-dimensional 132. Zhang W, Williams DS, Detre JA, Koretsky AP. Measurement of brain
display. Radiology 1989;173:527–532. perfusion by volume localized nuclear magnetic resonance spectroscopy
108. Willig DS, Turski PA, Frayne R, et al. Contrast-enhanced 3D MR DSA using inversion of arterial water spins: accounting for transit time and
of the carotid artery bifurcation: preliminary study of comparison with cross relaxation. Magn Reson Med 1992;25:362–371.
unenhanced 2D and 3D time-of-flight MR angiography. Radiology 133. Warach S, Li W, Ronthal M, Edelman RR. Acute cerebral ischemia: evalua-
1998;208:447–451. tion with dynamic contrast-enhanced MR imaging and MR angiography.
109. Korosec FR, Frayne R, Grist TM, Mistretta CA. Time-resolved contrast- Radiology 1992;182:41–47.
enhanced 3D MR angiography. Magn Reson Med 1996;36(3): 134. Villringer A, Rosen RB, Belliveau JW, et al. Dynamic imaging with lan-
345–351. thanide chelates in normal brain: contrast due to magnetic susceptibility
110. Huston J, III, Rufenacht DA, Ehman RL, Wiebers DO. Intracranial aneu- effects. Mag Res Med 1988;6:164–174.
rysms and vascular malformations: comparison of time-of-flight and 135. Barajas Jr RF, Chang JS, Segal MR, et al. Differentiation of recurrent glio-
phase-contrast MR angiography. Radiology 1991;181:721–730. blastoma multiforme from radiation necrosis after external beam radia-
111. Mattle HP, Wentz KU, Edelman RR, et al. Cerebral venography with MR. tion therapy with dynamic susceptibility-weighted contrast-enhanced
Radiology 1991;178:453–458. perfusion MR imaging. Radiology 2009;253:486–496.
112. Widjaja E, Shroff M, Blaser S, Laughlin S, Raybaud C. 2D time-of-flight 136. Caseiras GB, Chheang S, Babb J, et al. Relative cerebral blood volume
MR venography in neonates: anatomy and pitfalls. Am J Neuroradiol measurements of low-grade gliomas predict patient outcome in a multi-
2006;27:1913–1918. institution setting. European Journal of Radiology 2010;73(2):215–220.
113. Rollins N, Ison C, Reyes T, Chia J. Cerebral MR venography in children: 137. Laswad T, Wintermark P, Alamo L, Moessinger A, Meuli R, Gudinchet F.
comparison of 2D time-of-flight and gadolinium-enhanced 3D gradient- Method for performing cerebral perfusion-weighted MRI in neonates.
echo techniques. Radiology 2005;235:1011–1017. Pediatr Radiol 2009;39(3):260–264.
114. Meckel S, Reisinger C, Bremerich J, et al. Cerebral venous thrombosis: 138. Williams DS, Detre JA, Leigh JS, Koretsky AP. Magnetic resonance imaging
diagnostic accuracy of combined, dynamic, and static contrast-enhanced of perfusion using spin inversion of arterial water. Proc Natl Acad Sci USA
4D MR venography. Am J Neuroradiol 2010;31:527–525. 1992;89:212–216.
115. Nitz WR, Bradley WG, Jr, Watanabe AS, et al. Flow dynamics of cere- 139. Williams DS, Grandis DJ, Zhang W, Koretsky AP. Magnetic resonance
brospinal fluid: assessment with phase-contrast velocity MR imag- imaging of perfusion in the isolated rat heart using spin inversion of arte-
ing performed with retrospective cardiac gating. Radiology 1992;183: rial water. Mag Reson Med 1993;30:361–365.
395–405. 140. Roberts DA, Detre JA, Bolinger L, Insko EK, Leigh JS. Quantitative mag-
116. Enzmann DR, Pelc NJ. Normal flow patterns of intracranial and spinal netic resonance imaging of human brain perfusion at 1.5 T using steady
cerebrospinal fluid defined with phase-contrast cine MR imaging. Radiol- state inversion of arterial water. Proc Nat Acad Sci 1994;91:33–37.
ogy 1991;178:467–474. 141. Ye FQ, Pekar JJ, Jezzard P, Duyn J, Frank JA, McLaughlin AC. Perfusion
117. Pelc LR, Pelc N, Rayhill S. Arterial and venous blood flow: noninvasive imaging of the human brain at 1.5 T using a single shot EPI spin tagging
quantitation with MR imaging. Radiology 1992;185:809–812. approach. Mag Reson Med 1996;36:219–224.
118. Reeder SB, Hargreaves BA, Yu H, Brittain JH. Homodyne recon- 142. Petersen ET, Lim T, Golay X. Model-free arterial spin labeling quantifica-
struction and IDEAL water-fat decomposition. Magn Reson Med Sep tion approach for perfusion MRI. Magn Reson Med 2006;55:219–232.
2005;54(3):586–593. 143. Hendrikse J, Petersen ET, van Larr PJ, Golay X. Cerebral border zones
119. Filler AG, Howe FA, Hayes CE, et al. Magnetic resonance neurography. between distal end branches of intracranial arteries: MR imaging. Radiol-
Lancet 1993;341:659–661. ogy 2008;246(2):572–580.
120. Howe FA, Filler AG, Bell BA, et al. Magnetic resonance neurography. Magn 144. Alsop DC, Detre JA. Multisection cerebral blood flow MR imaging with
Reson Med 1992;28:328–338. continuous arterial spin labeling. Radiology 1998;208:410–416.
121. Chew WM, Rowley HA, Barkovich AJ. Magnetization transfer contrast 145. Cox IH, Roberts TPL, Moseley ME. Principles and techniques in neuroim-
imaging in pediatric patients. Radiology 1992;185(P):281. aging. In: Kucharczyk J, Moseley ME, Barkovich AJ, eds. Magnetic Reso-
122. Dousset V, Grossman RI, Ramer KN, et al. Experimental allergic encepha- nance Neuroimaging. Boca Ratan: CRC; 1994:1–102.
lomyelitis and multiple sclerosis: lewion characterization with magnetiza- 146. Melhem ER, Itoh R, Jones L, Barker PB. Diffusion tensor MR imaging of
tion transfer imaging. Radiology 1992;182:483–491. the brain: effect of diffusion weighting on trace and anisotropy measure-
ments. Am J Neuroradiol 2000;21:1813–1820.

Barkovich_Chap01.indd 17 5/5/2011 11:43:33 PM


18 Pediatric Neuroimaging

147. Sasaki M, Yamada K, Watanabe Y, et al. Variability in absolute apparent callosum: a 3T diffusion tensor imaging and Q-ball tractography study.
diffusion coefficient values across different platforms may be substantial: Am J Neuroradiol 2009;30:282–289.
a multivendor, multi-institutional comparison study. Radiology 2008; 170. Widjaja E, Blaser S, Raybaud C. Diffusion tensor imaging of midline pos-
249(2):624–630. terior fossa malformations. Pediatr Radiol 2006;36(6):510–517.
148. Basser P, Matiello J, Le Bihan D. MR diffusion tensor spectroscopy and 171. Maas L, Mukherjee P, Carballido-Gamio J, et al. Early laminar organization
imaging. Biophys J 1994;66:259–267. of the human cerebrum demonstrated with diffusion tensor imaging in
149. Basser P, Pierpaoli C. Microstructural and physiological features of tis- extremely premature infants. Neuroimage 2004;22:1134–1140.
sues elucidated by quantitative-diffusion-tensor MRI. J Magn Reson 172. Filippi C, Lin D, Tsiouris A, et al. Diffusion-tensor MR imaging in children
B 1996;111:209–219. with developmental delay: preliminary findings. Radiology 2003;229:44–50.
150. Le Bihan D, Mangin J, Poupon C, et al. Diffusion tensor imaging: concepts 173. Barkovich A, Baranski K, Vigneron D, et al. Proton MR spectroscopy for
and applications. J Magn Reson Imaging 2001;13:534–546. the evaluation of brain injury in asphyxiated, term neonates. Am J Neuro-
151. Partridge SC, Mukherjee P, Henry R, et al. Diffusion tensor imaging: serial radiol 1999;20:1399–1405.
quantitation of white matter tract maturity in premature newborns. Neu- 174. Hanrahan J, Cox I, Azzopardi D, et al. Relation between proton magnetic
roimage 2004;22:1302–1314. resonance spectroscopy within 18 hours of birth asphyxia and neurode-
152. Hasan KM, Alexander AL, Narayana PA. Does fractional anisotropy have velopment at 1 year of age. Dev Med Child Neurol 1999;41:76–82.
better noise immunity characteristics than relative anisotrophy in diffu- 175. Grodd W, Krageloh-Mann I, Klose U, Sauter R. Metabolic and destructive
sion tensor MRI? Magn Reson Med 2004;51:413–417. brain disorders in children: findings with localized proton MR spectros-
153. Mukherjee P, Berman JI, Chung SW, Hess CP, Henry RG. Diffusion tensor copy. Radiology 1991;181:173–181.
MR imaging and fiber tractography: theoretic underpinnings. Am J Neu- 176. Degrauw T, Salomons G, Cecil K, et al. Congenital creatine transporter
roradiol 2008;29(4):632–641. deficiency. Neuropediatrics 2002;33:232–238.
154. Mukherjee P, Chung SW, Berman JI, Hess CP, Henry RG. Diffusion tensor 177. Filippi C, Ulug A, Deck M, Zimmerman R, Heier L. Developmental delay
MR imaging and fiber tractography: technical considerations. Am J Neu- in children: assessment with proton MR spectroscopy. Am J Neuroradiol
roradiol 2008;29(5):843–852. 2002;23:882–888.
155. Mori S, Crain BJ, Chacko VP, van Zijl PCM. Three dimensional tracking 178. Tzika A, Vajapeyam S, Barnes P. Multivoxel proton MR spectroscopy
of axonal projections in the brain by magnetic resonance imaging. Ann and hemodynamic MR imaging of childhood brain tumors: preliminary
Neurol 1999;45:265–269. observations. Am J Neuroradiol 1997;18:203–218.
156. Neil J, Miller J, Mukherjee P, Huppi PS. Diffusion tensor imaging of 179. Tzika A. Proton magnetic resonance spectroscopic imaging as a cancer
normal and injured developing human brain - a technical review. NMR biomarker for pediatric brain tumors. Int J Oncol 2008;32:517–526.
Biomed 2002;15(7–8):543–552. 180. Kimmich R, Hoepfel D. Volume-selective multipulse spin-echo spectros-
157. Horsfield MA, Jones DK. Applications of diffusion-weighted and diffu- copy. J Magn Reson 1987;72:379–384.
sion tensor MRI to white matter diseases - a review. NMR Biomed 2002; 181. Barkovich AJ, Miller SP, Bartha A, et al. MRI, MRS, and DTI of sequen-
15(7–8):570–577. tial studies in neonates with encephalopathy. Am J Neuroradiol 2006;27:
158. Beaulieu C. The basis of anisotropic water diffusion in the nervous system - 533–547.
a technical review. NMR Biomed 2002;15(7–8):435–455. 182. Pirzkall A, Nelson S, McKnight T, et al. Metabolic imaging of low-grade
159. Sotak CH. The role of diffusion tensor imaging in the evaluation of gliomas with three-dimensional magnetic resonance spectroscopy. Int J
ischemic brain injury - a review. NMR Biomed 2002;15(7–8):561–569. Radiat Oncol Biol Phys 2002;53:1254–1264.
160. Moseley M, Kucharczyk J, Mintorovitch J, et al. Diffusion-weighted MR 183. Vigneron DB. Magnetic resonance spectroscopic imaging of human brain
imaging by susceptibility contrast NMR. Am J Neuroradiol 1990;11: development. Neuroimaging Clin N Am 2006;16:75–85.
423–429. 184. Vigneron DB, Barkovich AJ, Noworolski SM, et al. Three-Dimensional
161. McKinstry R, Mathur A, Miller J, et al. Radial organization of developing Proton MR Spectroscopic Imaging of Premature and Term Neonates. Am
preterm human cerebral cortex revealed by non-invasive water diffusion J Neuroradiol 2001;22:1424–1433.
anisotropy MRI. Cereb Cortex 2002;12:1237–1243. 185. Kim DH, Adalsteinsson E, Spielman DM. Spiral readout gradients for the
162. Nomura Y, Sakuma H, Takeda K, Tagami T, Okuda Y, Nakagawa T. Dif- reduction of motion artifacts in chemical shift imaging. Magn Reson Med
fusional anisotropy of the human brain assessed with diffusion-weighted Mar 2004;51(3):458–463.
MR: relation with normal brain development and aging. Am J Neuroradiol 186. Kim DH, Barkovich AJ, Vigneron D. Short echo time MR spectroscopic imag-
1994;15:231–238. ing for neonatal pediatric imaging. Am J Neuroradiol 2006;27:1370–1372.
163. Wimberger DM, Roberts TP, Barkovich AJ, Prayer LM, Moseley ME, 187. Nordahl CW, Dierker D, Mostafavi I, et al. Cortical folding abnormali-
Kucharczyk J. Identification of “premyelination” by diffusion-weighted ties in autism revealed by surface-based morphometry. J Neurosci Oct 24
MRI. J Comput Assist Tomogr 1995;19:28–33. 2007;27(43):11725–11735.
164. Neill JJ, Shiran SI, McKinstry RC, et al. Normal brain in human newborns: 188. Cachia A, Paillere-Martinot ML, Galinowski A, et al. Cortical folding
apparent diffusion coefficient and diffusion anisotropy measured by using abnormalities in schizophrenia patients with resistant auditory hallucina-
diffusion tensor MR imaging. Radiology 1998;209:57–66. tions. Neuroimage Feb 1 2008;39(3):927–935.
165. Mukherjee P, Miller J, Shimony J, et al. Normal brain maturation during 189. Dubois J, Benders M, Borradori-Tolsa C, et al. Primary cortical folding
childhood: developmental trends characterized with diffusion-tensor MR in the human newborn: an early marker of later functional development.
imaging. Radiology 2001;221:349–358. Brain Aug 2008;131(Pt 8):2028–2041.
166. Hoon Jr A, Lawrie Jr W, Melhem E, et al. Diffusion tensor imaging of 190. Dubois J, Benders M, Cachia A, et al. Mapping the early cortical folding pro-
periventricular leukomalacia shows affected sensory cortex white matter cess in the preterm newborn brain. Cereb Cortex Jun 2008;18(6):1444–1454.
pathways. Neurology 2002;59:752–756. 191. Gilmore JH, Lin W, Prastawa MW, et al. Regional gray matter growth, sex-
167. Vachha B, Adams RC, Rollins NK. Limbic tract anomalies in pediatric ual dimorphism, and cerebral asymmetry in the neonatal brain. J Neurosci
myelomeningocele and Chiari II malformation: anatomic correlations with Feb 7 2007;27(6):1255–1260.
memory and learning – initial investigation. Radiology 2006;240:194–202. 192. Habas PA, Kim K, Rousseau F, Glenn OA, Barkovich AJ, Studholme
168. Nakata Y, Barkovich AJ, Wahl M, et al. Diffusion Abnormalities and C. Atlas-based segmentation of developing tissues in the human
Reduced Volume of the Ventral Cingulum Bundle in Agenesis of the Cor- brain with quantitative validation in young fetuses. Hum Brain Mapp
pus Callosum: A 3 T Imaging Study. Am J Neuroradiol 2009;30(6):1142– 2010.
1148. 193. Ogawa S, Lee TM, Nayak AS, Glynn P. Oxygen sensitive contrast in mag-
169. Wahl M, Strominger Z, Jeremy RJ, et al. Variability of homotopic and netic resonance imaging of rodent brain at high magnetic fields. Magn
heterotopic callosal connectivity in partial agenesis of the corpus Reson Med 1990;14:68–78.

Barkovich_Chap01.indd 18 5/5/2011 11:43:33 PM


Chapter 1 • Techniques and Methods in Pediatric Neuroimaging 19

194. Engel SA, Rumelhart DE, Wandell BA, et al. fMRI of human visual cortex. 198. Eden GF, Zeffiro TA. PET and fMRI in the detection of task-related brain
Nature 1994;369:525. activity: implications for the study of brain development. In: Thatcher
195. Jack C, Thompson R, Butts K, et al. Sensory motor cortex: correlation of RW, Lyon GR, Rumsey J, Krasnegor N, eds. Developmental Neuroimaging.
presurgical mapping with functional MRI and invasive cortical mapping. San Diego: Academic; 1996:77–90.
Radiology 1994;190:85–92. 199. Martin E, Marcar V. Functional MR imaging in pediatrics. Magn Reson
196. McCarthy G, Blamire AM, Rothman DL. Echo planar MRI studies of Imaging Clin N Am 2001;9(1):231–246.
frontal cortex activation during word generation in humans. Proc Natl 200. DeLano MC, Cooper TG, Siebert JE, Potchen MJ, Kuppasamy K.
Acad Sci 1993;90:4952–4956. High-b-value diffusion-weighted MR imaging of adult brain: image con-
197. Hertz-Pannier L, Gaillard W, Mott S, et al. Noninvasive assessment of trast and apparent diffusion coefficient map features. Am J Neuroradiol
language dominance in children and adolescents with functional MRI: 2000;21:1830–36.
a preliminary study. Neurology 1997;48:1003–1012.

Barkovich_Chap01.indd 19 5/5/2011 11:43:33 PM


CHAPTER

Normal Development of the


2 Neonatal and Infant Brain,
Skull, and Spine
A. JAMES BARKOVICH AND PRATIK MUKHERJEE

Introduction Normal development of the skull and paranasal sinuses


Normal prenatal brain development The skull
Embryology The cranial fontanelles and sutures
Imaging of the preterm brain (premature infant and Maturation of the paranasal sinuses
fetus) Normal development of brain iron
Grading of brain maturation in premature infants Normal development of the spine
Normal postnatal brain development Embryology
Embryology CT of spine development
Spin-echo MRI of postnatal brain development MRI of spine development
Terminal zones Evaluation of brain development using physiologic
Milestones MR techniques
Postulated causes of T1 and T2 shortening associated with MR spectroscopy
myelination Diffusion tensor imaging and tractography
Other approaches to brain maturation by MR Magnetization transfer imaging
Normal postnatal MR development of the corpus callosum Perfusion imaging
Normal MR development of the pituitary gland Functional MRI

from the activity through the use of blood oxidation–level dependent


INTRODUCTION (BOLD) imaging. Imaging correlations of these changes that occur
The brain matures in an organized, predetermined pattern that during normal brain maturation are described in this chapter.
correlates with the functions the newborn or infant performs at various
stages of development. Prior to the development of modern neuroim-
aging techniques, it was not possible to analyze normal brain matura- NORMAL PRENATAL BRAIN
tion in vivo. Neuroimaging allows analysis of many aspects of brain DEVELOPMENT
maturation, including development of sulci, myelination, maturation
of brain chemistry, changes in free water diffusion, changes in blood
Embryology
velocity, and changes in location of specific brain activities. Although The bilateral cerebral vesicles that will form the cerebral hemispheres
transfontanelle ultrasonography, x-ray computed tomography (CT) first appear at about 35 days of gestation as outpouchings of the tel-
and magnetic resonance imaging (MRI) all show gross morphologic encephalon from the regions of the foramen of Monro. At the time of
changes in the maturing brain, MRI supplies the most information. these outpouchings, the walls of the vesicles are uniformly thin and
MRI permits highly sensitive assessment of the maturation of gray and are connected in the midline by the lamina terminalis, a midline area
white matter, in addition to assessment of microstructural changes derived from the roof plate that has shrunken due to apoptosis. The
including those secondary to myelination. Myelination is an important lamina terminalis does not grow as development proceeds; however, the
component of brain maturation because it facilitates the transmission cerebral vesicles exhibit marked expansion laterally, rostrally, ventrally,
of neural impulses through the central nervous system (CNS); myeli- and caudally. As the vesicles expand, cellular layers develop within their
nation can be studied by the changes in the T1 and T2 relaxation times walls, forming the germinal matrices from which the cells that form the
of the brain tissue, by assessing changes in magnetization transfer or, cerebrum will eventually develop. The germinal matrices are initially
indirectly, by assessing changes in the degree and direction of micro- composed only of a single region of proliferating cells (the ventricular
scopic motion (diffusion) of water in the brain. Magnetic resonance zone), but as development proceeds, a more peripheral subventricu-
spectroscopy allows us to assess some of the chemical changes that lar germinal zone develops, separated from the ventricular zone by a
occur as the brain develops. Finally, changes in regions of brain activity periventricular fiber-rich zone. These germinal zones are divided based
(sometimes called functional MRI or fMRI) may be determined upon their location, the type of neurons generated, and the ultimate
by looking at changes in local cerebral blood oxygenation resulting destination of the neurons: (a) medial ganglionic eminence (located

20

Barkovich_Chap02.indd 20 5/7/2011 2:41:49 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 21

near the developing third ventricle, GABAergic neurons generated,


including cortical interneurons, hippocampal interneurons, and glo-
bus pallidus interneurons), (b) lateral ganglionic eminence (located
near the developing third ventricle, GABAergic neurons generated,
mainly striatal projection neurons and olfactory interneurons, also
some thalamic interneurons), (c) preoptic area (located near the bot-
tom of the developing third ventricle, GABAergic neurons generated,
including cells to the preoptic area, amygdala, posterior globus palli-
dus, and cortex), and (d) dorsal neocortical germinal zone (located in
walls of developing lateral ventricles, glutamatergic neurons destined
for the neocortex) (1–7). Neurons migrate from these germinal zones
through the developing hemispheres to form the cerebral cortex, ini-
tially in an incomplete form that is often called the preplate. As the
neurons migrate, they develop axonal connections with other cortical
and subcortical neurons. These connections form a prospective zone
of white matter that is called the intermediate zone because it is in the
region intermediate between the ventricular zone and the develop-
ing cortex. In addition to afferent and efferent axons, the intermedi-
ate zone contains migrating neurons and oligodendrocyte progenitor
cells (8). Between the preplate and the intermediate zone is a transient
area of loosely packed and loosely organized neurons that form tem-
porary neuronal circuits, particularly with the thalamus (9); this zone
is known as the subplate. The subplate is largest at the 22nd gestational FIG. 2-1. Schematic demonstrating normal development of the fetal
week (10). At that time, it is about four times thicker than the cortex brain. During the early weeks of gestation, the cerebral hemispheres are
and is easily seen on fetal MRI as an area of T1 hypointensity and T2 smooth. The earliest fetal sulcus is the sylvian fissure, which first appears
hyperintensity between the intermediate zone and cortex (10–12). It during the fifth gestational month. By about 27 weeks, the Rolandic, inter-
parietal, and superior temporal sulci have appeared. Secondary and tertiary
gradually disappears after the 30th gestational week, as definitive corti-
sulci develop during the last 2 months of gestation. Because of the differ-
cal connections are established (10). Details of the development of the ent appearance of the brain in premature infants as compared with term
hemispheres are described in more detail in Chapter 5. For the pur- infants, it is important to know the gestational age of a child at the time of
poses of the discussion to follow, it is sufficient to understand that the delivery before assessing the structure of the brain.
occipital pole begins to develop at about the 43rd gestational day and
the temporal pole at approximately the 50th gestational day. During
the early weeks of gestation, the surfaces of the cerebral hemispheres proceeds most rapidly in the area of the sensorimotor and visual
are smooth. The fetal sulci appear in an orderly sequence; the phyloge- pathways. These are also the areas in which myelination occurs earliest
netically older sulci appear first, and the more recently acquired sulci (22 and following section), in which glucose uptake increases earliest
appear later. The principal sulci and gyri form the characteristic pat- (23), in which relative cerebral perfusion increases earliest (24), in
tern of the human cortex that can be identified in the full-term infant which cortical microstructure matures most rapidly (25), and in which
(Table 2-1). The primitive Sylvian fissure, the earliest fetal sulcus, is brain chemistry matures most rapidly (26). Gyral development takes
usually present when the fetus is imaged in the fourth gestational place most slowly in the frontobasal, frontopolar, and anterior tempo-
month. The next sulci to appear are the calcarine, parietooccipital, and ral regions, which are the slowest regions to myelinate and to mature
cingulate sulci during the fifth month (by 20–22 weeks); the Rolandic metabolically (23,24,26).
(central), interparietal, and superior temporal sulci that appear toward
the end of the sixth month (by 25 weeks); and the precentral, postcen- Imaging of the Preterm Brain (Premature Infant
tral, superior frontal, and middle temporal sulci that appear during the
and Fetus)
seventh gestational month (24–28 weeks) (13) (Fig. 2-1 and Table 2-1).
Because sulcal formation occurs so late in gestation, imaging studies of As mentioned earlier, the imaging modality being used determines
premature infants show sulci that are shallow and few in numbers. It which features of brain development can be evaluated. If the anterior
is, therefore, important to know the postconceptional age of a child before fontanelle is large enough, ultrasound shows development of the gyri
assessing the sulcal pattern. Otherwise, a false diagnosis of lissencephaly and sulci nearly as well as CT and MRI but does not give information
may be made. In addition, as will be discussed in more detail later in about brain myelination. CT allows fairly good information about sul-
the chapter, sulcation, myelination, and corpus callosum development cal development but gives a poor assessment of myelin development.
tend to be delayed in prematurely born neonates compared to fetuses of MRI allows excellent assessment of myelination, sulcation, and chemi-
the same postconceptual age. cal maturation. As CT requires the use of ionizing radiation and does
Van der Knaap et al. (19) have devised a method by which gyral not provide any information that cannot be obtained by ultrasound and
development can be divided into five stages: (a) before 32 weeks, (b) 33 MR, it is not recommended for brain imaging in the fetus or the prema-
to 34 weeks, (c) 35 to 37 weeks, (d) 38 to 41 weeks, and (e) beyond ture neonate. Because we no longer use CT for premature neonates and
41 weeks. The gyral maturity is determined by measurements of the because the changes visible on CT (restricted to progressively increas-
width of the gyri and depth of the sulci. The stage of gyral develop- ing sulcation) are seen better on MR, this edition does not show CT
ment is then assigned based upon the degree of gyral maturity in seven images of preterm infants, instead concentrating on the more extensive
different regions of the brain. Battin et al. (20) and Ruoss et al. (21) changes detected with maturation by MR.
have also developed systems of assessing sulcal development based on Fetal MRI has been successfully utilized in a large number of
ratios of the width and depth of the sulci and gyri. Gyral development centers for the past decade (11,12,27–33). Improvements in coil

Barkovich_Chap02.indd 21 5/7/2011 2:41:49 AM


22 Pediatric Neuroimaging

and ventricular volume parallel that of supratentorial volume, while


TABLE 2-1 Chronology of Sulcation supratentorial white matter volume grows more rapidly than overall
supratentorial volume (Dr. Piotr Habas, personal communication).
of the Brain Prior to 24 weeks of gestation, the brain is essentially agyric with
the exception of the wide, vertically oriented Sylvian fissures. The size
Location Age of Sulcation of the brain is very small, and the cerebral cortex is extremely thin,
Medial Surface so thin imaging sections (≤3 mm) must be used for optimal evalua-
tion. MR images show the cortex to be very hyperintense with respect
Callosal sulcus 14–23 wk to the underlying white matter on T1-weighted images and very
Parietooccipital sulcus 16–23 wk hypointense compared to white matter on T2-weighted images (Figs.
2-2 and 2-3). The germinal matrix has not yet involuted and is seen
Calcarine sulcus 16–25 wk
as a stripe in the walls of the lateral ventricles that is isointense to the
Cingulate sulcus 18–24 wk gray matter of the cortex on T1- and T2-weighted images (Fig. 2-2);
Marginal sulcus 22–27 wk it is thicker in younger fetuses (18–20 weeks) and becomes thinner
and discontinuous with further maturation (Fig. 2-3). The germinal
Secondary cingulate sulci 32–33 wk
matrix is thickest at the region of the caudate heads and should not
Secondary occipital sulci 34 wk be mistaken for a germinal matrix hemorrhage at this location. The
Ventral Surface more central ventricular zone and adjacent subventricular zone can-
not be distinguished by in vivo imaging. Between the germinal zones
Hippocampal sulcus 16–23 wk and the cortex, MRI also shows a layer of intermediate signal inten-
Collateral sulcus 23–26 wk sity, separated from the more peripheral cerebral cortex and more
Occipitotemporal sulcus 30–33 wk central germinal matrix (Fig. 2-2C–E). Although previously believed
to represent migrating glial cells, this layer has recently been identi-
Lateral Surface fied as the intermediate zone, or the developing fetal white matter
Superior frontal sulcus 25–29 wk (10). Projection and commissural axons are present and glial cells are
actively migrating through this region (34). Immediately peripheral
Inferior frontal sulcus 28–29 wk
to the intermediate zone and deep to the cerebral cortex is a region of
Superior temporal sulcus 23–27 wk T1 hypointensity/T2 hyperintensity (Fig. 2-2E), which is likely to be
(posterior part) the subplate, the region where thalamocortical afferent axons accu-
Superior temporal sulcus 28–32 wk mulate and wait before entering the cortical plate to establish defini-
(anterior part) tive synapses (10,35,36). These zones are most prominent in younger
fetuses (18–20 weeks); they can still be seen but are less conspicuous at
Inferior temporal sulcus 30–33 wk
23 to 24 weeks (12,37). The lateral ventricles and the cisterns around
Interparietal sulcus 26–28 wk the brainstem and cerebellum are visible and more prominent at this
Insular sulci 34 wk age than in the mature infant; they are smaller by 23 to 24 weeks
(Fig. 2-3) than at 18 to 20 weeks (Fig. 2-2). The third and fourth ven-
Central sulcus 20–26 wk tricles are difficult to visualize at this age unless very thin sections are
Precentral sulcus 24–27 wk obtained because the size of the brain is so small (38,39). The globi
Postcentral sulcus 25–28 wk pallidi typically appear hyperintense on T1-weighted images start-
ing at about 20 weeks. This likely represents premyelination changes,
Cerebellum such as the appearance of proteolipid protein in oligodendrocyte
Primary fissure 25–28 wk processes (40).
Between 24 and 30 weeks, the cerebral cortex shows development
In general, the earlier numbers are from pathology studies and the later ones of shallow Rolandic (central), calcarine, pericallosal/callosomarginal,
from radiology studies. Imaging numbers (later numbers) indicate when the
interparietal, and superior temporal sulci (Figs. 2-4 and 2-5); in some
sulcus is seen in more than 75% of studies.
patients, the precentral, postcentral, superior frontal, and middle tem-
From (14–18).
poral sulci may be visualized. The subplate is still seen in the subcortical
white matter as a layer of T1 hypointensity/T2 hyperintensity; it begins
to disappear on imaging in the posterior frontal and parietal lobe at
about 28 weeks, but remains visible in less mature areas such as the
technology (particularly the use of multiarray phased array coils and anterior frontal and temporal lobes for some time thereafter (12,37).
improved pulse sequences) have allowed progressive improvement in Myelination is seen in some brainstem structures during this period,
the quality of the images. Studies (18,27–29,33) indicate that brain including the median longitudinal fasciculus (MLF, bright at 25 weeks
development follows a similar progression of sulcation in fetuses as in on T1-weighted images, dark at 29 weeks on T2-weighted images),
prematurely born infants; sulci are increasingly identified with increas- the lateral lemnisci (bright at 26 weeks on T1-weighted images, dark
ing postconceptional age. However, it appears that sulcal development at 28 weeks on T2-weighted images), the medial lemnisci (bright at
occurs earlier in utero than ex utero. In other words, sulci are seen earlier 27 weeks on T1-weighted images, dark at 30 weeks on T2-weighted
in fetuses still in the uterus than in neonates of similar postconceptional images), and the superior and inferior cerebellar peduncles (bright at
ages that have been born prematurely. The reasons for this are not yet 28 weeks on T1-weighted images, dark at 29 weeks on T2-weighted
clear, nor are the precise differences in timing. Studies also show that images) (41). The basal ganglia and thalami are better seen at this age
the cerebrum and its components grow largely proportionally. For on MRI and have intensity similar to the cerebral cortex on both T1-
example, the volume of cortical gray matter, basal ganglia volumes, and T2-weighted images, although not as hyperintense on T1-weighted

Barkovich_Chap02.indd 22 5/7/2011 2:41:49 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 23

FIG. 2-2. Fetal MR at 20 postconceptional weeks. A–D. Axial images show that the germinal zones (low intensity rims around the ventricles, small
black arrows) and lateral ventricles (v) are rather large at this age. The intermediate zones (large white arrows) are best seen at higher levels (C and D).
E. Coronal image nicely shows the germinal zone (very low intensity, small white arrows), intermediate zone (intermediate intensity, large white arrow),
subplate (higher intensity, large black arrow), and cerebral cortex (similar low intensity to germinal zone, small black arrows).

images or as hypointense on T2-weighted images (Fig. 2-4E–L). The (38,39). The dorsal brainstem (relatively hyperintense on T1-weighted
ventrolateral nucleus of the thalamus becomes hypointense compared images and hypointense on T2-weighted images) is contrasted by the
with the remainder of the thalamus on T2-weighted images by about unmyelinated ventral pons (Fig. 2-5A and E). The thalami and globi
25 weeks and hyperintense on T1-weighted images by 27 to 28 weeks, pallidi are contrasted by the unmyelinated (relatively hypointense on
probably due to hypercellularity and possibly to early myelination, T1-weighted images and hyperintense on T2-weighted images) inter-
as well. The lateral ventricles, particularly the trigones and occipital nal capsule (Fig. 2-5B and F). The signal intensity of the entire cerebral
horns, are less prominent at this age than at 22 to 23 weeks, probably cortex is uniform at this age on both T1- and T2-weighted images. The
secondary to both growth of the cerebral white matter and develop- subplate is poorly seen on T1-weighted images and is best identified
ment of the calcarine sulci. in the anterior temporal lobes on T2-weighted images. The germinal
By 31 to 32 weeks, an increased number of gyri and shallow sulci matrix has involuted to a large degree and only a few scattered rem-
become visible in the cerebral cortex of prematurely born neonates nants may be seen. These remnants are most consistently seen just
(Fig. 2-5). The Sylvian fissures retain their immature appearance, anterior to the tips of the frontal horns of the lateral ventricles at this
although some development of the opercula can be detected. The cis- age (Fig. 2-5C and G) (42). The dorsal brainstem and superior and
terns around the brainstem and cerebellum remain large at this age and inferior cerebellar peduncles remain bright on T1-weighted images,
the CSF spaces in the occipital region and in the interhemispheric fissure but the middle cerebellar peduncles remain unmyelinated, isointense
remain prominent, although the interhemispheric fissure is more vari- to the cerebral white matter. T2-weighted MR shows hypointensity
able in size. The cavum septi pellucidi and cavum Vergae are prominent in the dorsal brainstem (predominantly due to the MLF, medial and
and will remain so throughout the first 40 postconceptual weeks lateral lemnisci), superior and inferior cerebellar peduncles, nuclei of

Barkovich_Chap02.indd 23 5/7/2011 2:41:49 AM


24 Pediatric Neuroimaging

FIG. 2-3. Fetal MR at 23 postconceptional weeks. A–D. Axial T1-weighted images show formation of calcarine and parietooccipital fissures.
The high intensity surrounding the lateral ventricles (open arrows) is the germinal matrix. Note the layer of migrating cells (solid arrows) in the
cerebral white matter, seen best on images (C and D); this is believed to represent migrating glia. Note also the small size of the cerebellum at
this age. (These images courtesy Dr. Nadine Girard).

the inferior colliculi, far lateral putamen, and ventrolateral thalamic maturity can be seen at this age, some infants having a gyral pattern
nucleus (43) (Fig. 2-5). The cerebral white matter still appears com- that resembles a term infant and others still appearing quite immature
pletely unmyelinated. (Fig. 2-6) (38,39).
At 34 to 36 weeks, the cerebral cortex has further thickened and By 38 to 40 weeks, the brain has a nearly normal adult sulcal pat-
more sulci have developed. Little change occurs in the signal intensity tern (Fig. 2-7); the sulci are formed but are not as deep as they will
of the white matter between 32 and 36 postconceptional weeks (43). become in the next several weeks. On T1-weighted MR studies, the
On T1-weighted MR, the posterior limb of the internal capsule remains dorsal brainstem, posterior portion of the posterior limb of the inter-
hypointense as compared to the lentiform nucleus (Fig. 2-6C and D); nal capsule, and the central portion of the corona radiata (the corti-
some patients will show a small dot of hyperintensity in the posterior cospinal tracts) are hyperintense compared to the rest of the brain. On
aspect of the posterior limb at 39 weeks (41). On T2-weighted images, T2-weighted images, the dorsal brainstem is hypointense and, at 39 to
the posterior limb of the internal capsule remains entirely hyperintense 40 weeks, a characteristic spot of hypointensity is present in the pos-
compared with surrounding structures (Fig. 2-6J). The Sylvian fissures terior limb of the internal capsule, lateral to the hypointense lateral
and, to a lesser extent, the CSF spaces near the occipital pole, remain thalamic nuclei. There are few differences between the CT images of
prominent (Fig. 2-6D, E, J and K). Considerable variation in brain a newborn term infant and those of older infants. The frontal white

Barkovich_Chap02.indd 24 5/7/2011 2:41:50 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 25

FIG. 2-3. (Continued) E–H. Axial single-shot half-Fourier RARE images through the same levels show the same features.
Note the complete absence of myelination in the white matter at this age and the smaller germinal zones compared with
the 20-week fetus illustrated in Figure 2-2. Note also that the subplate and intermediate zones, although still visible, are less
conspicuous at this age than at 20 weeks.

matter and parietooccipital white matter remain relatively low in may also remain somewhat large for several months. A cavum Vergae
attenuation compared to the gray matter (Fig. 2-7A–E). This probably and cavum septi pellucidi are usually present at birth; they disappear
results from the known high water content of the newborn brain and rapidly in the first few months after birth as the septal leaves fuse.
the lack of myelination. The MR appearance of the newborn brain, on The cisterna magna and basilar cisterns are relatively large through-
the other hand, is considerably different from that in older children, out infancy. This enlargement is quite apparent on MR scans, but less
as discussed in the following section. The Sylvian fissures may remain so on CT where only the axial plane is available and beam hardening
prominent in the immediate newborn period; the occipital CSF spaces artifact frequently obscures details in the basilar cistern area.

Barkovich_Chap02.indd 25 5/7/2011 2:41:51 AM


26 Pediatric Neuroimaging

FIG. 2-4. MR of 28-week fetus and of 28-week


prematurely born neonate. (A–D) are single-shot
half-Fourier RARE images, (E–H) are SE 550/15 MR
images, and (I–L) are SE 3000/120 MR images. Note
that sulcation in more advanced in the fetus than in
the prematurely born neonate of a similar gestational
age. By this age, gyri and sulci other than the sylvian
fissure become detectable. The development of shal-
low Rolandic (central), calcarine, pericallosal/callo-
somarginal, interparietal, and superior temporal sulci
can be visualized. In addition, the germinal matrix is
less prominent. The basal ganglia and thalami are
better seen at this age and have intensity similar to the
cerebral cortex on both T1- and T2-weighted images,
although not as hyperintense on T1-weighted images
or as hypointense on T2-weighted images. The lat-
eral ventricles, particularly the trigones and occipi-
tal horns, are less prominent at this age than at 22
to 23 weeks, probably secondary to both growth of
the cerebral white matter and development of the
calcarine sulci.

Barkovich_Chap02.indd 26 5/7/2011 2:41:53 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 27

FIG. 2-4. (Continued)

Grading of Brain Maturation fetal brain. A summary of these measurements is listed in Table 2-2. For
in Premature Infants more extensive lists of measurement, the book by Garel (11) and the
paper by Parazzini (31) are recommended.
Childs et al. (44) have proposed a scoring system for assessment of brain
maturation in premature infants. The scoring system takes into account
the degree of myelination, extent of sulcation, amount of involution of
NORMAL POSTNATAL BRAIN
the germinal matrix of the lateral ventricles, and the character of the DEVELOPMENT
band of migrating cells (if present) in the white matter of the brain.
Embryology
This method shows considerable potential for determining whether
the brains of neonates are developing properly. However, norms and From the imaging perspective, postnatal brain development consists
standard deviations will need to be established for each weekly post- primarily of changes in signal intensity secondary to the process of
conceptional age, and it must be determined whether these scores are myelination. Myelination of the brain begins during the fifth fetal month
sensitive and specific for predicting developmental abnormality. with the myelination of the cranial nerves and continues throughout
Another way to assess brain maturation is size. Several authors have life. In general, the myelination progresses from caudal to cephalad, and
performed the service of measuring many structures of the developing from dorsal to ventral. The brainstem, therefore, myelinates prior to

Barkovich_Chap02.indd 27 5/7/2011 2:41:54 AM


28 Pediatric Neuroimaging

FIG. 2-5. MR of normal 31-week premature infant. Images (A–D) are SE 600/11 images and (E–H) are SE 3000/120 images. More sulci have developed by
this age, although they are still rather shallow. The myelinated dorsal brainstem is contrasted by the unmyelinated ventral pons (A and E) and the thalami
and globi pallidi are contrasted by the completely unmyelinated internal capsule (B, C, F, and G). The germinal matrix has involuted considerably, but some
gray matter signal remains present along the lateral walls of the lateral ventricles, most prominently seen at the tips of the frontal horns (arrows in C and G);
this can persist until the end of the 44th gestational week. The cisterns in the occipital region remain prominent (C, G, and H). The signal intensity of the
entire cerebral cortex is uniform at this age on both T1- and T2-weighted images. Foci of gray matter intensity are seen just anterior to the tips of the frontal
horns of the lateral ventricles (arrows in C), representing foci of residual germinal matrix. Hyperintensity on T1-weighted images and hypointensity on
T2-weighted images is present in the dorsal brainstem, superior and inferior cerebellar peduncles, far lateral putamen, and ventrolateral thalamic nucleus.
The cerebral white matter still appears completely unmyelinated.

the cerebellum and basal ganglia, and the cerebellum and basal ganglia Another general trend in the maturation of the brain is that
myelinate prior to the cerebral hemispheres. Another generalization is myelination progresses more rapidly in functional systems that are
that, within any particular portion of the brain, the posterior region utilized in early life than in those that are not utilized until the child
tends to myelinate first. Therefore, the dorsal brainstem, containing the is older. Therefore, in the brainstem the medial longitudinal fascicu-
medial lemniscus and medial longitudinal fasciculus, myelinates prior lus, lateral and medial lemnisci, and inferior and superior cerebellar
to the ventral brainstem, which contains the corticospinal tracts. Like- peduncles, which transmit vestibular, acoustic, tactile, and proprio-
wise, the occipital lobes of the cerebral hemispheres myelinate early, ceptive sense, are myelinated at birth, whereas the middle cerebellar
whereas the prefrontal regions myelinate late. peduncles, which transmit motor impulses into the cerebellum, acquire

Barkovich_Chap02.indd 28 5/7/2011 2:41:55 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 29

FIG. 2-5. (Continued)

FIG. 2-6. Normal 34/35-week premature


infant. Images (A–F) are SE 550/15 MR
images, and (G–L) are SE 3000/120 MR
images. More sulci are forming, as can be
seen along the interhemispheric fissure
and over the convexities. Sulcal develop-
ment varies considerably at this age. The
sylvian fissures have markedly diminished
in prominence due to opercular develop-
ment. Notice that the dorsal brainstem
and globi pallidi have increased in signal
intensity on the axial T1-weighted MRI
(A–F). The posterior limb of the inter-
nal capsule remains completely unmyeli-
nated at this age. On T2-weighted images,
the brainstem nuclei, the periphery of
the cerebellar dentate nuclei, and the cer-
ebellar vermis are relatively hypointense
structures in the posterior fossa (G and
H). The subthalamic nuclei (arrows in I)
have become hypointense.

Barkovich_Chap02.indd 29 5/7/2011 2:41:57 AM


30 Pediatric Neuroimaging

FIG. 2-6. (Continued)

Barkovich_Chap02.indd 30 5/7/2011 2:41:58 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 31

FIG. 2-7. CT/MR of normal 38 to 40 week infant. A–E. With the exception of increased lucency of the frontal and temporo-parietooccipital white
matter, the CT scan of this infant resembles any normal infant during the first year of life. The sulcal pattern is nearly mature. A cavum septi pel-
lucidi is frequently prominent at this age. F–J. SE 550/15 images show high signal intensity in the dorsal brainstem, the decussation of the superior
cerebellar peduncles, the optic tracts, the posterior limbs of the internal capsules, the lateral thalamus, the optic radiations, and the central corona
radiata. There is also increased signal intensity in the Rolandic and perirolandic gyri, corresponding to known myelination of the white matter
within these gyri shortly after birth.

Barkovich_Chap02.indd 31 5/7/2011 2:42:01 AM


32 Pediatric Neuroimaging

FIG. 2-7. (Continued) K–O. SE 3000/120 images show low signal intensity in the cerebellar
vermis, dorsal brainstem, posterior aspect of the posterior limb of the internal capsule, the
ventrolateral thalamus and the perirolandic gyri of the cortex. The T2-weighted images cor-
respond more closely to the temporal sequence of brain myelination as demonstrated with
histochemical staining techniques.

Barkovich_Chap02.indd 32 5/7/2011 2:42:04 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 33

TABLE 2-2 Normal Fetal Brain Measurements


Measurement 20 wk 21 wk 22 wk 23 wk 24 wk
Antero-posterior vermis Minimum 5 mm 5 mm 6 mm 7 mm 7 mm
Median 6 mm 7 mm 7 mm 8 mm 8 mm
Maximum 7 mm 9 mm 9 mm 10 mm 11 mm
Supero-inferior vermis Minimum 8 mm 8 mm 8 mm 9 mm 10 mm
Median 9 mm 10 mm 11 mm 11 mm 12 mm
Maximum 10 mm 12 mm 12 mm 13 mm 14 mm
Transverse cerebellum Minimum 18 mm 19 mm 20 mm 21 mm 23 mm
Median 19 mm 21 mm 22 mm 24 mm 25 mm
Maximum 21 mm 23 mm 24 mm 27 mm 28 mm
Length corpus callosum Minimum 16 mm 16 mm 17 mm 17 mm 23 mm
Median 18 mm 18 mm 21 mm 22 mm 24 mm
Maximum 18 mm 23 mm 24 mm 27 mm 28 mm
Fronto-occipital diameter Minimum 49 mm 50 mm 54 mm 57 mm 65 mm
Median 52 mm 56 mm 58 mm 63 mm 67 mm
Maximum 55 mm 60 mm 62 mm 67 mm 74 mm
Cerebral biparietal diameter Minimum 37 mm 36 mm 40 mm 41 mm 47 mm
Median 39 mm 40 mm 43 mm 46 mm 49 mm
Maximum 40 mm 45 mm 45 mm 49 mm 52 mm

Adapted from Parazzini C, Righini A, Rustico M, Consonni D, Triulzi F. Prenatal magnetic resonance imaging: brain normal linear biometric values
below 24 gestational weeks. Neuroradiology 2008;50:877–883. With permission.

myelin later and more slowly. Similarly, in the cerebrum, the geniculate change by conventional MR imaging probably reflects the fact that the
and calcarine (optic), postcentral (somesthetic), and precentral (pro- relaxation rates of gray and white matter change proportionately; thus,
prio-kinesthetic) regions acquire myelin early, whereas the posterior no relative change is appreciated.
parietal, temporal, and frontal areas, which are association areas that
integrate the sensory experience, acquire myelin later (22,45–47). T1-Weighted Images
Yakovlev and Lecours (47), staining the brain with the Weigert stain The changes of white matter maturation are visually detected at dif-
for myelin, showed that myelination proceeds rapidly within the brain ferent rates and at different times on T1-weighted images than on
up to about of 2 years of age. The process slows markedly after 2 years, T2-weighted images. On T1-weighted images, the appearance of the
although fibers to and from the association areas of the brain continue newborn brain is grossly similar to that of T2-weighted images in
to myelinate well into the third and fourth decades of life. adults in that white matter has lower signal intensity than gray matter.
As white matter matures, its signal intensity increases relative to gray
matter.
Spin-Echo MRI of Post-Natal Brain Development
Neonatal posterior fossa structures that exhibit high signal inten-
In general, changes in white matter maturation are seen best on sity at birth include the medial lemniscus, lateral lemniscus, MLF,
T1-weighted images during the first 6 to 8 months of life and on the long brachium of the inferior colliculus, and the inferior and superior cer-
echo time T2-weighted images between the ages of 6 and 18 months. ebellar peduncles (Fig. 2-7) (50). An increase in signal intensity of the
Maturation of both the brainstem and cerebellum seem to be more deep cerebellar white matter appears near the end of the first month of
sensitively assessed on T2-weighted images (48–50). We obtain both life and steadily increases, with high signal intensity developing in the
T1- and T2-weighted axial sequences for imaging patients in these age subcortical white matter of the cerebellar folia by the third month. At
groups, using parameters described in Chapter 1 (three-dimensional 3 months of age, the cerebellum has an appearance similar to that seen
[3D] spoiled gradient-echo sequence for T1-weighted images and in the adult on both axial and sagittal images. Signal intensity in the
conventional spin-echo T2-weighted images with long repetition and basis pontis (ventral pons) increases less rapidly, occurring during the
echo times). Other imaging sequences that give heavily T1-weighted third through the sixth months.
and T2-weighted images will work, as well. For example, fast spin-echo In the supratentorial region, the decussation of the superior cer-
T2-weighted sequences will show changes of myelination, although ebellar peduncles, the ventral lateral region of the thalamus, the globus
the brain appears slightly more myelinated on fast spin-echo than on pallidus, the posterior portion of the posterior limb of the internal
conventional spin-echo images (51). Although visually apparent signal capsule, and the central portion of the corona radiata (the corticospi-
changes of the white matter seem to be complete by about the age of nal tracts) exhibit high signal intensity at birth (Fig. 2-7) (50). In addi-
2 years, measurements of relaxation rates have shown that T1 shorten- tion, small foci of gray matter intensity are seen just anterior to the
ing of white matter and gray matter continues into adolescence, prob- tips of the frontal horns of the lateral ventricles in term neonates and
ably secondary to continued myelination and consequent diminution premature infants. These foci, which are believed to represent persis-
of brain water (52). The inability to qualitatively detect this continuing tent germinal matrix, disappear by about 44 postconceptional weeks

Barkovich_Chap02.indd 33 5/7/2011 2:42:06 AM


34 Pediatric Neuroimaging

(42,53). The development of high signal intensity proceeds rostrally are seen in the central centrum semiovale; this makes it difficult to
from the pons along the corticospinal tracts into the cerebral pedun- distinguish white matter from the surrounding cortex because both
cles, posterior limb of the internal capsule and the central portion of have low signal intensity. By age 4 months, the intensity of the precen-
the centrum semiovale. The white matter of the precentral and post- tral and postcentral gyri is indistinguishable from that of adjacent gyri,
central gyri is of high signal intensity compared with surrounding which have also diminished in signal intensity (Fig. 2-8). Low signal
cortex by about 1 month of age (Fig. 2-7). The change to high sig- intensity is seen in the optic tracts at birth in some patients and at
nal intensity in the subcortical motor tracts is essentially complete by age 1 month in most; the decrease in signal intensity extends poste-
age 3 months. In infants less than 1-month-old, high signal intensity riorly along the optic radiations during the subsequent 2 months; by
is present in the optic chiasm and optic tracts; by age 3 months, the 4 months of age, the calcarine fissure shows some low signal intensity.
occipital white matter surrounding the calcarine fissure is of high sig- By age 4 months, the globi pallidi are slightly hyperintense compared
nal intensity. The posterior aspect of the posterior limb of the internal to the putamina; they will remain hyperintense until 8 to 10 months,
capsule is of high signal intensity at birth; high signal intensity does when the globi pallidi and putamina become isointense again.
not develop in the anterior limb until 2 to 3 months of age. The sple- Most deep white matter tracts of the cerebrum decrease in signal
nium of the corpus callosum shows high signal intensity in all infants intensity between 6 and 12 months of age (Figs. 2-9 to 2-12). The inter-
by age 4 months (Fig. 2-8). The increase in signal intensity proceeds nal capsule matures in a posterior-to-anterior fashion. The more ante-
anteriorly; the genu is always of high signal intensity by age 6 months rior portion of the posterior limb contains a thin strip of hypointensity
(Fig. 2-9). Typically, at 4 to 5 months of age, the splenium is high in by approximately 7 months; progressive thickening of the hypointense
signal intensity, while the genu is low in signal intensity. Maturation of area continues up to 10 months of age. The anterior limb of the inter-
the subcortical white matter, other than the visual and motor regions, nal capsule is completely hypointense by 11 months in normal patients;
begins at 3 months. The deep white matter matures in a posterior- hypointensity can be detected as early as 7 months in some patients but
to-anterior direction, with the deep occipital white matter maturing is always preceded by the presence of low signal intensity in the poste-
first and the anterior frontal and temporal white matter last. Peripheral rior limb. The corpus callosum matures from posterior to anterior; the
extension and increasing hyperintensity of the subcortical white mat- splenium shows low signal intensity by age 6 months and the genu by
ter continue until approximately age 7 months in the occipital white age 8 months (Figs. 2-9 and 2-10). The basal ganglia begin to dimin-
matter and 8 to 11 months in the frontal and temporal white mat- ish in signal intensity relative to the subcortical white matter at 5 to
ter (Fig. 2-10). Only minimal changes are seen on the T1-weighted 7 months of age. This appearance gradually fades as the surrounding
images after 11 months, consisting of increasing signal intensity in the brain decreases in signal intensity as a result of myelination. The basal
most peripheral regions of the anterior frontal, anterior temporal, and ganglia appear essentially isointense with the subcortical white matter
parietal white matter (22). by the age of approximately 10 months (Fig. 2-11). The globus pallidus
will become hypointense with respect to white matter again around the
T2-Weighted Images end of the first decade of life; this decrease in intensity results from iron
The overall appearance of the newborn brain on T2-weighted images deposition and will be described later in this chapter.
is grossly similar to that of adult T1-weighted images in that the white The subcortical white matter (other than the calcarine and Rolan-
matter has higher signal intensity than the gray matter. On T2-weighted dic areas) matures last, proceeding from the occipital region anteri-
sequences, white matter maturation is seen as a reduction in signal orly to the anterior frontal and temporal lobes. In the past, subcortical
intensity. As stated earlier, T2-weighted images are probably superior white matter maturation was described as being complete by about
to T1-weighted images for assessment of maturation of the cerebellum 24 months (22). With improved signal-to-noise and thinner imag-
(54) and brainstem (48). ing sections on modern MR scanners, some unmyelinated subcortical
At birth, low signal intensity is present in the inferior and superior white matter can still be detected a bit later, but it is still essentially
cerebellar peduncles and the cranial nerve nuclei (particularly cranial complete by 30 months. Myelination of deep white matter begins at 9
nerves VI, VII, and VIII) (50). Small foci of gray matter intensity are to 12 months of age in the occipital lobe and at 11 to 14 months fron-
seen just anterior to the tips of the frontal horns in premature and term tally (Figs. 2-12 and 2-13); deep white matter in the anterior temporal
neonates. As discussed in the prior section on T1-weighted images, lobe matures last. Extension of the low signal intensity into the subcor-
these represent foci of persistent germinal matrix that disappear by tical white matter begins at about 1 year in the perirolandic region and
about 44 postconceptional weeks (42,53). The cerebellar vermis (Fig. is essentially complete, even on modern images, by 24 to 28 months
2-7K) and the flocculi cerebellum are also of low signal intensity. The (Fig. 2-14). Thus, with the exception of the so-called terminal zones
ventral brainstem becomes of similar low intensity to the dorsal brain- (see following section), white matter maturation, as assessed by MR, is
stem at about the fifth postnatal month. The middle cerebellar pedun- complete in the early part of the third year of life. During the progress
cles begin to decrease in signal intensity during the second month of of the peripheral extension of the low signal intensity within the white
life and are of uniform low intensity by age 3 months (55). The cerebral matter, the mantle of gray matter gives the appearance of progressive
peduncles become low in intensity by 4 months and the red nuclei by thinning, and the subcortical white matter often has a heterogeneous
5 months (55). Low signal intensity is seen in the subcortical white appearance.
matter of the cerebellar folia during the fifth to eighth month and the
cerebellum reaches an adult appearance at approximately 18 months.
Terminal Zones
Supratentorial structures that show low signal intensity at birth
include the decussation of the superior cerebellar peduncles, the medial As maturation in the centrum semiovale progresses, nearly all sub-
and lateral geniculate bodies, the subthalamic nuclei, the ventral lateral jects have persistent areas of high signal intensity in the white matter
regions of the thalami, a small patch of the posterior portion of the lateral to the bodies of the lateral ventricles and, more prominently,
posterior limbs of the internal capsules, and, a small linear region in dorsal and superior to the ventricular trigones on T2-weighted images
the lateral putamina (50). By less than 1 month of age, the cortex in the (Fig. 2-15). These areas are most often homogeneous, although in
precentral and postcentral gyri has lower intensity than the surround- some patients they may be patchy. They are more difficult to identify on
ing cortex (Fig. 2-7). By age 2 months, patches of low signal intensity the first echo of the long TR sequence than on the second and usually

Barkovich_Chap02.indd 34 5/7/2011 2:42:06 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 35

FIG. 2-8. MRI of the brain of a normal 4-month-old. A–E. SE 550/15 images show rostral progres-
sion of the maturation of the internal capsule; the anterior limbs of the anterior capsule are now well
myelinated. The splenium of the corpus callosum should always have high signal intensity by this age.
Notice the isointensity of the cortical gray matter and subcortical white matter, resulting in difficulty
in the identification of structural abnormalities at this age on T1-weighted images. F–G. Note the
relative lack of change on the T2-weighted image from the neonate (Fig. 2-7).

Barkovich_Chap02.indd 35 5/7/2011 2:42:06 AM


36 Pediatric Neuroimaging

FIG. 2-9. MRI of the brain of a normal 6-month-


old. A–E. SE 550/15 images show further progres-
sion of brain maturation. Both the splenium and
the genu of the corpus callosum are of high signal
intensity at this age. There has been progression
of the maturation of the centrum semiovale with
increasing hyperintensity of subcortical white
matter, most notably in the occipital and para-
central regions. F–J. SE 3000/120 images reveal a
diminution of signal intensity within the centrum
semiovale. Additionally, there is a relative decrease
in the signal intensity of the basal ganglia with
respect to the surrounding brain. The splenium
of the corpus callosum is of low signal intensity at
this age and there are patches of low signal inten-
sity within the callosal genu.

Barkovich_Chap02.indd 36 5/7/2011 2:42:08 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 37

FIG. 2-9. (Continued)

FIG. 2-10. MRI of the brain of a normal


8-month-old. A–E. SE 550/15 images at
this age show essentially an adult appear-
ance at first inspection. Hyperintensity of
the subcortical white matter tracts is seen
in the paracentral and occipital regions
but is not yet present in the frontal or
parietal regions.

Barkovich_Chap02.indd 37 5/7/2011 2:42:11 AM


38 Pediatric Neuroimaging

FIG. 2-10. (Continued) F–J. On SE 3000/120


images, the anterior limbs of the internal capsule are
starting to show diminished signal intensity. Both
the splenium and the genu of the corpus callosum
are of low signal intensity at this age. The white
matter in the occipital and paracentral regions is
now isointense with the overlying cortex.

appear isointense to surrounding white matter on a proton density in irregularity of the ventricular wall, abnormally deep cortical sulci,
image. The primary cause of this high signal intensity is probably the with the cortex sometimes extending down to the ventricular surface,
known delayed myelination of the fiber tracts involving the associa- and thinning of the body of the corpus callosum (see examples of white
tion areas of the posterior and inferior parietal and posterior temporal matter injury of prematurity in Chapter 4). Baker et al. (56) suggested
cortex. Large perivascular spaces probably contribute to the high signal that the differentiation between these normal peritrigonal areas of high
intensity, particularly in the peritrigonal area (see subsequent para- signal intensity and white matter injury of prematurity is very difficult.
graph in this section). Yakovlev and Lecours have called these regions They noted, however, that a layer of myelinated white matter is present
the “terminal zones” because some of the axons in these regions do not between the trigone of the ventricle and the terminal zones in normal
stain for myelin until the fourth decade (47). These areas of persistent patients (Fig. 2-15). When the peritrigonal high signal is due to periven-
high signal intensity are seen throughout the first decade and, in some tricular leukomalacia, this layer of normally myelinated white matter is
patients, into the second decade of life. absent. Differentiation from hydrocephalus is usually easier as a shunt
It is important to differentiate the terminal zones from white mat- or third ventriculostomy (see Chapter 8) can be detected on the MR
ter injury resulting from prematurity (“periventricular leukomalacia” study. White matter disease due to metabolic disorders is typically much
[see Chapter 4]) and hydrocephalus (see Chapter 8), and from meta- more extensive than peritrigonal; details are described in Chapter 4.
bolic disorders (see Chapter 3); all can also be associated with areas
of prolonged T2-relaxation in the peritrigonal region. In general, the Large Perivascular Spaces
white matter injury of prematurity is more sharply defined and is situ- Another condition that can mimic both the terminal zones and periven-
ated more inferiorly, lateral to the trigones and near the optic radia- tricular leukomalacia on MR is enlargement of perivascular spaces
tions. Injured white matter is of very high intensity and is typically (Fig. 2-15D–F). Indeed, as stated above, the high signal of the perivas-
bright on the first echo of the T2-weighted sequence. Moreover, white cular spaces may contribute to the T2 hyperintensity of the terminal
matter injury is associated with loss of brain tissue, typically resulting zones. Studies have shown that perivascular spaces are commonly seen

Barkovich_Chap02.indd 38 5/7/2011 2:42:13 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 39

FIG. 2-11. SE 3000/120 MRI of the normal


10-month-old infant. T2-weighted images
demonstrate decreasing signal intensity of
the white matter diffusely throughout the
brain. This causes a recession of the appear-
ance of hypointensity of the basal ganglia
(C,D) with respect to the white matter noted
in the 6- and 8-month-old patients. The cor-
tex and underlying white matter are essen-
tially isointense throughout most of the brain
at this age. For this reason, structural abnor-
malities of the brain at this age are identified
best on T1-weighted images. The anterior
limbs of the internal capsule are hypointense
with respect to surrounding structures in
essentially all patients at this age (C,D).

with modern, high quality MR studies. With thin section volumetric the first 6 months of life, T1-weighted images are most useful for
T1-weighted images, Groeschel et al. (57) were able to identify them assessing normal brain maturation. On T1-weighted images, high
in 125 out of 125 normal volunteers below the age of 30 years; with signal intensity should appear in the anterior limbs of the internal
routine 5 to 6 mm sections, they were identified in 80% of a group capsules and should extend distally from deep cerebellar white mat-
of pediatric clinical scans. Focally expanded or “dilated” perivascular ter into the cerebellar folia, by 3 months of age. The splenium of the
spaces, as defined by Groeschel et al., are seen in 1.5% to 3% of nor- corpus callosum should be of moderately high signal intensity by
mal subjects (57,58). Although most patients with dilated perivascular the fourth month, and the genu of the corpus callosum should be
spaces, and even those with “giant” perivascular spaces (57), are neu- of high signal intensity by age 6 months. An essentially adult pat-
rologically normal, evidence has been presented that affected patients tern is seen by approximately 8 months of age with the exception
have a higher incidence of neuropsychiatric disorders than the general that some of the most subcortical white matter fibers (particularly in
pediatric population (58). the anterior frontal and anterior temporal lobes) have not acquired
high signal intensity. After age 6 months, T2-weighted images are
more useful in the assessment of normal brain maturation. On
Milestones
T2-weighted images, the splenium of the corpus callosum should
Barkovich et al. (22) charted the ages at which the changes of myeli- be of low signal intensity by 6 months of age, the genu of the corpus
nation appeared on T1- and T2-weighted images, as well as nor- callosum by 8 months of age, and the anterior limb of the internal
mal MRI milestones for myelination of brain (Table 2-3). During capsule by 11 months of age. The deep frontal white matter should

Barkovich_Chap02.indd 39 5/7/2011 2:42:14 AM


40 Pediatric Neuroimaging

FIG. 2-12. SE 2500/80 MRI of the brain of


a normal 12-month-old. There is increasing
low signal intensity of the white matter in the
paracentral and occipital regions. The images
are otherwise very similar to the 10-month-
old infant.

be of low signal intensity by age 14 months. With the exception of T2 relaxation times. The second component, composed of axonal
the subcortical white matter, the entire brain should have an adult and extracellular water, has longer T1 and T2 relaxation times (61).
appearance by 18 months, and the subcortical white matter should The new technique of myelin water imaging allows specific analysis
be mature by about 30 months. of myelination by this technique (62), but such techniques are not yet
available for clinical use.
Postulated Causes of T1 and T2 Shortening In the time scale of typical T1 values, myelin water can diffuse
across axonal and myelin membranes and interact with water mol-
Associated with Myelination
ecules in the other compartments. It is known that the T1 shortening
It is interesting to note that brain maturation occurs at different rates in developing white matter correlates temporally with the increase in
and times on T1-weighted SE images, T2-weighted SE images, and cholesterol and glycolipids that accompany the formation of myelin
inversion recovery images with short inversion time (STIR) (22,59). from oligodendrocyte processes (22,63). In addition, galactocerebro-
The exact reasons for these differences have not been entirely worked sides, which are among the most prevalent glycolipids in myelin, cause
out. It is known that there are two distinct components of water con- marked T1 shortening and magnetization transfer in saline solutions
tributing to MR images in white matter; these lead to a biexponential in vitro (64). Galactocerebrosides are prevalent on the oligodendrocytic
curve of T2 relaxation during white matter development (60). The processes as they begin to envelop the axon in response to increasing
first, composed of intramyelinic water, has relatively short T1 and electrical activity (65). Thus, the aqueous interaction of cholesterol (66)

Barkovich_Chap02.indd 40 5/7/2011 2:42:15 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 41

FIG. 2-13. SE 2500/80 MRI of the


brain of a normal 15-month-old. The
maturation of the deep white matter
has progressed significantly since the
12-month-old stage. Although somewhat
patchy, the subcortical white matter is
now hypointense in a great deal of the
cerebrum. The maturation of the white
matter is slowest in the frontal and tem-
poral lobes.

FIG. 2-14. SE 2500/80 MRI


of the brain of a normal
22-month-old. The appearance
of the brain is essentially identi-
cal to that of an adult. Notice,
however, that there is still some
patchy high signal intensity in
the anterior frontal white mat-
ter and in the white matter
parallel to the lateral ventricles.
This is most prominent dorsal
to the bodies and trigones of the
lateral ventricles.

Barkovich_Chap02.indd 41 5/7/2011 2:42:16 AM


42 Pediatric Neuroimaging

FIG. 2-14. (Continued)

FIG. 2-15. Terminal zones and large perivascular spaces. A–C. Axial SE 2500/80 (A) and coronal SE 2500/80 (B) and FLAIR (C) images show persistent
increased signal intensity lateral, superior, and posterior to the lateral ventricles (arrows), particularly in the region of the trigones. These regions probably
represent areas of known slow myelination within the brain (sometimes called “terminal zones”) and should not be mistaken for areas of ischemia or brain
damage. They are seen from about age 16 months until age 10 years. D–F. T1 (C) and first and second echo T2-(D and E) weighted images show curvilinear
periventricular areas that are isointense to CSF on all imaging sequences. This is the classic appearance for perivascular spaces.

Barkovich_Chap02.indd 42 5/7/2011 2:42:18 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 43

TABLE 2-3 Ages when Changes of Myelination Appear


Anatomic Region T1-Weighted Images T2-Weighted Images
Superior cerebellar peduncle 28 gest wk 27 gest wk
Median longitudinal fasciculus 25 gest wk 29 gest wk
Medial lemnisci 27 gest wk 30 gest wk
Lateral lemnisci 26 gest wk 27 gest wk
Middle cerebellar peduncle Birth Birth to 2 mo
Cerebral white matter Birth to 4 mo 3–5 mo
Posterior limb internal capsule
Anterior portion First month 4–7 mo
Posterior portion 36 gest wk 40 gest wk
Anterior limb internal capsule 2–3 mo 7–11 mo
Genu corpus callosum 4–6 mo 5–8 mo
Splenium corpus callosum 3–4 mo 4–6 mo
Occipital white matter
Deep 3–5 mo 9–14 mo
Subcortical 4–7 mo 11–15 mo
Mid-frontal white matter
Deep 3–6 mo 11–16 mo
Subcortical 7–11 mo 14–18 mo
Anterior frontal white matter
Deep 5–8 mo 12–18 mo
Subcortical 10–15 mo 24–30 mo
Centrum semiovale 2–4 mo 7–11 mo
gest wk, gestational weeks; mo, month.

and galactocerebrosides on the surface of the myelin bilaminar Altered packing of these lipids results in increases in T2 relaxation
membrane are most likely responsible for the early T1 shortening we times (72), a point that is important in the imaging of inborn errors
see in developing white matter. of metabolism, discussed in Chapter 3. Cholesterol, glycolipids, and
At the relatively long echo times used in acquiring T2-weighted portions of the myelin proteins are hydrophilic; that is, they hydrogen
images in neonates, only the signal from axonal and extracellular water bond strongly with water molecules (73–75). Therefore, it is likely that
is likely to contribute to the MR signal (67). Furthermore, the increas- the initial T1 shortening seen by MR results from an increase in the
ing hypointensity seen on T2-weighted images of the maturing brain amount of bound water in the brain (and consequent decrease in the
correlates temporally with chemical maturation of the myelin sheath, amount of free water) secondary to hydrogen bonding of free water to
specifically with tightening of the myelin spiral around the axon (asso- surface galactocerebrosides and, possibly, cholesterol and proteins.
ciated with conformational changes of myelin proteins) and satura-
tion of polyunsaturated fatty acids within the myelin membranes
Other Approaches to Brain Maturation by MR
(22,68–71). If, indeed, this T2 signal is entirely from extracellular and
axonal water, it may reflect decreasing proton density. This decrease in Many different approaches have been used to grade brain matura-
proton density is possibly related to the further (hydrophobic) chemi- tion according to the changes in T1 and T2 relaxation. Some authors
cal maturation of myelin, to decreasing axonal water secondary to (59,76–78) have been largely descriptive. Others (22,39) have tried to
microtubule and microfilament production, or to decreasing extracel- quantify myelination and create milestones of normal myelination
lular free water secondary to myelin production and the elaboration of by which delayed myelination can be identified. Dietrich et al. (79)
glia and glial processes in the white matter. approached the subject of normal brain maturation by dividing the
Other factors are likely to be of importance, as well, and some of appearance of the brain on T2-weighted spin-echo images into three
these are important for our understanding of white matter diseases. patterns: (a) infantile (birth to 6 months), (b) isointense (8–12 months),
Proper packing of lipids in the bilaminar membrane is critical to the and (c) early adult (10 months onward). In the infantile pattern, the
stability of myelin and results in short T2 relaxation times in vitro (72). cerebral white matter is hyperintense relative to gray matter, whereas

Barkovich_Chap02.indd 43 5/7/2011 2:42:19 AM


44 Pediatric Neuroimaging

the adult pattern shows hypointense white matter. The appearance of may be difficult to see on routine sagittal images, particularly in fetuses
the isointense and early adult patterns is delayed in patients with devel- of gestational age less than 24 weeks (Fig. 2-16A–C). In the near-term
opmental delay. A similar, but more complex staging system, dividing and term infant, the corpus is usually visible and the signal intensity
brain maturation into five stages, has been proposed by Staudt et al. approaches that of cortical gray matter on T1-weighted images. The
(80,81). Bird et al. (82) determined that the gray and white matter callosal shape at this age is thin and flat; the bulbous enlargements seen
should be isointense by age 4 months on T1-weighted images and by at the adult genu and splenium are not present (103) (Fig. 2-16D and E).
9 to 10 months on T2-weighted images. They considered the age at The first postnatal change is a substantial, albeit variable in time, thick-
which gray and white matter are isointense as a critical factor in evalu- ening of the genu, which frequently occurs as early as the second and
ating patients for developmental delay. Some authors have analyzed the third months of life. DeLacoste et al. (104) have demonstrated that the
images in terms of patterns and attempted to assess degree of, and delay axons crossing through the genu come from the inferior frontal and
in, maturation on the basis of the pattern (48,49,83–85). We choose to anterior, inferior parietal regions. The enlargement of the genu, there-
assess maturation through the use of the normal milestones described fore, presumably relates to the myelination of the interhemispheric
above, as it is a rapid and reliable method that does not require any post connections of the inferior portions of the precentral and postcentral
processing of the imaging data. Indeed, any of the methods described gyri; these areas, which are involved with basic motor and sensory
above are easy to use and reliable. function, develop early in life.
A number of studies (86,87) have postulated the existence of a win- At birth, the splenium is intermediate in size between the body
dow of time during which delayed myelination can be detected by MR. and the genu of the corpus callosum. It enlarges slowly until the fourth
This window seems to be between the ages of 4 months and 2 years. or fifth postnatal month and then rapidly increases in size (Fig. 2-16F
Delayed myelination can be detected in children older than 24 months and G). By the end of the seventh month, the splenium is equal in size
only when it is very severe. to the genu; it then gradually enlarges in proportion with the genu
The advent of diffusion tensor imaging (DTI) (88) allows brain and the rest of the brain through the remainder of the first year (103)
maturation to be quantified by the calculation of regional water diffu- (Fig. 2-16G and H). By about 9 to 10 months, the appearance of the
sion and diffusion anisotropy in the developing brain (25,89–92). This corpus callosum becomes similar to that in the adult (Fig. 2-16I). The
is extremely useful in research but is not essential in the routine assess- axons in the splenium arise from the visual and visual association areas
ment of pediatric brain MR in the reading room. DTI and its use in of the cortex (104,105). Not surprisingly, the rapid development of the
assessing brain maturation are discussed in more detail in section on splenium corresponds temporally with increasing visual awareness at
“Diffusion Tensor Imaging and Tractography” of this chapter. 4 to 6 months of age. It is during this period that the infant devel-
ops binocular vision and visual accommodation and begins to iden-
NORMAL POSTNATAL MR DEVELOPMENT tify objects (106). Both binocular vision and object identification are
dependent on interhemispheric connection. Thus, the enlargement of
OF THE CORPUS CALLOSUM the splenium presumably relates to the myelination of connections
The embryological development of the corpus callosum is discussed in between the visual cortex and the association areas of the brain in the
Chapter 5. Briefly, the axons of the corpus callosum navigate from the increasingly visually aware child.
cerebral cortex to the midline in response to cues from the developing The body of the corpus callosum steadily enlarges throughout
hemisphere (93–96). These axons initially cross the midline near the childhood without any detectable growth spurts. The size of the body
foramina of Monro, at a point that will eventually be the junction of is relatively uniform, except that a focal thinning is frequently seen at
the callosal genu and body, and in the hippocampal commissure, the the junction of the body and splenium (Fig. 2-16H); Raybaud refers
axons of which act as guides that are followed by axons forming the to this section of the corpus as the callosal “isthmus” (107). This focal
callosal splenium. Specialized groups of midline glial cells form a bed thinning is seen in adults as well. McLeod et al. (108) found an isthmus
for ingrowth and midline crossing of the callosal fibers; axons will not in 22% of 450 randomly selected patients. The normal variant should
cross if the proper substrates are not present in the interhemispheric not be mistaken for a segment of hypoplasia.
fissure (97–100). The earliest pioneer axons cross at approximately The lesser enlargement of the callosal body compared with the
12 weeks; the last axons cross the midline at 18 to 20 weeks of gestation. genu and splenium probably represents the anatomic origin of these
Although all the components of the corpus callosum are present by fibers and the phylogenetic development of the brain. Sensory and
20 weeks, growth of the structure is far from complete. From 20 weeks visual function is important early in life for all animals. It is, there-
to term, the length increases 25%; the thickness of the body increases fore, not surprising that the areas of the brain serving these functions
by 30% and the genu by 270% (101). The length of the corpus can myelinate first and that the association tracts related to these func-
be measured postnatally by cranial ultrasound and has been found to tions, which run through the corpus callosum, develop early as well.
grow an average of 0.11 mm/d (range 0.05–0.29), with the rate being The more gradual growth of the body of the corpus callosum probably
similar for infants born at all gestational ages (102). As all of the cal- reflects the lesser importance of association areas in the temporal and
losal axons are presumed to be present at the time of birth, the post- parietal lobes (from which these fibers originate [104,105]) in early life
natal callosal growth presumably directly reflects the myelination of and in lower orders of animal life. The fact that the axons from the
the axons. Because the corpus callosum is easily evaluated by MRI and association areas myelinate later as well (as seen by the late myelina-
transfontanelle sonography, an understanding of the normal appear- tion of the temporal lobes and the persistent areas of prolonged T2
ance of the developing corpus has become important. relaxation superior and dorsal to the trigones) further supports this
The appearance of corpus callosum is quite different in the fetus hypothesis.
and preterm infant than the term neonate and different in the term The length of the corpus callosum changes slowly and erratically
neonate and infant than in the adult. An adult appearance evolves during the first year of life. Callosal length seems to vary more with
slowly over the first 8 to 10 months of life. In the fetus and preterm neo- respect to head size and shape than with age (103), probably because
nate, the corpus callosum is hypointense compared with cortical gray normal infants vary more in head size and shape than in head enlarge-
matter on T1-weighted images and isointense with surrounding brain ment during the first year of life. This hypothesis is supported by
on T2-weighted images; it is extremely thin and of uniform size and the fact that the ratio of callosal length to AP brain diameter is quite

Barkovich_Chap02.indd 44 5/7/2011 2:42:19 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 45

FIG. 2-16. Normal corpus callosum development. A. Midline sagittal single-shot half-Fourier RARE image of a 23 gestational week fetus shows that the
corpus callosum (arrows) is uniformly thin and isointense to surrounding brain. It is, therefore, somewhat difficult to differentiate from surrounding brain.
B. In a 28 gestational week fetus, the corpus is more easily seen, but remains uniformly thin and isointense to surrounding brain. C. Corpus callosum in
a premature neonate of 29 postconceptional weeks. On this SE 500/11 image, the corpus is very hypointense compared to gray matter and very thin. The
presence of the corpus is verified by the normal cingulate gyrus and sulcus above the expected callosal position. D. Corpus callosum in a premature infant of
34 postconceptional weeks. On this SE 500/11 image, the corpus is slightly hypointense compared with gray matter. It is still very thin, but is barely visible.
Note that the cerebellum is still very small at this age.

constant in infants throughout the first year of life (103). Some authors of judging normal maturation of the corpus callosum and, thereby,
have investigated the relationship of the area of the corpus callosum to normal brain development.
maturity (109). Although analysis of callosal area on midline sagittal
images is an accurate method of quantification of white matter matu- NORMAL MR DEVELOPMENT
ration, it is time consuming and, in the clinical setting, has not proven
useful in most practices. The length of the corpus callosum has been
OF THE PITUITARY GLAND
studied in fetuses and premature neonates; findings indicate that the In the fetus and neonate, the pituitary gland has a convex superior
corpus grows more slowly in prematurely born infants than in fetuses surface and a shorter T1 relaxation time (higher signal intensity on
of the same gestational age (102). As described earlier, similar delays of T1-weighted images) than the remainder of the brain (Fig. 2-17A)
maturation have been found in sulcation and myelination of prema- (110–112). The signal intensity and size diminish slowly and linearly
turely born neonates. Overall, however, owing to the insensitivity of until approximately 2 months after birth (Fig. 2-17B), the time at which
callosal length and the difficulty of calculating callosal area as markers the anterior lobe of the pituitary gradually assumes the appearance of a
of normal growth, the changes in callosal shape and signal intensity child, with a flat or slightly concave superior margin and a signal inten-
(see earlier section on “Normal Myelination”) are the preferred means sity of normal gray matter on T1-weighted spin-echo images (113,114).

Barkovich_Chap02.indd 45 5/7/2011 2:42:20 AM


46 Pediatric Neuroimaging

FIG. 2-16. (Continued) E. Normal 1-month-old. On this SE 600/20


image, the corpus callosum is isointense with the rest of the brain. The
corpus callosum is uniformly thin at this age without the normal bulbous
enlargement of the genu and splenium; the genu, body and splenium are
all of the same thickness. F. Normal 4-month-old. By 3 to 4 months of
age, the splenium of the corpus callosum increases in size and begins to
show an increased signal intensity as compared to the rest of the brain
on SE 600/20 images. These changes probably result from the process
of myelination in the visual association fibers. G. Normal 7-month-old.
By 6 to 7 months of age, the corpus callosum is of uniform high signal
intensity as compared with surrounding brain. The genu and splenium
of the corpus are now large as compared to the body. The corpus is still
relatively thin. H. Normal 10-month-old. By 8 to 9 months, the corpus
callosum begins to thicken in the genu and splenium, taking on more of
an adult appearance. The thinning of the corpus at the junction of the
posterior body and splenium (arrow) is a normal variant and does not
connote pathology. I. Normal mature corpus callosum in a 19-year-old.

Barkovich_Chap02.indd 46 5/7/2011 2:42:21 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 47

FIG. 2-17. Development of normal pituitary gland. A. Normal newborn pituitary. The gland is upwardly convex and is uniformly bright on T1-weighted
images. B. Normal infant pituitary gland. By age 2 to 3 months, the adenohypophysis (anterior pituitary) loses its high signal on T1-weighted images,
allowing the bright neurohypophysis (posterior pituitary) to be identified as a separate structure.

Of note, the evolution of signal is similar in infants born at term and in The low signal intensity is believed to result from the presence of red
those born prematurely, indicating that the changes result from adjust- (actively hematopoietic) marrow. By the end of the second year of life,
ments to extrauterine life and are not related to the infant’s devel- the marrow in the crista galli and the nasal processes of the frontal
opmental stage (115). The low signal of the anterior pituitary gland bone develop high signal intensity on T1-weighted images (Fig. 2-19B).
contrasts with the intrinsic high signal of the posterior pituitary gland During the third year of life, patchy areas of increased signal intensity
on T1-weighted images. This high signal is postulated to result from begin to appear in the body of the sphenoid bone and in the clivus on
the effects of the proteins (neurophysins) that transport the posterior T1-weighted images (Fig. 2-19C). These foci of T1-shortening may ini-
pituitary hormones from the hypothalamus to the neurohypophysis tially appear in the basiocciput or the basisphenoid. As time progresses,
(116). During childhood, the pituitary gland slowly grows in all dimen- most of these regions of fatty marrow will undergo pneumatization
sions, maintaining a flat or mildly concave upper surface, with a height to become parts of the paranasal sinuses, although the mechanisms
of 2 to 6 mm in the sagittal plane. Note that during childhood, until by which this occurs are unknown (122). Slowly, over the next 3 to
puberty begins, the anterior lobe of the pituitary is typically small. 4 years, these foci of short T1 relaxation time enlarge and coalesce
Although the normal size of the developing pituitary stalk (infundibu- (Fig. 2-19C–E). By 10 years of age, the marrow in almost all children
lum) has never been determined, the size of the mature infundibulum is almost entirely of high signal intensity on the T1-weighted images
is established and this number can be used in children. The infundibu- (Fig. 2-19F). A few small foci of low signal intensity may remain well
lum should be no more than 2.6 mm in its thickest portion on coro- into the second decade.
nal and sagittal MR images; greater thickness is highly suspicious for After intravenous administration of paramagnetic contrast, the
pathologic infiltration (see the section on “Suprasellar Tumors” in immature skull and skull base show variable enhancement in infants
Chapter 7) (117,118). The size of the stalk relative to the size of the and young children (123). Most likely, the enhancement is the result
brain never changes, and, as a general rule, it should never be as large of the vascularity of the hematopoietic marrow in the calvarial diploë
as the basilar artery on axial images (119). and, particularly, the skull base of infants. The amount of enhance-
With the arrival of puberty, the pituitary gland increases dramati- ment is age-related, as the immature homogeneous marrow of younger
cally in size and (in girls, but not boys) demonstrates a marked upward children enhances more and to a greater degree than the heterogeneous
convexity (Fig. 2-18). Of note, the gland is homogeneous on all imag- marrow of older children (123). The amount of enhancement seen in
ing sequences, both prior to and after contrast administration; this aids older children after fat suppression pulses have been applied is not
in differentiation from an adenoma and a Rathke cleft cyst. The height known. The normal enhancement of bone marrow in infants can be
of the gland may reach 10 mm in girls and 7 to 8 mm in boys (120,121). problematic in patients with diseases such as leukemia and neuroblas-
The appearance then slowly evolves into that of the adult gland over toma; differentiation of normal hematopoietic marrow from meta-
the subsequent 5 to 8 years. static neuroblastoma may not be possible by MR alone unless the bone
is expanded or periosteum is lifted from the surface of the bone. Cor-
relation with CT and radionuclide studies is essential.
NORMAL DEVELOPMENT OF THE SKULL Prior to pneumatization of the sphenoid sinus, an area of mark-
AND PARANASAL SINUSES edly increased signal intensity appears in the presphenoid portion of
the sphenoid bone (Fig. 2-19C). This high signal, believed to represent
The Skull
fatty change, appears between 7 months and 2 years of age, and then
At birth, the T1 signal intensity of the bone marrow in the clivus and regresses as pneumatization of the sphenoid sinus occurs after the age
in the diploic space of the calvarium is of low signal intensity with of 2 years (Fig. 2-19D–G). Some of the high signal may persist into the
respect to brain and to the sphenooccipital synchondrosis (Fig. 2-19A). second decade (124).

Barkovich_Chap02.indd 47 5/7/2011 2:42:22 AM


48 Pediatric Neuroimaging

FIG. 2-18. Menarchal pituitary gland. As seen on these contrast-enhanced T1-weighted sagittal (A) and coronal (B) images, children entering puberty have
enlarged pituitaries, typically larger in girls than in boys.

The maturation of the diploic space of the calvarium occurs at areas of elastic, pliable connective tissue allow the calvarium to mold
almost the identical time as the clivus. Foci of high signal intensity during the process of birth and also allow the head to grow rapidly dur-
on T1-weighted images begin to appear within the diploic space at ing the first 2 years after birth. These sutures and fontanelles are well
approximately age 3 years. More and more areas of high signal intensity described in most standard pediatric texts and will not be discussed
develop over the next 4 years until, at about 7 years of age, the diploic at length here. Suffice it to say that the neonate has six major sutures
space is of nearly uniform high signal intensity (125). Enhancement of in the cranial vault that may be involved in pathologic conditions:
the diploic space parallels that of the clivus. one midline metopic suture between the paired frontal bones, derived
On MR, the sphenooccipital synchondrosis remains prominent from metopon (Greek for forehead); two coronal sutures between the
throughout childhood and can be seen well into the third decade of frontal and parietal bones bilaterally, from corona (Latin for crown
life. This persistence of the synchondrosis on MR is in marked contrast or garland); two lambdoid sutures between the parietal and occipital
to the appearance on CT. On CT, the sphenooccipital synchondrosis bones (named because of the resemblance to the Greek letter lambda);
appears widely patent until the age of 8 years, when ossification centers and one midline sagittal suture that separates the two parietal bones,
appear. In boys, the ossification center is single and appears in the mid- derived from sagitta (Latin for arrow) (128). In addition, one major
line, whereas in girls, bilateral paramedian ossification centers appear. fontanelle is present in the neonate: the anterior (at the junction of the
In the study of Okamoto et al. (126), fusion of the basisphenoid and sagittal, metopic, and two coronal sutures). Five smaller fontanelles, the
basiocciput appeared complete by CT at age 13 years; no sphenooccipi- paired anterolateral fontanelles at the junction of the squamosal and
tal synchondrosis was seen beyond that age. Madeline and Elster found coronal sutures, the (single) posterior fontanelle (at the junction of the
complete fusion of the sphenooccipital synchondrosis in 95% of girls lambdoid sutures and the sagittal suture), and the paired posterolateral
by age 16 years and in 95% of boys by age 18 years (127). fontanelles at the junction of the mendosal and lambdoid sutures, are
The importance of understanding the normal temporal sequence important mainly as potential acoustic windows for sonography.
of skull maturation lies in the evaluation of the patient with systemic The normal times of closure of the sutures and fontanelles is of
disease. For example, several disease entities will cause reactivation of some importance, as premature closure often requires surgical cor-
erythropoiesis within the bone marrow. In particular, states of chronic rection or may be a sign of and underlying syndrome or metabolic
anemia, such as sickle cell disease and thalassemia, will cause a rever- disorder (see section “Craniosynostosis”, Chapter 5). The fontanelles
sion of bone marrow to the immature low signal intensity. Other close first, with the posterior closing by 8 weeks, the anterolateral by
disease processes, such as leukemia, can infiltrate the bone marrow; 3 months, the anterior by 15 to 18 months, and the posterolateral by
such infiltration will also cause a lengthening of the T1 relaxation time 24 months. The first major suture to close is the metopic, which starts
resulting from replacement of fat by a proliferative marrow. If low sig- to close as early as 3 months and is completely closed by 9 months,
nal intensity is seen diffusely throughout the clivus beyond the age of as assessed by 3D CT reformations (129). The precise time of closure
4 years, some underlying condition such as an anemia or a systemic, of the other sutures has not been established; these sutures don’t fully
infiltrative disease should be sought. close until adulthood (128). However, as a general rule, no part of the
sagittal, coronal, or lambdoid sutures should appear closed within the
first year of life.
The Cranial Fontanelles and Sutures
In the newborn, the calvarium is composed of many small bones that
Maturation of the Paranasal Sinuses
are separated by linear strips of connective tissue (sutures) and carti-
lage (synchondroses). At several areas, broader, more irregular patches The maxillary sinus is the first of the paranasal sinuses to develop. The
of connective tissue, known as fontanelles, separate the bones. These sinuses are rudimentary at birth, lying entirely medial to the orbit.

Barkovich_Chap02.indd 48 5/7/2011 2:42:23 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 49

FIG. 2-19. Development of normal bone marrow in the clivus as


seen on T1-weighted images. A. Normal clivus of a 3-month-old.
The marrow within the basiocciput and basisphenoid (white arrows)
is of uniform low signal intensity as compared to the sphenooccipi-
tal synchondrosis. The presphenoid region of the sphenoid bone
is isointense to the basisphenoid. B. Normal 12-month-old infant.
During the second year of life, the crista galli (closed arrow) and
the nasal process of the frontal bone (open arrow) develop high sig-
nal intensity within their marrow. C. Normal 18-month-old infant.
Between the age of 6 months and 2 years, fatty change occurs within
the presphenoid portion of the sphenoid bone. The basisphenoid
and basiocciput remain of uniform low intensity. D. Normal 4-year-
old child. The fatty change in the presphenoid region has begun to
recede as pneumatization occurs. There are patches of high signal
intensity within the basisphenoid and basiocciput as normal yellow
marrow begins to appear. E. Normal 5-year-old child. The regions
of high signal intensity within the basisphenoid and basiocciput are
becoming more apparent and beginning to coalesce.

Barkovich_Chap02.indd 49 5/7/2011 2:42:23 AM


50 Pediatric Neuroimaging

FIG. 2-19. (Continued) F. Normal 10-year-old. The sphenoid sinus is nearly completely pneumatized at this age. Only a small amount of fatty
change remains in the most inferior portion of the presphenoid (arrow). The basisphenoid and basiocciput are now composed of mainly yellow
marrow; the clivus therefore shows mainly a high signal intensity compared to the synchondrosis with a few patchy areas of low signal intensity
persisting. G. Mature clivus in a normal 16-year-old. The basiocciput and basisphenoid are now of uniform high signal intensity.

They are commonly partially or completely opacified. The sinuses the same prepneumatization transition as the sphenoid sinuses in that
grow rapidly through childhood, with a growth rate of 2 mm/y in yellow, fatty marrow develops prior to pneumatization (131). Earliest
the vertical dimension and 3 mm/y in the anteroposterior dimension pneumatization is seen around the age of 2 years in the orbital pro-
(130). Growth continues until the end of puberty, when facial growth cesses of the frontal bone. By age 4 years, pneumatization reaches the
ceases. The following milestones are useful: (a) Lateral margin of the nasion, advancing to the level of the orbital roof by age 8 years and into
sinus projects under the medial orbital wall by the end of the first year. the vertical portion of the frontal bone by age 10 years. Growth contin-
(b) Sinus extends laterally past the infraorbital canal by age 4 years. ues until the end of puberty. The extent of frontal sinus development is
(c) Sinus reaches maxillary bone and plane of hard palate by age extremely variable (131).
9 years (131).
At birth, the ethmoid sinuses are developed more anteriorly than
posteriorly. Lack of aeration of the posterior ethmoid sinuses is consid-
NORMAL DEVELOPMENT OF BRAIN IRON
ered normal until the age of approximately 6 years (130). Pneumatiza- In the adult, certain regions of the brain show a marked diminution in
tion progresses posteriorly, with resultant enlargement of the posterior signal intensity on T2-weighted spin-echo and gradient-echo images,
air cells. By the late phases of pneumatization, the posterior cells are particularly at high field strength; the diminution in signal intensity is
larger and fewer than the anterior cells. The last phase of ethmoid less apparent on fast-spin echo images. This diminished T2 relaxation
pneumatization involves development of the extramural air cells, the time has been suggested to result from the presence of relatively high
agger nasi, Haller cells, and concha bullosa, which lie in the anterior quantities of iron in those locations (132), although the cause and effect
ethmoidal region (131). relationship is disputed (133). The most prominent areas in which this
The fetal sphenoid sinuses consist of small cavities (conchal phenomenon occurs are the basal ganglia, particularly the globus pal-
sinuses) within the developing sphenoid bones. Soon after birth, the lidus, the substantia nigra (mostly in the medial pars reticularis), the
conchal sinuses fuse to the sphenoid bone and begin to pneumatize red nuclei, and the dentate nuclei of the cerebellum. Please note that
(130). High resolution CT may show pneumatization of the sphenoid this work was done at 1.5 T, and that the iron deposition will become
sinuses as early as age 2 years; pneumatization is preceded by conver- apparent sooner if MR is performed at 3 T.
sion of red marrow to fatty marrow, as described earlier in this sec- At birth, this accentuated T2 shortening is not seen anywhere in
tion in the discussion of maturation of the skull (124). Pneumatization the brain (134) (Fig. 2-20). The basal ganglia begin to manifest low
proceeds inferiorly, posteriorly, and laterally, with the presphenoid intensity with respect to the cerebral cortex on T2-weighted images at
portion developing first (usually between the ages of 5 and 10 years approximately age 6 months. However, the globus pallidus and puta-
[122]), followed by the basisphenoid (typically after age 10 years [122]) men at this age are isointense to one another and hypointense with
and, in some patients, greater sphenoid wings and pterygoid processes. respect to the internal capsule. This initial decrease in signal intensity
As noted in the previous section, care must be taken not to mistake on T2-weighted images is, therefore, believed to be secondary to myeli-
the normal fatty changes that occur prior to pneumatization (124) for nation of the axons within the lentiform nuclei. Indeed, as the cerebral
hemorrhage, proteinaceous fluid, or inclusion tumor. white matter begins to myelinate, the basal ganglia become hyperin-
The frontal sinuses are the last of the paranasal sinuses to develop. tense with respect to the surrounding white matter on T2-weighted
They originate as extensions of the anterior ethmoidal air cells (130). images. A second phase of T2 shortening in the globus pallidus, sub-
At birth, they are normal bone containing red marrow. They follow stantia nigra, and red nuclei becomes noticeable at 9 or 10 years of age

Barkovich_Chap02.indd 50 5/7/2011 2:42:25 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 51

FIG. 2-20. Immature pattern of brain iron. A–C. In this normal 4-year-old, we see that the globus pallidus, red nucleus, pars reticularis of the substantia
nigra, and dentate nuclei of the cerebellum are all hyperintense as compared to normal white matter of the brain (arrows). This appearance usually changes
to isointensity with white matter before the age of 10 in the globus pallidus, substantia nigra, and red nucleus. The changes in the dentate nucleus occur
later and less consistently.

(134) (Fig. 2-21). At this age, the signal intensity of these areas becomes
equal to, and then less than, that of the surrounding white matter. The
NORMAL DEVELOPMENT OF THE SPINE
globus pallidus will be of lower signal intensity than surrounding brain Embryology
in 90% of patients by age 15 years (134) (Fig. 2-22). The signal intensity
At the end of the second gestational week, the normal human embryo
continues to diminish on T2-weighted images throughout the second
is a bilaminar structure comprised of a flat sheet of cells adjacent to the
decade and may continue at a slower rate throughout life.
amnion, termed the epiblast, and a second layer adjacent to the yolk sac,
The dentate nuclei of the cerebellum begin to show lower signal
known as the hypoblast. The hypoblast will eventually be replaced by
intensity at a slightly later age, with noticeable changes beginning to
the endoderm, which is believed to be a derivative of the epiblast. Soon
occur at about age 15 years (Fig. 2-21). This low signal intensity also
after, cells in the caudal midline of the embryo proliferate to form a
progresses slowly throughout life; however, there is less accumula-
primitive knot (also known as Hensen node, which defines the cephalic
tion of iron in the dentate nuclei than in the other iron-containing
end) and the primitive streak caudal to it. By about embryonic day 16,
regions of the brain. The dentate nuclei will be of lower signal inten-
the primitive streak begins to regress and cells at the rostral lip of the
sity than surrounding cerebellum in only 30% of patients at age
primitive knot migrate between the epiblast and hypoblast, forming the
25 years (134).

FIG. 2-21. Intermediate stage of maturation of brain iron. A–C. In this normal 13-year-old, the iron-concentrating regions of the brain are now isoin-
tense with white matter. Iron accumulates more rapidly in the globus pallidus than the other areas of the brain and the globus pallidus will be of lower
signal intensity than surrounding brain in 90% of patients by age 15. The substantia nigra and red nuclei are hypointense with respect to white matter in
50% of normal patients by age 20.

Barkovich_Chap02.indd 51 5/7/2011 2:42:25 AM


52 Pediatric Neuroimaging

FIG. 2-22. Brain iron in normal young adult.


A–C. By the age of 25, the globus pallidus, sub-
stantia nigra, and red nuclei will be hypointense
with respect to white matter in 80% of normal
patients. However, only 30% of normal 25-year-
old people will show hypointensity of the dentate
nuclei, which accumulate iron more slowly and
less consistently than the other areas.

notochordal process. The notochordal process elongates as cells from between days 40 and 60. Finally, ossification begins in four ossification
the primitive knot are added to it at its caudal end; after canalization, the centers (two in the vertebral body and one in each side of the vertebral
more caudal portion will be known as the notochord. The notochord arch), a process that continues into postnatal life (135,136).
induces surrounding mesoderm (the paraxial mesoderm, derived from The newborn vertebral body has a membranous central compo-
the primitive streak) to condense into paired blocks of somites. The nent, with ossification centers and endplates of hyaline cartilage that
somites eventually develop into myotomes, which will form the para- lie superior and inferior to the central component. The endplates are
spinous muscles and overlying skin, and sclerotomes, which will form each about one half the size of the central vertebral component (137).
the cartilage, bones, and ligaments of the vertebral column (135,136.) The vertebral body and cartilaginous endplates of the newborn have a
During the fourth or fifth gestational week, resegmentation of the more substantial vascular supply than those of the adult. The vertebral
sclerotomes occurs, with eventual development of the vertebral bodies. bodies contain hematopoietic marrow, with large vascular pools and
In this resegmentation, the cells within the caudal half of each sclero- sinusoidal channels that lack blood–brain barrier and contain large
tome separate from the cranial half (of the same sclerotome) at the extracellular spaces. The cartilaginous endplates are perfused by vessels
sclerotomal cleft and then fuse with the cranial half of the inferior scle- from the margins of the vertebral bodies and by vessels branching from
rotome (Fig. 2-23) to form a new, primitive vertebral body (135,136). the lumbar arteries (138,139).
As a result, the intersegmental arteries become trapped in the centers of
the new vertebral bodies. At the same time, the segments of notochord
CT of Spine Development
within the newly formed vertebrae disintegrate and the notochordal
segments in the intervertebral regions proliferate and convert into In the spine, the major developmental process that is demonstrated by
nucleus pulposis. The mesenchymal vertebrae then undergo chondrifi- CT is the ossification of the synchondroses between ossification cen-
cation, starting at specific chondrification centers within the vertebrae, ters. The persistence of these synchondroses in infants and children

Barkovich_Chap02.indd 52 5/7/2011 2:42:27 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 53

FIG. 2-23. Diagram of formation of vertebral bodies. A. Each sclerotome consists of a cranial mass of loosely packed cells (anterior sclero-
tome, A, dotted areas) and a caudal mass of densely packed cells (posterior sclerotome, P, vertical lines). The sclerotomes are closely related
to the notochord (N) and the myotomes (M, cross-hatched areas). B. The caudal, dense cell mass of one sclerotome unites with the cranial
loose cell mass of the adjacent caudal sclerotome to form a structure (the centrum of the vertebral body) that contains equal parts of adjacent
sclerotomes. The notochord has regressed; its remnants (N) will become nuclei pulposi. Myotomes are evolving into paraspinous muscles
(M). Note relationship of intersegmental arteries (A) to the vertebrae and sclerotomal segments.

can create problems in the interpretation of CT scans. Therefore, it is on axial images as oblique low intensity lines. The lines are seen from
important to recognize their normal sequence of closure (Table 2-4). infancy and presumably represent dense cortical bone plus cartilage.
Except for C-2, all of the vertebrae develop from three primary ossifica- The synchondroses disappear from MR at a different time than they do
tion sites: the body (anteromedial) and the two neural arches (postero- on CT and plain films, suggesting that MR and CT visualize different
lateral). The neural arches are separated from the body by neurocentral aspects of the process. On MR, the lines begin to disappear laterally,
synchondroses (140). At C-1, the anteromedial ossification center is the at a point about midway between the superior and inferior endplates
anterior arch. C-2 differs from the other vertebrae in that it has four (143,144). In the lumbar spine, the synchondroses appear approxi-
ossification centers: the body, the two neural arches, and the dens. The mately 75% closed at age 4 years. Complete disappearance of the lines
dens forms in utero from two separate paramedian ossification centers begins in the mid- and lower cervical region at about age 6 years. In the
that fuse in the midline, usually by the seventh fetal month; the syn- lumbar region, the lines disappear between the ages of 10 and 14 years,
chondrosis between the two ossifications centers may persist in infancy while in the thoracic region they disappear between the ages of 10 years
and should not be mistaken for a fracture (141). (upper thoracic) and 15 to 16 years (midthoracic) (140,143,144).
At birth, nonossified synchondroses are present within the ante-
rior arch of C-1, within the posterior arch of C-1, and between the two
arches. The anterior arch synchondrosis typically ossifies between the TABLE 2-4 Closure of Synchondroses of
ages of 8 and 12 months, the posterior arch synchondrosis between the
ages of 1 and 7 years, and the synchondrosis between the two arches the Cervical Spine (Assessed
between the ages of 7 and 9 years (142). At the C-2 level, the synchon- by CT)
drosis between the two posterior ossification centers ossifies between
the ages of 4 and 7 years. The center between the C-2 vertebral body and Synchondrosis Age at Ossification
the base of the odontoid ossifies between 3 and 7 years. The superior
odontoid ossification center appears on CT between the ages of 2 and 6 Anterior arch of C-1 8–12 mo
years and fuses with the odontoid between the ages of 11 and 12 years. Posterior arch of C-1 1–7 y (usually by 4 y)
Below the level of C-2, the vertebral bodies consist of one anterior ossi-
Lateral masses of C-1 7–9 y
fication center and two posterolateral ossification centers (Fig. 2-24). At
C-3 and below, the ossification of the synchondroses is fairly constant. C-2 body-odontoid 3–7 y
The posterior synchondrosis ossifies between the ages of 4 and 7 years, Superior odontoid center
and the synchondrosis between the vertebral body and the posterior
Appears 2–6 y
ossification center ossifies between the ages of 3 and 7 years (142).
Fuses 11–12 y
MRI of Spine Development Posterior C-2 synchondrosis 4–7 y
The neural synchondroses (between the neural arches and the verte- Below C-2 level
bral body) can be assessed by MR as well as by CT. These synchon- Posterior synchondrosis 4–7 y
droses can be seen on both T1- and T2-weighted images as linear
bands of low signal intensity. They can be assessed on sagittal and axial Vertebral body-posterior 3–7 y
images (Fig. 2-25) appearing on the sagittal images as low-intensity ossification center
lines running from the superior endplate to the inferior endplate and

Barkovich_Chap02.indd 53 5/7/2011 2:42:28 AM


54 Pediatric Neuroimaging

osseous portion. T2-weighted images show the vertebral body ossifica-


tion centers to be hypointense centrally and slightly more hyperintense
peripherally (Fig.2-26B). Administration of paramagnetic contrast at
this age will result in mild to marked enhancement of both the verte-
bral body and the cartilaginous endplates. The intervertebral disc does
not change significantly during childhood, appearing hypointense on
T1-weighted images, hyperintense on T2-weighted images, and show-
ing minimal to no enhancement (145,146).
Stage II (Fig. 2-27A–E), taking place from approximately 1 to
6 months, is characterized by T1 shortening in the vertebral bodies that
starts at the superior and inferior borders of the body and moves cen-
trally, eventually affecting the entire vertebral body. Thus, the upper
and lower portions of the vertebral body are more hyperintense than
the central portion (Fig. 2-27A). A band of hypointensity (the exact
correlate of which is not known) separates outer portions of the ver-
tebral body from the cartilagenous endplate. The intervertebral disc
is hypointense. Note that the cartilagenous synchondrosis between
the body of C-2 and the dens, the cartilagenous apical dens, and the
sphenooccipital synchondrosis (in the clivus) are all hyperintense on
T1-weighted images during this stage (Fig. 2-27E). T2-weighted images
also show increasing intensity in the superior and inferior portions of
the vertebrae; the vertebral body slowly becomes isointense with the
endplates by about age 3 months. The vertebral endplates are mildly
hypointense compared to the vertebral body, while the intervertebral
disc is very hyperintense. Note that different techniques of T2 and T2*
FIG. 2-24. CT of immature vertebral body shows the anterior ossification weighting will result in a slightly different appearance of the immature
center (A) and the two posterolateral ossification centers (L). Small arrows spine (Fig. 2-27B–D). The enhancement pattern of the vertebral body
show the anterior synchondroses and the large arrow marks the posterior and endplate at Stage II is not significantly different than that at Stage I;
synchondrosis. the vertebral body uniformly enhances (Fig. 2-27C) (145,146).
Stage III (Fig. 2-27F–H) takes place from about age 7 months
The MR appearance of the vertebral bodies and intervertebral discs onward. At this stage, the vertebral bodies are becoming hyperintense
of the infant spinal column has been described as evolving through with respect to the cartilaginous endplates and surrounding muscle on
three stages (145). Stage I, occurring from birth through the first month T1-weighted images. The cartilaginous endplates gradually become
of life, is characterized by biconvex-appearing vertebral bodies that are ossified and are incorporated into the vertebral body, which becomes
markedly hypointense centrally on T1-weighted images; this likely rep- rectangular in shape by about age 2 years. T2-weighted images
resents the vertebral body ossification center (Fig. 2-26A). The carti- (Fig. 2-27G) show the vertebral bodies to be homogeneous, isointense
laginous endplates of the vertebral bodies, which are about one half the with the cartilaginous endplates, and mildly hyperintense compared
size of the osseous body at this age, are mildly hyperintense compared with surrounding muscle. Uniform contrast enhancement of variable
with muscle, but significantly hyperintense compared with the central intensity (Fig. 2-27H) is seen in the endplates and vertebral bodies in

FIG 2-25. Appearance of immature vertebrae


on MR. Parasagittal T2-weighted image (A)
and axial SE T1-weighted image (B) show the
synchondroses (arrows) between the anterior
(A) and posterolateral (L) ossification centers
as linear bands of low signal intensity.

Barkovich_Chap02.indd 54 5/7/2011 2:42:29 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 55

FIG. 2-26. Neonatal spine, Stage I. A. T1-weighted image shows


horizontal bands of high intensity (small white arrows) located within
the centers of the vertebral bodies, probably representing the basiver-
tebral venous plexuses. The cartilagenous endplates of the vertebral
bodies (larger white arrows), which are about one half the size of the
osseous body at this age, are mildly hyperintense compared with mus-
cle, but markedly hyperintense compared with the hemiovoid-shaped
central osseous portion. B. T2-weighted image shows the ossification
centers to be markedly hypointense and the endplates (black arrows)
mildly hyperintense compared with muscle. The intervertebral discs are
markedly hyperintense.

FIG. 2-27. Spine at age 3 months (Stage II) and


9 months (Stage III). A. Fat suppressed sagittal
T1-weighted image shows horizontal bands of low
intensity (small white arrow) located within the cen-
ters of the vertebral bodies, probably representing
the residual vertebral body ossification center. The
periphery of the vertebral body shows alternating
hyperintensity (large black arrows) bordered by a
curvilinear band of hypointensity. Just peripheral to
the thin curvilinear hypointensity is a slightly thicker
curvilinear hyperintense band (white arrowheads) that
represents the cartilagenous endplate of the vertebral
bodies (large white arrowheads); these are markedly
hyperintense compared with the central osseous por-
tion of the vertebrae. The intervertebral disc (small
black arrow) is hypointense. B. Sagittal T2-weighted
image shows vertebral body ossification center to be
hypointense (small black arrow). This is surrounded
by a thin rim of hyperintensity relatively hyperin-
tense (large black arrow) and the markedly hypoin-
tense cartilagenous endplate (small white arrows). The
intervertebral disc (large white arrow) is markedly
hyperintense. C. Sagittal STIR image shows findings
similar to the T2-weighted image except that the ver-
tebral body ossification center is less well differenti-
ated from the surrounding cartilagenous endplate and
the intermediate layer is poorly seen. D. Sagittal T2*-
weighted gradient-echo image nicely differentiates the
hypointense vertebral body ossification center (large
black arrow) from the more hyperintense surround-
ing peripheral vertebral body (black arrowheads).
However, the hyperintense cartilagenous endplate
and intervertebral disc are both hyperintense (white
arrows) and cannot be differentiated.

Barkovich_Chap02.indd 55 5/7/2011 2:42:30 AM


56 Pediatric Neuroimaging

FIG. 2-27. (Continued) E. Sagittal T1-weighted image of the


craniocervical junction shows that the synchondrosis between
the body of C-2 and the dens (white arrowhead), the cartilag-
enous apical dens (large white arrow), and the sphenooccipital
synchondrosis (small white arrows) are all hyperintense com-
pared to bone at this stage. F. Sagittal noncontrast image at age
9 months. The vertebral bodies now appear nearly homogeneous
and the cartilagenous endplates are no longer hyperintense. The
intervertebral disc is hyperintense compared with the vertebral
bodies. G. T2-weighted image shows a homogenous vertebral
body; the superior and inferior cartilagenous endplates remain
hypointense, while the intervertebral disc remains hyperin-
tense. H. Postcontrast sagittal T1-weighted image shows nearly
uniform enhancement of the vertebral bodies.

most children up to the age of 9 or 10 years, during which time the rates, depending of their chemical surroundings (T2 relaxation). As
hematopoietic marrow is being slowly replaced (145,146). they lose coherence, their respective MRS peaks broaden and their
amplitude is decreased. In general, short TE spectra are used to assess
patients with suspected metabolic disorders where one needs to analyze
EVALUATION OF BRAIN DEVELOPMENT as many peaks as possible and look for the appearance of new, unex-
USING PHYSIOLOGIC MR TECHNIQUES pected ones. Long TE spectra generally detect peaks from only four
MR Spectroscopy compounds (N-acetylaspartate [NAA], choline, creatine, and lactate).
However, they are easier to quantify and are used in our institution
The Normal MR Spectrum to assess patients with brain injury (147–150) and brain tumors (151).
The evaluation of the developing brain with MR spectroscopy is still at Another important concept is that each area of brain has slightly
an early stage of development. Before dealing with specifics of changes different concentrations of metabolites. For example, the cerebellum
in spectra during development, two important concepts need to be dis- has a different spectrum than the brainstem, the basal ganglia have dif-
cussed. The first is that the observed spectra vary with the echo time ferent spectra than the cerebral white matter, and the thalamus has a dif-
(TE) used in acquiring them. As discussed in Chapter 1, many more ferent spectrum than the putamen. Beyond that, each thalamic nucleus
peaks are seen on short (20–30 milliseconds) TE than on intermedi- has a slightly different spectrum. Furthermore, the spectra change with
ate (135–144 milliseconds) or long (270–288 milliseconds) TE spectra. maturation. Infants have high choline and myo-inositol and relatively
The reason for this is that excited protons lose coherence at variable low creatine and NAA peaks; as they grow the choline and myo-inositol

Barkovich_Chap02.indd 56 5/7/2011 2:42:31 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 57

peaks shrink, while the NAA and creatine peaks grow. Therefore, it will exclusively in neurons and axons, and because glia are not affected in
be necessary to establish norms based upon small voxel spectra in every many neurodegenerative processes, NAA is reduced in most neurode-
part of the brain in very large numbers of completely normal infants generative processes (163). Animal experiments have confirmed that
in order to quantitatively establish whether spectra are normal in every reduced NAA correlates with neuronal necrosis (164). Thus, an abso-
different part of the brain. This has been accomplished in small studies lute or relative (to Cr) decrease of this peak is generally considered to
(152), but a study of sufficient size to establish definitive norms will be an indicator of neuronal and/or axonal damage (165–167). How-
require many years. In the mean time, it suffices to look at patterns of ever, because NAA is synthesized in the mitochondria, energy deple-
peak heights and peak ratios in different parts of the brain to deter- tion, without permanent neuronal damage, can also result in transient
mine whether gross abnormalities are present. Before discussing these reduction of NAA. Indeed, recovery of NAA has been seen in mito-
patterns, let us first discuss the different peaks seen on proton (1H) MR chondrial disorders (168), epilepsy (169), and treatment of AIDS with
spectra (Fig. 2-28), the origin of their signals, and their significance. antiviral agents (170). NAA is broken down to acetate and aspartate in
After understanding the origin of the signals, the major changes that oligodendrocytes, where the acetate is subsequently used in the syn-
occur during development will be discussed. thesis of myelin lipids (155). It is converted to NAAG in presynaptic
terminals; NAAG is then released into the synapse, where it may block
Metabolites Seen in MR Spectra of the Brain
presynaptic calcium channels, thus inhibiting glutamate (Glu) release
Peaks Seen on Long Echo Time (Long TE) Spectra (155). In the infant, concentrations of NAA in the gray and white mat-
NAA is the most obvious peak on 1H MRS and is the peak (at 2.01 parts ter are similar. As NAA breakdown products may be used for myelino-
per million [ppm]) that is used as a reference for chemical shift deter- genesis, the relatively high NAA concentrations of immature white
mination (153,154). The peak actually includes contributions from matter have been attributed to very active membrane synthesis (156);
NAA, N-acetylaspartylglutamate (NAAG), glycoproteins, and amino- work in the immature brain has shown that oligodendroglia precursors
acid residues in peptides (153,154); therefore, the peak is, perhaps, contain twice as much NAA as immature neurons (171). As a conse-
more appropriately termed “N-acetyl groups.” NAA is synthesized in quence, NAA levels have potential as an indicator of normal oligoden-
the mitochondria of neurons and present in very high concentrations droglial development.
in the CNS. NAA is a marker of the functional integrity of neuronal The choline (sometimes called trimethylamine) peak at 3.21 ppm
mitochondrial metabolism and may be critical for myelinogenesis, is composed of contributions of the trimethylammonium (−N(CH3)3+)
whereas NAAG modulates glutamatergic transmission and may have protons in choline, betaine, and carnitine plus the H5 proton of myo-
a role in neuroprotection and synaptic plasticity (155). NAA has been inositol and taurine (153,154). The “choline” contribution is a sum
postulated to have at least two functions in the adult brain: (a) it is a signal from several choline-containing compounds (i.e., phosphoryl-
precursor of brain lipids and (b) it is involved in coenzyme A inter- choline, phosphorylethanolamine, glycerophosphorylcholine, glycero-
actions (155,156). Others have suggested that NAA is an osmolyte phosphorylethanolamine, and free choline), possibly in conjunction
(a metabolically inert compound that functions only to protect neural with the choline that is present as a polar head group in membrane
tissues during metabolic disturbances of extracellular osmotic pressure lipids. In the neonate, phosphorylethanolamine dominates the spec-
[157]), that it is a neurotransmitter/neuromodulator precursor, and trum; it decreases with age along with phosphorylcholine (172); both
that it functions as a free storage form for aspartate (158,159). In adult of these compounds are precursors for membrane synthesis (173).
brains, NAA concentrations are higher in the cortex than in the white Glycerophosphorylethanolamine and glycerophosphorylcholine are
matter (160), as most NAA is located in neurons and their branches. products of membrane breakdown; they increase in concentration
Acetyl-CoA-L-aspartate-N-acetyltransferase, the synthetic enzyme with postnatal age (172). Membrane-bound choline compounds may
for making NAA, is localized in mitochondria, where NAA is synthe- not be visible by MRI (174); however, when the cell membranes are
sized from aspartate and acetylcoenzyme A (161). Acetoaspartase, the broken down in disease processes, the bound choline is released and
enzyme that degrades NAA, is located primarily in astrocytes (162); becomes visible (174). The choline reflects the structural components
thus, NAA breakdown occurs primarily in glial cells, explaining the low of cell membranes, especially myelin sheaths (153,154). Thus, the cho-
concentration of NAA in mature glia. Because NAA is located almost line peak tends to be enlarged in highly cellular processes such as high

FIG. 2-28. 1H spectral peak identification (short TE). 1. CH3 of lac-


tate. 2. CH3 of NAA and NAAG, C-2 CH2 of Glu and Gln, C-3 CH2 of
GABA. 3. C-4 CH2 of Glu and C-2 CH2 of GABA. 4. C-4 CH2 of Gln
and C-3 CH2 of NAA. 5,6. C-3 CH2’s of NAA. 7. CH3 of creatine and
C-4 CH2 of GABA. 8. CH3 of cholines. 9. C-1 CH2 of taurine. 10. C-1
and C-3 CH of inositol and C-2 CH2 of glycine. 11. C-2 CH of Glu and
Gln, C-4 and C-6 CH of inositol. 12. C-2 CH2 of creatine. 13. CH2’s of
cholines. 14. C-2 CH of lactate.

Barkovich_Chap02.indd 57 5/7/2011 2:42:32 AM


58 Pediatric Neuroimaging

grade tumors and in neurodegenerative disorders. Focal inflamma- amounts in AGA (appropriate for gestational age) term neonates (see
tion, which results in marked local cellularity and, often, in significant Chapter 3). More than a trace amount, particularly after the first few
breakdown of cell membranes, can also result in large choline peaks. hours of life, is generally considered to be indicative of some degree of
In the mature brain, choline concentration is higher in cerebral white brain injury (26). However, lactate, is a normal finding on 1H MRS in
matter than in cerebral cortex and higher in the thalami and cerebel- the cerebrospinal fluid of premature and term neonates, with a con-
lum than in the cerebral cortex or cerebral white matter. centration up to 2.7 mm/L (26,180–182). This lactate disappears by the
The creatine peak at 3.03 ppm is from methyl (CH3) protons of time the baby is a few months old. The normal presence of the lactate
creatine and phosphocreatine plus minor contributions from gamma- in the CSF is important, because one may falsely diagnose the presence
aminobutyrate, lysine, and glutathione (153,154). A second, smaller of anaerobic glycolysis, and hence ischemia, in the parenchyma if the
creatine peak is seen at 3.94 ppm (175,176). Phosphocreatine appears spectroscopy voxel includes a large third ventricle or a large portion
to be a crucial molecule in maintenance of energy-dependent systems of the lateral ventricle. Exclusion of CSF is, therefore, very important in
in all brain cells (177). Its concentration is highest in the cerebellum, analyzing neonatal proton MR spectra in the presence of encephalopathy.
followed by the thalami, basal ganglia, cortical gray matter, and white It is important to realize that propane-1,2-diol, an injection sol-
matter (178). The overall level of Cr is usually considered to remain vent for the administration of anticonvulsants given to neonates, has
stable in most situations; therefore, Cr is used as a standard for com- a spectroscopic appearance nearly identical to lactate (183). Propane-
parison with other metabolites. In rats, Cr concentrations are higher 1,2-diol appears as a doublet centered at approximately 1.1 ppm
in astrocytes than in neurons; therefore, Cr may be elevated in injured (Fig. 2-29) (183). Therefore, if a doublet is seen in the aliphatic region
tissue (179). (about 1–1.4 ppm) in an encephalopathic neonate, the precise chemical
Lactate, detected by a characteristic doublet centered at 1.3 ppm shift of the doublet should be determined before assuming the presence
(the peaks are at 1.27 and 1.36 ppm) on 1H MRS, can be seen in trace of lactate.

FIG. 2-29. Presence of propane-1,2-diol in neona-


tal spectrum. (A) is a proton spectrum (TE = 270
milliseconds) from the basal ganglia. (B) is a proton
spectrum from the frontal watershed white matter.
Note that two doublets are present, the one cen-
tered at 1.1 ppm (p) representing propan-1,2-diol
and the one centered at 1.3 ppm (la) representing
lactate.

Barkovich_Chap02.indd 58 5/7/2011 2:42:32 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 59

Peaks Seen on Short Echo Time (Short TE) Spectra from all sorts of aliphatic hydrocarbons (lipids) and amino acids.
When proton spectra are acquired using a short TE, many additional These peaks gradually diminish in size over the first 12 to 18 months
peaks may be identified (these peaks disappear at long echo times after birth and are usually of no clinical importance unless the peak
because of their relatively short T2-relaxation times). Several small heights approach about half that of NAA; when large, the peaks may
peaks that correspond to protons from glutamine (Gln) and Glu are indicate cellular breakdown rather than production and, thus, a degen-
seen in the 2.1 to 2.4 ppm region (153,177). A second peak, at 3.75 erative process. These peaks are seen more commonly in inborn errors
ppm, can be seen and is from resonance of the alpha CH moiety. Glu is of metabolism and in child abuse than in the “normal” infant popula-
an excitatory neurotransmitter that is released from excited axons into tion (Dr. William Ball, personal communication).
the synapse, where it binds to postsynaptic Glu receptors. Some of the Most commercially available MR spectroscopy programs do not
released Glu is then transported into adjacent glial processes, while the allow phosphorus MRS to be performed. However, some knowledge
rest is taken up into presynaptic and postsynaptic neuronal processes. of 31P MRS is useful. Presently, the progressive decrease of phosph-
The Glu absorbed into glial cells is converted into Gln by the enzyme omonoester and the complementary increase of phosphodiester,
Gln synthetase or metabolized to oxoglutarate (by Glu dehydrogenase) caused by lipid metabolism, seem to be the best indicators of brain
and aspartate (by aspartate aminotransferase) (184). Glu is an impor- development on 31P MRS (194). Spectra from preterm infants show
tant molecule in brain injury, from hypoxia-ischemia, seizures, and increased resonances from phosphomonoesters and comparatively
possibly trauma (185). Unfortunately, at clinically used magnetic-field lower signals from phosphodiesters compared with full-term infants.
strengths, the peaks at 2.1 to 2.4 ppm overlap each other and cannot be Phosphodiesters arising from phospholipid degradation products
separated well from the NAA peak, gamma-aminobutyric acid (GABA) and phospholipids are found to increase further in infants and adults
peaks, and each other, while the peak at 3.75 ppm is difficult to separate but there is relatively little diminution in the signal intensity from
from the creatine peak at 3.9 ppm. It is possible to better separate and the phosphomonoesters (which are mainly phospholipid precur-
evaluate these peaks at higher static magnetic-field strengths, such as sors). However, changes in the chemical shift and spectral width of
3 T. As higher field strength scanners become used more frequently in the phosphomonoester signals are observed between the neonates and
clinical practices, it is possible that analysis of these peaks will become the adults, indicating that a change occurs in the composition of the
important in assessing excitotoxic brain injury, which is often mediated phosphomonoesters. This is likely the result of changing proportions
by Glu (186). Higher field strength may also allow better assessment of of the contributions of phosphorylethanolamine, phosphorylcholine
astroglial reaction to brain injury, as Gln is mainly seen in astrocytes, (195), glycerophosphorylethanolamine, and glycerophosphorylcho-
while Glu is synthesized in neurons. line (172). Additionally, the correlation of 31P MRS and neurologic
Myo-inositol, which has two peaks, at 3.56 and at 4.06 ppm, is exam (examination of reflexes, motor-and sensory functions) in new-
thought to be the storage pool for membrane phosphoinositides, which born dogs demonstrated that exponential increases in phosphocre-
are second messengers that are involved in many hormonal systems and atine, inorganic phosphate, and phosphodiesters, with maintenance
in enzyme regulation in the CNS (158,187). It is an essential growth of the phosphocreatine/inorganic phosphate ratio, preceded matura-
factor (188) and is the precursor of phosphatidylinositol, a constituent tional changes in the neurological examination (196). The spectro-
of phospholipid membranes (153). It is located primarily in glial cells, scopic evolution is postulated to result from increasing ATP turnover
with particularly high concentrations in astrocytes (173) and, there- in the maturing mitochondria.
fore, may be a specific marker for astrogliosis. Other possible roles
include osmoregulation, cell nutrition, and detoxification (158,187).
Variations of Spectra in Brain Regions
Minor contributions to the peak at 3.56 ppm come from glycine and Proton spectra vary with the location of the prescribed voxel in which
inositol-1-phosphate (174). Scyllo-inositol is a nonmetabolized isomer the spectrum is acquired. This should not be surprising, as the cyto-
of myo-inositol that may inhibit the transport and incorporation of architecture of the cerebral cortex differs considerably from one region
myo-inositol into phospholipids (189,190). Most experts believe that of the cortex to the other (197). The importance of this concept can-
the singlet peak at 3.35 ppm is from the six equivalent methine protons not be overstated. Spectra obtained from the mature frontal cortex
of the cyclic alcohol of scyllo-inositol, not from taurine as previously differ from those in the mature parietal cortex; both differ from the
believed, based upon the exact chemical shift, the singlet nature of the spectra of the mature hippocampus (198). Spectra obtained from the
resonance, agreement with biochemical concentration levels, a link to thalamus differ from those obtained from the striatum (199). It is not
myo-inositol levels, and exclusion of other metabolites in 1H MR spec- possible to show spectra from every location in the brain at every age.
tra of mammalian brain in vivo and in vitro (178,190). Cerebral glucose However, a few patterns can be described. The height of the NAA peak
may be detected on short TE proton MR spectra by a singlet at 3.43 varies in different regions of the brain. NAA/Cr values are lower in
ppm. The area under the singlet may be used as a gross measurement the brainstem and cerebellum than in the cerebral hemispheres and
of glucose concentration in the brain (191). are lower in the basal nuclei (thalami, caudates, putamina) than in
Taurine is an amino acid that is reported to act as an osmoregulator the cerebral white matter (Fig. 2-30). Within the cerebral white mat-
and as a modulator of the action of neurotransmitters (192). It is found ter, NAA/Cr is higher in the frontal than the parietal white matter. In
at high concentration in infants but the concentration drops with mat- addition, the ratios of choline and creatine vary, with choline usually
uration, to approximately 1.5 mmol in adults. The spectrum contains being larger than creatine in white matter and thalami, while creatine
two triplets centered at 3.25 and 3.42 ppm (192). Unfortunately, on is usually larger than choline in mature striatum and in cerebral cortex
most clinical scanners, these peaks overlap with the resonances from (198–203).
myo-inositol, scyllo-inositol, and choline, so they are difficult to see
unless markedly elevated, as sometimes seen in brain tumors (193). Changes in Spectra with Brain Maturation
Two broad peaks are typically seen in short echo spectra of infants, Changes in in vivo MR spectra with brain maturation vary with tech-
one between 0.5 and 1.0 ppm approximately and another between 1.0 nique. As discussed earlier, spectra acquired with short echo times
and 1.6 ppm approximately. They are commonly referred to as “macro- (Fig. 2-31) have a different appearance than those acquired with long
molecular peaks.” They are composed primarily of methylene (centered echo times (Fig. 2-32) because T2 relaxation and J-coupling cause
at about 1.3 ppm) and methyl (centered at about 0.9 ppm) protons broadening and decreased amplitude of peaks (187). As many more

Barkovich_Chap02.indd 59 5/7/2011 2:42:33 AM


60 Pediatric Neuroimaging

FIG. 2-30. Variation in proton spectra with location in the brain (TE = 288 milliseconds) in a mature (3-year-old) child. A. Spectrum of the cerebellar
cortex shows large choline (Ch) and creatine (Cr) peaks compared with NAA. B–F. Spectrum of the frontal white matter (B), frontal cortex (C), occipital
cortex (D), putamen (E), and thalamus (F) all show slightly different ratios of choline, creatine, and NAA.

peaks are identified, the maximum information is obtained by using during the first year of life (Fig. 2-31) (158,205). The scyllo-inositol
short echo times. However, a stable baseline is more easily obtained (190) peak is highest in newborns (158,206).
and peaks are more easily quantified using long echo times. Absolute concentrations of metabolites are very difficult to
Changes in MRS that reflect brain maturation include a rela- determine by in vivo MR spectroscopy (205,207,208). Therefore, the
tive decrease in the size of the phosphomonoester peak and relative time course of most metabolites has been expressed most exactly by
increases in the size of the phosphocreatine and phosphodiester peaks comparing the spectral peaks with each other and calculating ratios
on 31P spectra and increase in the size of the large NAA peak (at chemi- (158,159,167). Compared to Cr, for example, a decrease of choline (the
cal shift 2.01 ppm) relative to the choline (at 3.21 ppm) and the cre- Ch/Cr ratio) and myo-inositol (myo-Ins/Cr ratio) develops as matura-
atine-phosphocreatine peaks (at 3.03 ppm) on 1H spectra (Figs. 2-31 tion progresses (167); the choline/myo-inositol ratio is stable (158,159).
and 2-32) (187,202). This pattern of evolution is seen in fetuses (204) In addition, the temporal evolution of the peaks differs in gray matter
and premature infants (199,205), as well as term infants (199,205) and as compared to white matter. Whereas the Cr/Ch ratio stays nearly con-
young children (177,202). In addition, the large myo-inositol peak pres- stant in white matter, the ratio increases in gray matter during the first
ent at 3.56 ppm in the spectra of newborns diminishes substantially 2 years of life (167).

Barkovich_Chap02.indd 60 5/7/2011 2:42:34 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 61

FIG. 2-31. Changes in short echo time (26 milliseconds) 1H spectra with maturation. Basal ganglia spectra from a 5-day-old neonate (A),
a 3-month-old (B), a 14-month-old (C), and a 22-month-old (D). Myo-inositol and choline are large peaks in neonates. Choline and creatine remain large
in older infants. NAA is the dominant peaks in older infants, children, and adults. In older children and adults, choline is usually larger in white matter,
creatine in cortex. Broad peaks at 0.9 and 1.3 ppm (macromolecular peaks, arrows in A) are normal in infants on short TE spectra.

Quantification of proton MRS signals from specific compounds The thalami, basal ganglia, sensorimotor system, and visual system
is difficult because the signal intensity of each peak depends upon its mature earlier than the prefrontal and temporal association areas, for
concentration, spin saturation effects, and T2 relaxation times (209). example, and spectra of the more mature areas will give lower values of
All of these must be taken into account before any accurate data can be myo-inositol and higher values of NAA than less mature areas on pro-
constructed. Nonetheless, methods for absolute quantification of peak ton spectra (199,212,213). On phosphorus spectra, the more mature
areas have been developed and, because they are more reproducible areas will give lower values of phosphomonoester and higher values
and reliable than ratios of compounds, these methods are becoming of phosphodiester, phosphocreatine, and ATP (195). Using absolute
the standard way of analyzing spectra (177,187,193,205,210). However, measurements, myo-inositol has been found to be the dominant peak
different groups obtain slightly different values, even using these “abso- in neonates, with a concentration of 10 to 12 mmol/kg (177,211). Cho-
lute” measurements (177,211–213). One reason for these differences is line, with a concentration of 2.5 to 3.5 mmol/kg, is the dominant peak
that the spectra of different parts of the brain mature at different times in older infants (177,211). Creatine and N-acetyl groups, which have
and different rates (see Fig. 2-32A–D) (199,205). Thus, just as certain concentrations of 5 to 6 mmol/kg and 4 to 5 mmol/kg, respectively,
regions of the brain myelinate earlier than others (see earlier parts of in the neonate, increase in concentration to 7 to 10 mmol/kg and 9 to
this chapter), the same regions undergo earlier biochemical maturation. 10 mmol/kg, respectively in the adult; thus, they become the dominant

Barkovich_Chap02.indd 61 5/7/2011 2:42:36 AM


62 Pediatric Neuroimaging

FIG. 2-32. Changes in long echo time (288 milliseconds) 1H spectra with maturation. A. Normal 35-week neonate. Spectrum through the basal nuclei
shows a dominant choline peak, with relatively small NAA and creatine/phosphocreatine peaks. B. Normal 35-week neonate. Spectrum in frontal white
matter is less mature than that through the basal nuclei. The creatine/phosphocreatine and NAA peaks are relatively lower than the choline and myo-
inositol peaks compared to (A). Some lactate (arrows in A and B) is normal in the white matter at this stage. C. Normal term neonate. By this stage, the
NAA is relatively more abundant compared to choline. Lactate has essentially disappeared. D. Normal term neonate. Spectrum in frontal white matter
is still less mature than that through the basal nuclei, but more mature than at 35 weeks (B). The creatine/phosphocreatine and NAA peaks are relatively
lower than the choline and myo-inositol peaks compared to the basal ganglia (C) but the NAA is relatively larger compared with 35 weeks. E. Normal
12-month-old infant. A relative increase is seen in the size of the creatine/phosphocreatine and NAA peaks compared to the choline peak. Myo-inositol
remains fairly prominent at this age. F. Normal 24-month-old child. The spectrum begins to resemble that of an adult, with dominant NAA peak and
relatively small creatine/phosphocreatine and myo-inositol peaks.

Barkovich_Chap02.indd 62 5/7/2011 2:42:38 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 63

peaks in the adult proton spectrum. Myo-inositol, conversely, decreases to overlying cortex and remains that way until late adulthood when
to a concentration of 6 mmol/kg in the adult, resulting in its decreasing degenerative changes develop.
prominence in the spectrum of the older child and adult (177,211). On ADC maps, the signal intensity is proportional to the amount
of water diffusion only, without T2 information. As discussed in Chap-
ter 1, areas of reduced diffusion appear brighter than normal tissue on
Diffusion Tensor Imaging and Tractography
DWIs, whereas reduced diffusion appears darker on diffusivity (ADC)
As discussed in Chapter 1, special techniques that were initially devel- images. As premature and term neonates have a great amount of water
oped in the 1960s (214,215) allow MRI to be used in measuring the in their unmyelinated brain and, as a result, very long T1 and T2 relax-
rate and direction of diffusion of water in the brain (216,217). The ation times of the white matter, diffusivity maps are typically more
rate of diffusion of free water in the brain seems to be primarily related sensitive to subtle injury than DWIs. Therefore, a series of normal
to the maturity of the white matter; as the brain matures, the amount diffusivity images during development are shown in Figure 2-34. On
of motion of the water molecules (as measured by the diffusivity, also diffusivity images of preterm and term neonates, marked contrast is
called average diffusivity [Dav] or apparent diffusion coefficient [ADC], present between hypointense gray matter and hyperintense white mat-
see Chapter 1) decreases, probably as a result of increased complexity ter (Fig. 2-34A–H). This contrast is more marked in premature neo-
of white matter pathways and increasing myelination. The develop- nates (Fig. 2-34A–D) than in term neonates (Fig. 2-34E–H). If residual
ment of anisotropy of water motion along white matter pathways, as germinal matrix is present, it has diffusion characteristics similar to
measured by fractional anisotropy (FA), seems to correlate best with cortex (Fig. 2-34B and C). The posterior limb of the internal capsule,
the development of immature oligodendrocytes (218). High levels of which myelinates early and has compact axons, has reduced diffusion
anisotropy can be present in the absence of myelin (219–221), sug- (hypointense, Fig. 2-34B and D) compared to the hemispheric white
gesting that factors other than myelin—neurofilament development, matter. As the child matures during the first year, the contrast between
aligned axons, single membrane barriers, cohesive and compact fiber gray and white matter diminishes (Fig. 2-34I–P) until they become
tracts, and changes in the extracellular and intracellular matrices—are isointense in much of the cerebrum by age 9 months. The early devel-
all important factors in the development of anisotropic water motion oping pathways, such as the sensorimotor pathways, become hypoin-
in white matter (218,219). tense compared to cortex by about 9 months, while the late developing
pathways in the anterior frontal and parietal lobes remain hyperin-
Diffusion Imaging of Brain Maturation tense compared to cortex (Fig. 2-34M–P). Beyond age 12 months, the
Depending upon the manufacturer of the MR scanner, diffusion infor- cerebral cortex and cerebral white matter remain largely isointense
mation may be displayed in several different forms. The most common (Fig. 2-34Q–X) except for persistent white matter hyperintensity in the
of these forms are the diffusion-weighted image (DWI), which con- peritrigonal and frontal subcortical regions, which persists until the
tains both T2 and diffusion information, and the diffusivity map (ADC end of the second year.
map), in which image contrast is determined only by the magnitude of
water motion in each voxel. As information on water diffusion may be Diffusion Tensor Imaging of Brain Maturation
useful in assessing injury to (148,222,223), or metabolic derangement As discussed in Chapter 1, diffusion measurements can also be used to
of (224) the neonatal and infant brain, an understanding of the appear- calculate measurements of diffusion anisotropy (88). Several measure-
ance of diffusion-based images in normal children of various ages is ments can be used to calculate anisotropy. Of these, FA provides the
critical. However, when the brain is diffusely injured, DWI and diffusivity most detailed and extensive depiction of anisotropy and achieves high-
maps may appear normal despite widespread reduced diffusion. In such est signal-to-noise, volume ratio provides the strongest contrast between
cases, calculation of diffusivity values and correlation of those values with low and high anisotropy areas (but has decreased signal-to-noise and
established norms for the patient’s age and the region being measured therefore decreased resolution of mildly anisotropic areas), and rela-
(25,90,225,226) are absolutely essential. tive anisotropy is intermediate between the other two (227). The dif-
DWIs contain information from both T2 relaxation and diffu- fusion information can be displayed numerically, or as images. Several
sion. In premature neonates, the diffusion effects dominate, resulting groups have published the evolution of calculated diffusivity and diffu-
in nearly all white matter being hypointense relative to cortical gray sion anisotropy of the brain during development (89,90,225,226,228).
matter (Fig. 2-33A–D). The posterior limbs of the internal capsules are Sometimes, however, display of the data as an image is more useful
exceptions to this, being iso- to slightly hyperintense to cortical gray from the perspective of the radiologist, particularly when management
matter (Fig. 2-33A and B). In term infants (Fig. 2-33E–H), the white decisions need to be made quickly on a sick neonate or child.
matter in the perirolandic region is slightly more hyperintense than If DTI is performed with adequately high signal-to-noise, aniso-
in premature infants, becoming nearly isointense to overlying cortex, tropic water motion is seen throughout the white matter of the brain,
and the posterior limbs of the internal capsules are isointense to sur- even in the unmyelinated brains of premature neonates (226,229,230).
rounding structures (Fig. 2-33D). By age 3 months (Fig. 2-33I–L), the Although the amount of anisotropy in any region varies depending
central white matter becomes isointense to relatively hypointense to upon the voxel size and the signal-to-noise ratio, the greatest degree
overlying cortex, presumably due to an increasing predominance of of anisotropy is found in the corpus callosum, internal capsules, brain-
T2 effects over diffusion effects. The frontal white matter, which is the stem, and cerebellar peduncles (89,224), and all of the major white
last area to mature, remains hyperintense compared with overlying matter tracts show some degree of anisotropy. The maximum eigen-
gray matter (Fig. 2-33K and L). Between the ages of 3 and 9 months value, corresponding to the magnitude in the direction of greatest dif-
(Fig. 2-33M–P), the anterior and posterior cerebral white matter also fusion, is larger in white matter than in gray matter throughout brain
becomes more hypointense compared to overlying cortex. During the development. In contradistinction, the intermediate and minimum
first few post-term months, the internal capsule becomes progres- eigenvalues are larger in white matter than in gray matter during the
sively hypointense, with the posterior limb attaining mature contrast newborn period, but become smaller in white matter than in gray mat-
(hypointensity) by age 3 to 4 months (Fig. 2-33K) and the anterior limb ter with progressive myelination. As the brain matures, anisotropic
by age 20 months. Beyond the age of about 9 months (Fig. 2-33Q–X), water motion becomes visible in all white matter regions, including
the cerebral white matter becomes uniformly hypointense compared the subcortical association axons. The amount of anisotropy increases

Barkovich_Chap02.indd 63 5/7/2011 2:42:40 AM


64 Pediatric Neuroimaging

FIG. 2-33. DWIs at two levels of the devel-


oping brain. Signal intensity in these images
reflects both T2 weighting and diffusion
weighting. A–D. Premature neonate with
adjusted gestational age of 32 weeks. In pre-
mature neonates, the diffusion effects domi-
nate, resulting in nearly all white matter being
hypointense relative to cortical gray matter.
The posterior limbs of the internal capsules
are exceptions to this, being iso- to slightly
hyperintense to cortical gray matter (A and
B). E–H. Two-week-old neonate, born at
term. In term infants, the white matter in the
perirolandic region is slightly more hyperin-
tense than in premature infants, becoming
nearly isointense to overlying cortex, and the
posterior limbs of the internal capsules are
isointense to surrounding structures (D).

Barkovich_Chap02.indd 64 5/7/2011 2:42:40 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 65

FIG. 2-33. (Continued) I–L. Three-month-old


infant. By age 3 to 4 months, the central white
matter becomes isointense to relatively hypoin-
tense to overlying cortex, presumably due to an
increasing predominance of T2 effects over dif-
fusion effects. The frontal white matter, which
is the last area to mature, remains hyperintense
compared with overlying gray matter (K and L).
During the first few post-term months, the inter-
nal capsule becomes progressively hypointense,
with the posterior limb attaining mature con-
trast (hypointensity) by age 3 to 4 months (K)
and the anterior limb by age 20 months. M–P.
Nine-month-old infant. Between the ages of 3 and
9 months, the anterior and posterior cerebral white
matter also becomes more hypointense compared
to overlying cortex. Note, however, that the less
mature regions, such as the anterior frontal and
anterior temporal lobes, remain isointense.

Barkovich_Chap02.indd 65 5/7/2011 2:42:41 AM


66 Pediatric Neuroimaging

FIG. 2-33. (Continued) Q–T. Fifteen-month-


old infant. Beyond the age of about 9 months, the
cerebral white matter progressively becomes uni-
formly hypointense compared to overlying cortex.
U–X. Four-year-old child. By the end of the sec-
ond postnatal year, the white matter is uniformly
hypointense and remains that way until late
adulthood when degenerative changes develop.

Barkovich_Chap02.indd 66 5/7/2011 2:42:43 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 67

FIG. 2-34. ADC images of the developing brain.


On these images, low signal intensity reflects
a small amount of free water motion, and high
signal intensity results from a high degree of free
water motion. A–D. Diffusion images of a pre-
term (28 week) neonate. The white matter is very
hyperintense compared with the cortex and deep
gray matter. Note the normal hypointensity of
the residual germinal matrix in the walls of the
lateral ventricles. The internal capsule is slightly
more hypointense than the white matter of the
centrum semiovale. E–H. Term neonate. The
appearance is similar to that in the preterm infant
with the exception that some hypointensity, rep-
resenting reduced water motion, is now pres-
ent in the corticospinal tracts within the corona
radiata (H).

Barkovich_Chap02.indd 67 5/7/2011 2:42:44 AM


68 Pediatric Neuroimaging

FIG. 2-34. (Continued) I–L. 3-month-old


infant. At age 3 months, the white matter is
becoming isointense to the gray matter in the
sensorimotor pathways. The deep gray nuclei
are slightly hyperintense. M–P. 9-month-old
infant. At this age, most hemispheric white mat-
ter is isointense to cortex. In the regions that
mature late (anterior frontal and parietal, P),
the white matter remains slightly hyperintense.

Barkovich_Chap02.indd 68 5/7/2011 2:42:45 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 69

FIG. 2-34. (Continued) Q–T. Fifteen-month-old


infant. At this age, most white matter is isointense to
cortex, and the sensorimotor white matter is slightly
hypointense compared to cortex. U–X. Four-
year-old child. In the mature brain, white matter
has become slightly hypointense compared to cor-
tex except for some patchy hyperintensity (in W)
posterior and superior to the ventricular trigones.

over the first decade of life, with the compact white matter pathways matter, anisotropy continues to increase beyond the first decade of life
(corticospinal tracts and corpus callosum) showing early matura- (231,232).
tion of anisotropy while the anisotropy of the noncompact white It seems rather clear that much of the diffusion anisotropy in the
matter of the centrum semiovale matures later (231). In the slowest maturing brain results from the presence of myelin sheaths around the
maturing regions, the subcortical white matter and the frontal white axons (216,233,234). However, the initial development of anisotropy

Barkovich_Chap02.indd 69 5/7/2011 2:42:47 AM


70 Pediatric Neuroimaging

precedes the formation of myelination (220,221) and seems to corre- perpendicular to the cortical surface) while the two minor eigenvalues
late with the developmental expansion of immature oligodendrocytes remain unchanged. This analysis of cortical maturation via DTI may
during the premyelination period (218). Additionally, diffusion anisot- prove useful in predicting long-term developmental outcome in pre-
ropy occurs in unmyelinated nerve bundles (235,236) and this anisot- mature neonates.
ropy can be eliminated by administration of tetrodotoxin, suggesting DTI can also be used to identify transient regions of differing
that it results from a physiologic process, possibly the rapid flux of ions anisotropy in the developing brain. For example, the subplate (also
into the developing axon immediately prior to myelination (220,221). called cortical layer 7) is a transient region that lies deep to the cerebral
Thus, DTI, by allowing us to assess characteristics of water motion in cortex but superficial to the intermediate zone (developing white mat-
the brain, has the potential to help us assess cerebral maturation, and, ter). Many axons that will eventually synapse with neurons in the cor-
perhaps to better understand cerebral physiology and maturation. tex temporarily synapse with neurons in the subplate until the cortex is
However, it is important to remember that anisotropy is not exclusively sufficiently mature. The moderate diffusivity and low anisotropy of the
caused by myelination; many of the causes of anisotropy are yet to be subplate differentiate it from the low diffusivity and high anisotropy
determined. of the overlying cortex and from the low diffusivity and medium-to-
Although the brains of near-term and term neonates, infants, and high anisotropy of the intermediate zone (239). Altered development
children show no anisotropy of gray matter, DTI of very preterm neo- of the subplate may be a clue to future developmental abnormalities
nates and infants shows an interesting phenomenon of anisotropic in the child and to what aspects of prematurity and its therapy put the
water diffusion in the cerebral cortex (Fig. 2-35). This anisotropy is neonate at risk.
believed to result from the radial organization of the immature cere-
bral cortex (237). In the immature cortex, the predominant features Diffusion Tensor Tractography of Brain Maturation
are radial glial cells and large pyramidal neurons with prominent, Since white matter pathways in the brain exist in three dimensions, two
radially oriented apical dendrites; these structures cause horizontal dimensional (2D) representations of DTI such as ADC and FA maps
water motion to be relatively impaired, resulting in radially oriented are intrinsically limited. This shortcoming has led to the development
anisotropy. As the cortex matures, neurons extend horizontally ori- of 3D DTI fiber tractography techniques. There are many different
ented basal dendrites and horizontally oriented thalamocortical axons techniques for performing fiber tractography, but most of the currently
invade the cortex (237). In addition, intracortical association axons used methods are variations on the same underlying principle of track-
navigate horizontally through the developing cortex. The developing ing along the orientation of the primary eigenvector of the diffusion
horizontal, or laminar, organization causes impairment of radially tensor from voxel to voxel (240–242). Note that, because the process of
directed water motion, resulting in increasing isotropic water motion water diffusion exhibits antipodal symmetry, DTI tractography cannot
in the cortex. The cortical anisotropy disappears first in the periro- distinguish forward from backward (i.e., antegrade versus retrograde)
landic region of the cortex, where it is no longer seen after about 30 along a fiber trajectory. Tractography can be used to separate func-
postconceptional weeks (238). Anisotropy persists the longest in the tionally distinct white matter pathways using the multiple region-of-
anterior temporal and prefrontal cortex, where is seen as late as 37 interest (ROI) method in which a priori anatomic knowledge of the
postconceptional weeks (Fig. 2-35B) (238). This loss of cortical anisot- course of a white matter tract is used to delineate its entire 3D trajec-
ropy is the result of a decrease in the maximum eigenvalue (oriented tory (242). The fiber tracking is initiated at an ROI placed in one part

FIG. 2-35. Diffusion anisotropy of the premature cerebral cortex. A. FA images of a 28-week preterm neonate. Note the hyperintensity in much of the
cerebral cortex (other than the central regions), indicating high anisotropy of most of the cortex. The most mature cortical area, the sensorimotor region
(white arrows) is hypointense, indicating low anisotropy due to more cortical maturation. B. FA images of a 37-week preterm neonate. Most of the cortex
is now hypointense, indicating low anisotropy. The prefrontal cortex (white arrows) is still slightly hyperintense, indicating persistent anisotropy in this
late-maturing region. Central hyperintensity (white asterisks) shows anisotropy in the developing white matter pathways.

Barkovich_Chap02.indd 70 5/7/2011 2:42:48 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 71

of the pathway, and only those fiber tracks that pass through an ROI that the T1 shortening on spin-echo images results from a magnetiza-
placed in another portion of the pathway are retained. This power- tion transfer interaction of glycocerebrosides and cholesterol on the
ful method, which has been experimentally verified (243), can be used surface of the myelin molecule with free water in the brain. Because
to “dissect” out adjacent but functionally distinct white matter con- magnetization transfer ratios can be calculated by use of magnetiza-
nections of the developing human brain, such as the pyramidal tract tion transfer in imaging sequences, it may ultimately be possible to
and the somatosensory radiation. 3D tractography can also be used to use increases in magnetization transfer to quantify brain maturation.
measure tract-based diffusivity, FA, or other DTI parameters, by inte- This can be done by looking at changes in magnetization transfer
grating the parameters in all the voxels intersected by the fiber tracks. in regions of the brain or in global magnetization transfer changes
The advantages of this tract-based quantitation over traditional ROI (256). It remains to be seen which is more sensitive to subtle changes
measurements from DTI are that it is more specific to the functionally in myelination.
distinct axonal pathway of interest, that it reflects the entire 3D course
of the pathway rather than just one location within the pathway, and
Perfusion Imaging
that it is more reproducible than manually placed ROIs, as has been
shown in the adult brain by a cross-sectional study (244) and by a lon- Perfusion imaging is uncommonly performed in children. It is occa-
gitudinal study (245). sionally necessary in children with vasculopathies such as moyamoya,
Applying 3D DTI fiber tracking to the developing brain of pre- where revascularization is contemplated. Susceptibility-weighted per-
mature newborns, Partridge et al. (246) found that tract-based mea- fusion imaging, as described in Chapter 1, is usually performed, as it
surements of ADC, FA, and the three diffusion tensor eigenvalues in allows the evaluation of relative cerebral blood flow, blood volume,
the pyramidal tract are all more reproducible than manual 2D ROI and the time it takes a bolus to reach the region of interest (257,258).
measurements, with less interoperator and intraoperator variability. However, age-related changes require the use of a quantitative method
DTI tractography has also been used by Berman et al. (247,248) to such as arterial spin labeling (ASL) (259); at the time of writing of this
gauge maturational changes separately in the pyramidal tract, soma- chapter, ASL is still not widely available and is rather difficult to apply
tosensory radiations, and visual pathways of premature newborns. in infants. Some preliminary work suggests that ASL can be used in
At any given age, tract-based diffusivity in the motor pathway was children and that cerebral blood flow increases with age in both gray
less than that of the somatosensory radiations, whereas tract-based matter and white matter (260). The cerebral blood flow appears to sud-
FA was greater in the motor pathway, while visual maturation was denly decrease in adolescence (260). It is likely that ASL will be applied
strongly associated with FA of the optic radiations. More recently, more frequently in the future and that this section will grow in future
large-scale DTI investigations have characterized the time course of editions of this book.
age-related white matter microstructural changes across the entire
lifespan, all of which have a “U” or inverted “U” shape reflecting the
Functional MRI
effects of maturation early in life and of senescence late in life. A fiber
tractography study of the corpus callosum in 315 normal volunteers Blood oxygenation level–dependent (BOLD) fMRI has been a pow-
of ages 5 to 59 years showed that peak FA values were reached in erful research tool for mapping human brain activity for the past
the third decade of life, whereas the minimum diffusivity values were two decades (261,262). The T2*-weighted echo-planar acquisition
reached somewhat later, often during the fourth decade of life (249). required for BOLD fMRI is sensitive to differences in the oxygenation
An atlas-based DTI study of 16 different white matter tracts in 430 content of blood. The ratio of oxyhemoglobin to deoxyhemoglobin
healthy subjects ranging in age from 8 to 85 years showed similar rises when cortical regions become more metabolically active due to
results, with peak FA values in the third decade of life and minimum a concomitant increase in blood flow; this results in an increase of
diffusivity values in the fourth to fifth decade of life (250). Notably, the BOLD signal and thereby enables identification of activated areas
the corticospinal tract showed evidence of particularly early and in response to a specific sensorimotor, cognitive, or behavioral task.
rapid maturation whereas the dorsal and ventral cingulum bundles However, compared to the adult brain, there has been relatively little
showed more protracted development compared to the other tracts application of BOLD fMRI techniques to the study of brain devel-
examined. Clearly, fiber tracking will be used extensively in the future opment in newborns, infants, and children or for the investigation
to study white matter tracts, both in development and in aging. of developmental disorders. This is largely because these pediatric
populations are often unable to cooperate with the task paradigms,
such as tests of speech, language, and motor function, required to
Magnetization Transfer Imaging
localize activation using fMRI. This has recently changed with the
Another technique that may be useful in the analysis of brain develop- advent of “resting-state” fMRI techniques, which do not require a
ment is magnetization transfer. It has been shown that the amount task paradigm, but rather rely on BOLD “functional connectivity” to
of magnetization transfer in the brain increases during myelination map functionally-specific cortical regions. Functional connectivity
(251). In addition, the increase in magnetization transfer parallels refers to the phenomenon of temporal synchronization of function-
that of myelination as determined by histology (251). As discussed ally related brain areas. For BOLD fMRI, this was first demonstrated
in Chapter 1, almost all magnetization transfer in the brain seems to in the motor cortex where it was observed that the spontaneous
result from interactions of free water with components of myelin, in fluctuations of the BOLD signal were highly correlated between left
particular the hydroxyl and amine moieties of cholesterol and glyco- and right motor cortex even in the absence of a motor task or any
cerebrosides on the myelin surface (252,253). Destruction of myelin other goal-directed behavior (263). Currently, two different meth-
results in decreased magnetization transfer (254). Moreover, the onset ods are widely used to map functionally connected brain regions,
of T1 shortening seen during the early phases of myelination on one hypothesis-driven and the other data-driven. The hypothesis-
T1-weighted images corresponds temporally and topographically to driven method is called seed-voxel correlation and requires placing
the onset of magnetization transfer (Fig. 2-36) (255). Thus, it appears a ROI on the area to be studied and calculating the temporal cor-

Barkovich_Chap02.indd 71 5/7/2011 2:42:49 AM


72 Pediatric Neuroimaging

FIG. 2-36. Magnetization transfer images in a neonate. A and B. SE 550/15 images show T1 shortening in the
dorsal brainstem and the posterior limb of the internal capsule/lateral thalamus. C and D. Magnetization transfer
images show considerable magnetization transfer in the areas of T1 shortening on the T1-weighted images, but
nowhere else.

relations of the BOLD signal with those of other regions of inter- inferior parietal lobules, ventromedial prefrontal cortex, and variable
est in the brain (263). The data-driven method, called independent other regions including temporal cortex (267,268).
component analysis (ICA), is fully automated and can be applied Resting-state fMRI has begun to be used in the research setting
to the whole brain to derive multiple functionally connected brain to investigate the process of human brain development. The hypoth-
networks (264). esis is that, as white matter tracts in the brain mature and myelinate,
Examples of resting-state networks, also known as intrinsically the functional connectivity between intrinsically connected cortical
connected networks, computed from ICA of BOLD fMRI at 3 Tesla regions will strengthen. More importantly, it is hoped that abnormali-
(3 T) are shown in Figure 2-37. More than a dozen such cortical net- ties of functional connectivity might help to determine the effect of
works have been identified in the human brain to date (265,266). The brain injury on brain function and be a means of directing therapy
motor network represents the temporal synchrony between left and in neonates and children with brain injury. Resting state networks,
right motor cortical regions; a visual network similarly includes left including motor, auditory, visual, and DMN, can be detected in prema-
and right visual cortex. The default mode network (DMN) is the name ture infants scanned at term-equivalent age (269), indicating that cor-
given to cortical areas that have the greatest metabolic activity when tical regions are functionally connected well before the onset of white
the subject is at rest; they deactivate (have less activity) during goal- matter myelination in many pathways connecting these networks. In a
directed behavior. The DMN is more complex than the motor or visual study of preterm infants as young as 26 weeks postmenstrual age, the
networks, consisting of bilateral posterior cingulate gyrus, precuneus, most mature functional connections were found between homologous

Barkovich_Chap02.indd 72 5/7/2011 2:42:49 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 73

FIG. 2-37. Resting-state functional connectivity MRI (fcMRI) of infants. The colored areas of cortex in
these images show those cortical areas that have the greatest metabolic activity when the subject is at rest;
they deactivate (have less activity) during goal-directed behavior. A. ICA of resting-state fMRI reveals cor-
related BOLD activity of bilateral regions of the posterior cingulate and adjacent precuneus, inferior pari-
etal lobules, ventromedial prefrontal cortex, and lateral temporal cortex. These comprise the DMN, those
areas that are maximally activated in the resting state. B. ICA of visual network fMRI reveals correlated
BOLD activity of bilateral regions of the occipital lobes corresponding to visual cortex.

cortical regions of the two cerebral hemispheres (270). The DMN could white matter tracts compared to subcortical-cortical projection tracts.
not be detected in premature newborns, but was identified in term-born A larger resting-state fMRI study of 210 subjects ranging in age from
infants (270). In a study of 71 healthy infants and children ages 2 weeks 7 to 31 years of age reported that children showed more short-range
to 2 years, only a few elements of the DMN were present in newborns, functional connectivity and less long-range functional connectivity
but the number of connected cortical regions of the DMN increased than adults (273). This may relate to the protracted development of
up to 1 year of age (271). By 2 years of age, the DMN resembled that long white matter fibers compared to shorter connections and reflect
of the adult brain. A comparison of resting state networks between an overall change in cortical information processing during develop-
23 children ages 7 to 9 years and 22 young adults ages 19 to 22 years ment from more local to more distributed. However, research into the
revealed that subcortical to cortical connectivity was relatively greater development of brain networks using functional connectivity informa-
in children, whereas adults showed more predominant corticocortical tion from fMRI and structural connectivity information from DTI is
connectivity (272). This may be explained by the relatively delayed and itself in its infancy, and much more remains to be discovered that may
prolonged maturation of corticocortical association and commissural paint a more complex picture than these initial investigations.

Barkovich_Chap02.indd 73 5/7/2011 2:42:50 AM


74 Pediatric Neuroimaging

FIG. 2-37. (Continued) C. ICA of auditory network fMRI reveals correlated BOLD activity of bilateral
regions of the superior temporal gyri corresponding to auditory cortex. D. ICA of dorsal attention mode
fMRI reveals correlated BOLD activity of bilateral parietal lobe cortex.

REFERENCES 4. Miyoshi G, Hjerling-Leffler J, Karayannis T, et al. Genetic fate mapping reveals


that the caudal ganglionic eminence produces a large and diverse population
1. Markram H, Toledo-Rodriguez M, Wang Y, Gupta A, Silberberg G, Wu C. of superficial cortical interneurons. J Neurosci 2010;30(5):1582–1594.
Interneurons of the neocortical inhibitory system. Nat Rev Neurosci 5. Nobrega-Pereira S, Gelman D, Bartolini G, Pla R, Pierani A, Marin O. Ori-
2004;5:793–807. gin and molecular specification of globus pallidus neurons. J Neurosci 2010;
2. Wonders CP, Anderson SA. The origin and specification of cortical interneu- 30(8):2824–2834.
rons. Nat Rev Neurosci 2006;7:687–696. 6. Flandin P, Kimura S, Rubenstein JLR. The progenitor zone of the ven-
3. Tucker ES, Segall S, Gopalakrishna D, et al. Molecular specification and pat- tral medial ganglionic eminence requires Nkx2-1 to generate most of the
terning of progenitor cells in the lateral and medial ganglionic eminences. globus pallidus but few neocortical interneurons. J Neurosci 2010;30(8):
J Neurosci 2008;28:9504–9518. 2812–2823.

Barkovich_Chap02.indd 74 5/7/2011 2:42:52 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 75

7. Xu Q, Cobos I, De La Cruz E, Rubenstein JLR, Anderson SA. Origins of 34. Sidman R, Rakic P. Development of the human central nervous system.
cortical interneuron subtypes. J Neurosci 2004;24:2612–2622. In: Haymaker W, Adams R, eds. Histology and Histopathology of the Nervous
8. Jakovcevski I, Zecevic N. Sequence of oligodendrocyte development in the System. Springfield: Charles C. Thomas, 1982:3–145.
human fetal telencephalon. Glia 2005;49:480–491. 35. Allendoerfer K, Shatz C. The subplate, a transient neocortical structure:
9. Arber S. Subplate neurons: bridging the gap to function in the cortex. its role in the development of connections between thalamus and cortex.
Trends Neurosci 2004;27:111–113. Annu Rec Neurosci 1994;17:185–218.
10. Kostovic I, Judas M, Radios M, Hrabac P. Laminar organization of the 36. Kostovic I, Rakic P. Developmental history of the transient subplate zone in
human fetal cerebrum revealed by histochemical markers and magnetic the visual and comatosensory cortex of the macaque monkey and human
resonance imaging. Cereb Cortex 2002;12:536–544. brain. J Comp Neurol 1990;297:441–470.
11. Garel C. MRI of the Fetal Brain. Normal Development and Cerebral 37. Rados M, Judas M, Kostovic´ I. In vitro MRI of brain development. Eur J
Pathologies. Berlin: Springer, 2004. Radiol 2006;57:187–198.
12. Perkins L, Hughes E, Srinivasan L, et al. Exploring cortical subplate evo- 38. Fitz CR. Developmental anomalies of the brain. In: Heinz ER, ed.
lution using magnetic resonance imaging of the fetal brain. Dev Neurosci Neuroradiology, vol 4. New York: Churchill Livingstone, 1984:215–224.
2008;30:211–220. 39. McArdle CB, Richardson CJ, Nicholas DA, Mirfakhraee M, Hayden CK,
13. Lemire RJ, Loeser JD, Leech RW, Alvord EC. Normal and Abnormal Devel- Amparo EG. Developmental features of the neonatal brain: MR imag-
opment Of The Human Nervous System. Hagarstown: Harper & Row, 1975. ing. Part I. Gray-white matter differentiation and myelination. Radiology
14. Chi J, Dooling E, Gilles F. Gyral development of the human brain. Ann 1987;162:223–229.
Neurol 1977;1:86–93. 40. Iai M, Yamamura T, Takashima S. Early expression of proteolipid protein in
15. Larroche J. Critères morphologiques du développement du systeme human fetal and infantile cerebri. Pediatr Neurol 1997;17:235–239.
nerveux central du foetus hemain. J Neuroradiol 1981;8:93–108. 41. Counsell S, Maalouf E, Fletcher A, et al. MR imaging assessment
16. Garel C, Chantrel E, Elmaleh M, Brisse H, Sebag G. Fetal MRI: normal ges- of myelination in the very preterm brain. Am J Neuroradiol 2002;23:
tational landmarks for cerebral biometry, gyration and myelination. Childs 872–881.
Nerv Syst 2003;19(7–8):422–425. 42. van Wezel-Meijler G, van der Knaap MS, Sie LTL, et al. Magnetic resonance
17. Adamsbaum C, Moutard ML, Andre C, et al. MRI of the fetal posterior imaging of the brain in premature infants during the neonatal period.
fossa. Pediatr Radiol 2005;35:124–140. Neuropediatrics 1998;29:89–96.
18. Hill J, Dierker D, Neil J, et al. A surface-based analysis of hemispheric 43. Sie LTL, van der Knaap MS, van Wezel-Meijler G, Valk J. MRI assessment
asymmetries and folding of cerebral cortex in term-born human infants. of myelination of motor and sensory pathways in the brain of preterm and
J Neurosci 2010;30(6):2268–2276. term-born infants. Neuropediatrics 1997;28:97–105.
19. van der Knaap MS, van Wezel-Meijler G, Barth PG, Barkhof F, Adèr HJ, 44. Childs A-M, Ramenghi LA, Cornette L, et al. Cerebral maturation in
Valk J. Normal gyration and sulcation in preterm and term neonates: premature infants: quantitative assessment using MR imaging. Am J
appearance on MR images. Radiology 1996;200:389–396. Neuroradiol 2001;22:1577–1582.
20. Battin MR, Maalouf EF, Counsell SJ, et al. Magnetic resonance imaging 45. Brody BA, Kinney HC, Kloman AS, Gilles FH. Sequence of central nervous
of the brain in very preterm infants: visualization of the germinal matrix, system myelination in human infancy. I. An autopsy study of myelination.
early myelination, and cortical folding. Pediatrics 1998;101:957–962. J Neuropath Exp Neurol 1987;46:283–301.
21. Ruoss K, Lövblad K, Schroth G, Moessinger AC, Fusch C. Brain develop- 46. Kinney HC, Brody BA, Kloman AS, Gilles FH. Sequence of central ner-
ment (sulci and gyri) as assessed by early postnatal MR imaging in preterm vous system myelination in human infancy. II. Patterns of myelination in
and term newborn infants. Neuropediatrics 2001;32:69–74. autopsied infants. J Neuropathol Exp Neurol 1988;47:217–234.
22. Barkovich AJ, Kjos BO, Jackson DE, Jr, Norman D. Normal matura- 47. Yakovlev PI, Lecours AR. The myelogenetic cycles of regional maturation of
tion of the neonatal and infant brain: MR imaging at 1.5 T. Radiology the brain. In: Minkowski A, ed. Regional Development of the Brain in Early
1988;166:173–180. Life. Oxford: Blackwell, 1967:3–70.
23. Chugani HT, Phelps ME, Mazziotta JC. Positron emission tomogra- 48. Martin E, Krassnitzer S, Kaelin P, Boesch C. MR imaging of the brainstem:
phy study of human brain functional development. Ann Neurol 1987;22: normal postnatal development. Neuroradiology 1991;33:391–395.
487–497. 49. van der Knaap MS, Valk J. MR imaging of the various stages of normal
24. Barkovich AJ, Hallam D. Neuroimaging in perinatal hypoxic-ischemic myelination during the first year of life. Neuroradiology 1990;31:459–470.
injury. Ment Retard Dev Disabil Res Rev 1997;3:28–41. 50. Barkovich AJ. MR of the normal neonatal brain: assessment of deep
25. Mukherjee P, Gil K, Veervaraghevan S, et al. Region-specific maturation of structures. Am J Neuroradiol 1998;19:1397–1403.
the cerebral cortex in premature newborns demonstrated with high reso- 51. Shaw DWW, Weinberger E, Astley SJ, Tsuruda JS. Quantitative compari-
lution diffusion tensor imaging. Paper presented at: 11th Meeting of the son of conventional spin echo and fast spin echo during brain myelination.
International Society of Magnetic Resonance in Medicine 2003, Toronto, J Comput Assist Tomogr 1997;21:867–871.
Canada. 52. Steen RG, Ogg RJ, Reddick WE, Kingsley PB. Age-related changes in the
26. Penrice J, Cady EB, Lorek A, et al. Proton magnetic resonance spectroscopy pediatric brain: quantitative MR evidence of maturational changes during
of the brain in normal preterm and term infants and early changes after adolescence. Am J Neuroradiol 1997;18:819–828.
perinatal hypoxia-ischemia. Pediatr Res 1996;40:6–14. 53. Childs A-M, Ramenghi LA, Evans DJ, et al. MR features of develop-
27. Girard N, Raybaud C, Poncet M. In vivo MR study of brain maturation in ing periventricular white matter in preterm infants: evidence of glial cell
normal fetuses. Am J Neuroradiol 1995;16:407–413. migration. Am J Neuroradiol 1998;19:971–976.
28. Levine D, Barnes PD. Cortical maturation in normal and abnormal fetuses 54. van der Knaap MS, Valk J, de Neeling N, Nauta JJP. Pattern recognition in
as assessed with prenatal MR imaging. Radiology 1999;210:751–758. MRI of white matter disorders in children and young adults. Neuroradiology
29. Simon E, Goldstein R, Coakley F, et al. Fast MR imaging of fetal CNS 1991;33:478–493.
anomalies in utero. Am J Neuroradiol 2000;21:1688–1698. 55. Hittmair K, Kramer J, Rand T, Bernert G, Wimberger D. Infratentorial
30. Triulzi F, Parazzini C, Righini A. MRI of fetal and neonatal cerebellar brain maturation: a comparison of MRI at 0.5 and 1.5T. Neuroradiology
development. Semin Fetal Neonatal Med 2005;10(5):411. 1996;38:360–366.
31. Parazzini C, Righini A, Rustico M, Consonni D, Triulzi F. Prenatal mag- 56. Baker LL, Stevenson DK, Enzmann DR. End stage periventricular
netic resonance imaging: brain normal linear biometric values below 24 leukomalacia: MR imaging evaluation. Radiology 1988;168:809–815.
gestational weeks. Neuroradiology 2008;50:877–883. 57. Groeschel S, Chong WK, Surtees R, Hanefeld F. Virchow-Robin spaces on
32. Glenn OA. Normal development of the fetal brain by MRI. Semin Perinatol magnetic resonance images: normative data, their dilatiation, and a review
2009;33:208–219. of the literature. Neuroradiology 2006;48:745–754.
33. Chung R, Kasprian G, Brugger PC, Prayer D. The current state and future 58. Rollins NK, Deline C, Morriss MC. Prevalence and clinical significance of
of fetal imaging. Clin Perinatol 2009;36(3):685-699. dilated Virchow-Robin spaces in childhood. Radiology 1993;189:53–57.

Barkovich_Chap02.indd 75 5/7/2011 2:42:54 AM


76 Pediatric Neuroimaging

59. Hittmair K, Wimberger D, Rand T, et al. MR assessment of brain maturation: 85. van der Knaap MJ, Valk JM. White matter. In: van der Knaap MJ,
comparison of sequences. Am J Neuroradiol 1994;15:425–433. Valk J, eds. Magnetic Resonance of Myelin, Myelination, and Myelin Disor-
60. Ding X-Q, Kucinski T, Wittkugel O, et al. Normal brain maturation charac- ders, 2nd ed. Berlin: Springer, 1995:1–17.
terized with age-related T2 relaxation times: an attempt to develop a quan- 86. Kjos BO, Umansky R, Barkovich AJ. MR of the brain in children with
titative imaging measure for clinical use. Invest Radiol 2004;39:740–746. developmental retardation of unknown cause. Am J Neuroradiol 1990;11:
61. Whittall KP, MacKay AL, Graeb DA, Nugent RA, Li DK, Paty DW. In vivo 1035–1040.
measurement of T2 distributions and water contents in normal human 87. Barkovich AJ, Truwit CL. MR of perinatal asphyxia: Correlation of gesta-
brain. Magn Reson Med 1997;37:34–43. tional age with pattern of damage. Am J Neuroradiol 1990;11:1087–1096.
62. Kolind SH, Mädler B, Fischer S, Li DK, MacKay AL. Myelin water imag- 88. Basser PJ, Pierpaoli C. Microstructural and physiological features of tissues
ing: Implementation and development at 3.0T and comparison to 1.5T elucidated by quantitative-diffusion-tensor MRI. J Magn Reson B 1996;
measurements. Magn Reson Med 2009;62:106–115. 111:209–219.
63. Poduslo SE, Jang Y. Myelin development in infant brain. Biochem Res 89. Mukherjee P, Miller J, Shimony J, et al. Normal brain maturation during
1984;9:1615–1626. childhood: developmental trends characterized with diffusion-tensor MR
64. Kucharczyk W, Macdonald P, Stanisz G, Henkelman RM. Relaxivity and imaging. Radiology 2001;221:349–358.
magnetization transfer of white matter lipids at MR imaging: importance 90. Mukherjee P, Miller J, Shimony J, et al. Diffusion-tensor MR imaging of
of cerebrosides and pH. Radiology 1994;192:521–529. gray and white matter development during normal human brain matura-
65. Nave KA, Trapp BD. Axon-glial signaling and the glial support of axon tion. Am J Neuroradiol 2002;23:1445–1456.
function. Annu Rev Neurosci 2008;31:535–561. 91. Dubois J, Dehaene-Lambertz G, Perrin M, et al. Asynchrony of the early
66. Koenig SH, Brown RD, III, Spiller M, Lundbom N. Relaxometry of the maturation of white matter bundles in healthy infants: Quantitative land-
brain: why white matter appears bright on MRI. Mag Res Med 1990;14: marks revealed noninvasively by diffusion tensor imaging. Human Brain
482–495. Mapping 2008;29(1):14–27.
67. Baratti C, Barnett AS, Pierpaoli C. Comparative MR imaging study of 92. Gao W, Lin W, Chen Y, et al. Temporal and spatial development of axonal
brain maturation in kittens with T1, T2, and the trace of the diffusion maturation and myelination of white matter in the developing brain. Am
tensor. Radiology 1999;210:133–142. J Neuroradiol 2009;30:290–296.
68. Husted C, Montez B, Le C, Moscarello MA, Oldfield E. Carbon-13 “magic- 93. Tessier-Lavigne M, Goodman CS. The molecular biology of axon guidance.
angle” sample-spinning nuclear magnetic resonance studies of human Science 1996;274:1123–1133.
myelin, and model membrane systems. Mag Res Med 1993;29:168–178. 94. Shu T, Richards L. Cortical axon guidance by the glial wedge during the
69. Husted C. Carbon-13 Magic Angle Spinning NMR Studies of Myelin development of the corpus callosum. J Neurosci 2001;21:2749–2758.
Membranes. [Ph.D.], University of Illinois at Urbana-Champaign, 1991. 95. Serafini T, Colamarino S, Leonardo E, et al. Netrin-1 is required for com-
70. Barkovich AJ. Brain development: normal and abnormal. In: Atlas SW, ed. missural axon guidance in the developing vertebrate nervous system. Cell
Magnetic Resonance Imaging of the Brain and Spine. New York: Raven, 1996;87:1001–1014.
1991:129–175. 96. Rash BG, Richards LJ. A role for cingulate pioneering axons in the
71. Barkovich AJ. Concepts of myelin and myelination in neuroradiology. Am development of the corpus callosum. J Comp Neurol 2001;434:147–157.
J Neuroradiol 2000;21:1099–1109. 97. Raybaud C. Corpus callosum, other great cerebral commissures, septum
72. Lee J, Barry JA. Influence of membrane lipid packing on T2-weighted pellucidum: anatomy, development and malformation. Neuroradiology
magnetic resonance images: study of relaxation parameters in model 2010;52(6):447-477.
membrane systems. Magn Reson Med 1996;36:420–426. 98. Richards LJ, Plachez C, Ren T. Mechanisms regulating the development
73. Kosaras B, Kirschner DA. Radial component of CNS myelin: junctional of the corpus callosum and its agenesis in mouse and human. Clin Genet
subunit structure and supramolecular assembly. J Neurocytol 1990;19: 2004;66:276–289.
187–199. 99. Shu T, Li Y, Keller A, Richards LJ. The glial sling is a migratory population
74. Braun PE. Molecular organization of myelin. In: Morell P, ed. Myelin, of developing neurons. Development 2003;130:2929–2937.
2nd ed. New York: Plenum, 1984:97–116. 100. Shu T, Sundaresan V, McCarthy M, Richards L. Slit2 guides both pre-
75. Morell P, Quarles RH, Norton WT. Myelin formation, structure, and bio- crossing and postcrossing callosal axons at the midline in vivo. J Neurosci
chemistry. In: Siegel GJ, ed. Basic Neurochemistry: Molecular, Cellular, and 2003;23:8176–8184.
Medical Aspects, 5th ed. New York: Raven, 1994:117–143. 101. Rakic P, Yakovlev PI. Development of the corpus callosum and cavum
76. Johnson MA, Peccock JM, Bydder GM, et al. Clinical NMR of the brain in septae in man. J Comp Neurol 1968;132:45–72.
children: normal and neurologic disease. AJR 1983;141:1005–1018. 102. Anderson NG, Laurent I, Cook N, Woodward L, Inder TE. Growth rate
77. Holland BA, Haas DK, Norman D, Brant-Zawadzki M, Newton TH. MRI of corpus callosum in very premature infants. Am J Neuroradiol 2005;
of normal brain maturation. Am J Neuroradiol 1986;7:201–208. 26:2685–2690.
78. Christophe C, Muller MF, Baleriaux D, et al. Mapping of normal brain 103. Barkovich AJ, Kjos BO. Normal postnatal development of the corpus callo-
maturation in infants on phase-sensitive inversion-recovery images. sum as demonstrated by MR imaging. Am J Neuroradiol 1988;9:487–491.
Neuroradiology 1990;32(3):173. 104. DeLacoste MC, Kirkpatrick JB, Ross ED. Topography of the corpus cal-
79. Dietrich RB, Bradley WG, Zagaroza EJ, et al. MR evaluation of early myeli- losum. J Neuropath Exp Neurol 1985;44:578–591.
nation patterns in normal and developmentally delayed infants. Am J 105. Tovar-Moll F, Moll J, de Oliveira-Souza R, Bramati I, Andreiuolo PA, Lent R.
Neuroradiol 1988;9:69–76. Neuroplasticity in human callosal dysgenesis: a diffusion tensor imaging
80. Staudt M, Schropp C, Staudt F, Obletter N, Bise K, Breit A. Myelination of study. Cereb Cortex 2007;17:531–541.
the brain in MRI: a staging system. Pediatr Radiol 1993;23:169–176. 106. Liebman SD, Gellis SS. The Pediatrician’s Ophthalmology. St. Louis, :
81. Staudt M, Schropp C, Staudt F, et al. MRI assessment of myelination: an Mosby, 1966.
age standardization. Pediatr Radiol 1994;24:122–127. 107. Raybaud CA, Girard N. Étude anatomique par IRM des agénésies et dys-
82. Bird C, Hedberg M, Drayer BP, et al. MR assessment of myelination plasies commissurales télencéphaliques. Neurochirurgie 1998;44(Suppl 1):
in infants and children: usefulness of marker sites. Am J Neuroradiol 38–60.
1989;10:731–740. 108. McLeod NA, Williams JP, Machen B, Lum GB. Normal and abnormal
83. van der Knaap MS. Myelination and Myelin Disorders: A Magnetic Reso- morphology of the corpus callosum. Neurology 1987;37:1240–1242.
nance Study in Infants, Children and Young Adults. [Ph.D.], Netherlands: 109. Rauch RA, Jinkins JR. Analysis of cross-sectional area measurements of
Free University of Amsterdam and Universtiy of Utrecht, 1991. the corpus callosum adjusted for brain size in male and female subjects
84. Martin E, Boesch C, Zuerrer M, et al. MR imaging of brain maturation from childhood to adulthood. Behav Brain Res 1994;64:65–78.
in normal and developmentally handicapped children. J Comput Assist 110. Wolpert XM, Osborne M, Anderson M, Runge VM. Bright pituitary gland:
Tomogr 1990;14(5):685–692. a normal MR appearance in infancy. Am J Neuroradiol 1988;9:1–3.

Barkovich_Chap02.indd 76 5/7/2011 2:42:55 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 77

111. Cox TD, Elster AD. Normal pituitary gland: changes in shape, size, and 135. Sarwar M, Kier EL, Varapongse C. Development of the spine and spinal
signal intensity during the first year of life at MR imaging. Radiology cord. In: Newton TH, Potts DG, eds. Computed Tomography of the Spine
1991;179:721–724. and Spinal Cord, vol 1. San Anselmo, CA: Clavedel, 1983:15–30.
112. Dietrich RB, Lis LE, Greensite FS, Pitt D. Normal MR appearance of 136. Naidich T, McLone D. Growth and development. In: Kricun M, ed. Imaging
the pituitary gland in the first 2 years of life. Am J Neuroradiol 1995;16: Modalities in Spinal Disorders. Philadelphia: Saunders, 1988:1–19.
1413–1419. 137. Ho PSP, Yu S, Sether LA, Wagner M, Ho KC, Haughton VM. Progressive
113. Konishi Y, Kuriyama M, Sudo M, Hayakawa K, Konishi K, Nakamura K. and regressive changes in the nucleus pulposus. I. The neonate. Radiology
Growth patterns of the normal pituitary gland and in pituitary adenoma. 1988;169:87–91.
Dev Med Child Neurol 1990;32:69–73. 138. Guida G, Cigala F, Riccio V. The vascularization of the vertebral body in
114. Hayakawa K, Konishi Y, Matsuda T, et al. Development and aging of brain the human fetus at term. Clin Orthop 1969;65:229–234.
midline structures: assessment with MR imaging. Radiology 1989;172: 139. Ferguson W. Some observations on the circulation on foetal and infant
171–177. spines. J Bone Joint Surg (Am) 1950;32:640–648.
115. Kitamura E, Miki Y, Kawai M, et al. T1 signal intensity and height of 140. Zhang HMD, Sucato DJ, Nurenberg P, McClung A. Morphometric analy-
the anterior pituitary in neonates: correlation with postnatal time. Am J sis of neurocentral synchondrosis using magnetic resonance imaging in
Neuroradiol 2008;29:1257–1260. the normal skeletally immature spine. Spine 2010;35(1):76–82.
116. Fujisawa I, Asato R, Nishimura K, et al. Anterior and posterior lobes of the 141. Herman M, Pizzutillo P. Cervical spine disorders in children. Orthop Clin
pituitary gland: assessment by 1.5T MR imaging. J Comput Assist Tomogr North Am 1999;30:457–466.
1987;11:214–220. 142. Fauré C. Le rachis cervical de l’enfant. Les données de l’imagerie. Rivista di
117. Maghnie M, Arico M, Villa A, Genovese E, Beluffi G, Severi F. MR of Neuroradiol 1996;9:595–610.
the hypothalamic-pituitary axis in Langerhans cell histiocytosis. Am J 143. Rajwani T, Bhargava R, Moreau M, et al. MRI characteristics of the
Neuroradiol 1992;13:1365–1371. neurocentral synchondrosis. Pediatr Radiol 2002;32:811–816.
118. Maghnie M, Cosi G, Genovese E, et al. Central diabetes insipidus in chil- 144. Yamazaki A, Mason D, Caro P. Age of closure of the neurocentral cartilage
dren and young adults. N Eng J Med 2000;343:998–1007. in the thoracic spine. J Pediatr Orthop 1998;18:168–172.
119. Peyster RG, Hoover ED, Adler LP. CT of the normal pituitary stalk. Am J 145. Sze G, Baierl P, Bravo S. Evolution of the infant spinal column: evaluation
Neuroradiol 1984;5:45–47. with MR imaging. Radiology 1991;181:819–827.
120. Peyster RG, Hoover ED, Viscarello RR, et al. CT appearance of the ado- 146. Sze G, Bravo S, Baierl P, Shimkin PM. Developing spinal column:
lescent and preadolescent pituitary gland. Am J Neuroradiol 1983;4: gadolinium-enhanced MR imaging. Radiology 1991;180:497–502.
411–414. 147. Barkovich AJ, Baranski K, Vigneron D, et al. Proton MR spectroscopy
121. Elster AK, Chen MYM, Williams DW, III, Key LL. Pituitary gland: MR in the evaluation of asphyxiated term neonates. Am J Neurorad 1999;20:
imaging of physiologic hypertrophy in adolescence. Radiology 1990; 1399–1405.
174:681–685. 148. Barkovich A, Westmark K, Bedi H, Partridge J, Ferriero D, Vigneron D.
122. Yonetsu K, Watanabe M, Nakamura T. Age-related expansion and reduc- Proton spectroscopy and diffusion imaging on the first day of life after per-
tion in aeration of the sphenoid sinus: volume assessment by helical CT inatal asphyxia: preliminary report. Am J Neuroradiol 2001;22:1786–1794.
scanning. Am J Neuroradiol 2000;21:179–182. 149. Barkovich AJ. MR imaging of the neontal brain. Neuroimaging Clin N Am
123. Applegate GR, Hirsch WL, Applegate LJ, Curtin HD. Variability in the 2006;16(1):117–135.
enhancement of the normal central skull base in children. Neuroradiology 150. Barkovich AJ, Miller SP, Bartha A, et al. MRI, MRS, and DTI of sequential
1992;34:217–221. studies in neonates with encephalopathy. Am J Neuroradiol 2006;27:533–547.
124. Aoki S, Dillon WP, Barkovich AJ, Norman D. Marrow conversion prior to 151. Pirzkall A, Li X, Oh J, et al. 3D MRSI for resected high-grade gliomas
pneumatization of the spenoid sinus: assessment with MR imaging. Radi- before RT: tumor extent according to metabolic activity in relation to
ology 1989;172:373–375. MRI. Int J Radiat Oncol Biol Phys 2004;59:126–137.
125. Okada Y, Aoki S, Barkovich AJ, et al. Cranial bone marrow in children: 152. Bartha AI, Yap KR, Miller SP, et al. The normal neonatal brain: MR imag-
assessment of normal development with MR imaging. Radiology 1989; ing, diffusion tensor imaging, and 3D MR spectroscopy in healthy term
171:161–164. neonates. Am J Neuroradiol 2007;28(6):1015–1021.
126. Okamoto K, Ito J, Tokiguchi S, Furusawa T. High resolution CT findings in 153. Kreis R, Ross BD, Farrow NA, Ackerman Z. Metabolic disorders of the brain
the development of the sphenooccipital synchondrosis. Am J Neuroradiol in chronic hepatic encephalopathy detected with H-1 MR spectroscopy.
1996;17:117–120. Radiology 1992;182:19–27.
127. Madeline LA, Elster AD. Suture closure in the human chondrocranium: 154. Kreis R, Ross BD. Cerebral metabolic disturbances in patients with subacute
CT assessment. Radiology 1995;196:747–756. and chronic diabetes mellitus: detection with proton MR spectroscopy.
128. Gooding C. Cranial sutures and fontanelles. In: Newton T, Potts D, eds. Radiology 1992;184:123–130.
Radiology of the Skull and Brain. St. Louis: Mosby, 1971:216–240. 155. Benarroch EEMD. N-Acetylaspartate and N-acetylaspartylglutamate:
129. Vu H, Panchal J, Parker E, Levine N, Francel P. The timing of physiologic Neurobiology and clinical significance [Miscellaneous Article]. Neurology
closure of the metopic suture: a review of 159 patients using reconstructed 2008;70(16):1353–1357.
3D CT scans of the craniofacial region. J Craniofac Surg 2001;12:527–532. 156. Burri R, Steffen C, Herschkowitz N. N-acetyl-L-aspartate is a major source
130. Hasso AN, Vignaud J. Normal anatomy of the paranasal sinuses, nasal cav- of acetyl groups for lipid synthesis during rat brain development. Dev
ity and facial bones. In: Newton TH, Hasso AN, Dillon WP, eds. Computed Neurosci 1991;13(6):403–422.
Tomography of the Head and Neck, vol 3. New York: Raven, 1988:6.1–6.18. 157. Ross BD, Bluml S. New aspects of brain physiology. NMR Biomed 1996;
131. Scuderi A, Harnsberger HR, Boyer RS. Pneumatization of the paranasal 9:279–296.
sinuses: normal features of importance to the accurate interpretation of 158. Hüppi PS, Posse S, Lazeyras F, et al. Developmental changes im 1H spectros-
CT scans and MR images. Am J Roentgenol 1993;160:1101–1104. copy in human brain. In: Lafeber HN, ed. Fetal and Neonatal Physiological
132. Bizzi A, Brooks RA, Brunetti A, et al. Role of iron and ferritin in MR imag- Measurements. Elsevier Science Publishers B.V. 1991:33.
ing of the brain: a study in primates at different field strengths. Radiology 159. Hüppi PS, Boesch C, Fusch C, et al. Concentrations of cerebral metabolites
1990;177:59–65. in the developing human brain: comparison of autopsy data (HPLC) and
133. Chen JC, Hardy PA, Kucharczyk W, et al. MR of human postmortem brain in vivo 1H-MRS data. In Society of Magnetic Resonance in Medicine, Elev-
tissue: correlative study between T2 and assays of iron and ferritin in enth Annual Scientific Meeting, August 8–14, Berlin, Germany. 1992;1:231.
Parkinson and Huntington disease. Am J Neuroradiol 1993;14:275–281. 160. Miyake M, Kakimoto Y. Developmental changes of N-acetyl-L-aspartic
134. Aoki S, Okada Y, Nishimura K, et al. Normal deposition of brain iron acid, N-acetyl-L-aspartylglutamic acid, and beta-citryl-L-glutamic acid in
in childhood and adolescence: MR imaging at 1.5T. Radiology 1989;172: different brain regions and spinal cords of rat and guinea pig. J Neurochem
381–385. 1981;37:1064–1067.

Barkovich_Chap02.indd 77 5/7/2011 2:42:55 AM


78 Pediatric Neuroimaging

161. Alonso E, Garcia-Perez A, Bueso J, Rubio V. N-acetyl-L-glutamate in brain: 184. Erecinska M, Silver I. Metabolism and role of glutamate in mammalian
assay, levels, and regional and subcellular distribution. Neurochem Res brain. Prog Neurobiol 1990;35:245–296.
1991;16:787–794. 185. Johnston MV. Neurotransmitters and vulnerability of the developing
162. Matalon R, Michals K, Sebesta D, et al. Aspartoacylase deficiency and brain. Brain Dev 1995;17:301–306.
N-acetylaspartic aciduria in patients with Canavan disease. Am J Med 186. McDonald JW, Johnston MV. Physiological and pathophysiological roles
Genet 1988;29:463–471. of excitatory amino acids during central nervous system development.
163. Birken DL, Oldendorf WH. N-acetyl-L-aspartic acid: a literature review Brain Res Rev 1990;15:41–70.
of a compound prominent in 1H-NMR spectroscopic studies of brain. 187. Kreis R, Ernst T, Ross BD. Absolute quantitation of water and metabolites
Neurosci Biobehav Rev 1989;13:23–31. in the human brain. II. Metabolite concentrations. J Magn Res 1993;
164. Higuchi T, Graham SH, Fernandez E, et al. Effects of severe global isch- 102:9–15.
emia in N-acetylaspartate and other metabolites in the rat brain. Magn 188. Ross BD. Biochemical considerations in 1H spectroscopy. Glutamate and
Reson Med 1997;37:851–857. glutamine; myo-inositol and related metabolites. NMR Biomed 1991;
165. van der Knaap MS, Valk J. Magnetic Resonance of Myelin, Myelination, and 4:59–63.
Myelin Disorders, 2nd ed. Berlin: Springer, 1995. 189. Weisinger H. Myo-inositol transport in mouse astroglia-rich primary
166. Krägeloh-Mann I, Grodd W, Niemann G, Haas G, Ruitenbeek W. cultures. J Neurochem 1991;56:1698–1709.
Assessment and therapy monitoring of Leigh disease by MRI and proton 190. Michaelis T, Helms KD, Merboldt W, Haenicke W, Bruhn H, Frahm J. First
spectroscopy. Pediatr Neurol 1992;8(1):60–64. observation of scyllo-inositol in proton NMR spectra of human brain
167. Bruhn H, Kruse B, Korenke GC, et al. Proton NMR spectroscopy of cere- in vitro and in vivo. Paper presented at: Society of Magnetic Resonance in
bral metabolic alterations in infantile peroxisomal disorders. J Comput Medicine, Berlin. 1992.
Assist Tomogr 1992;16:335–344. 191. Gruetter R, Rothman DL, Novotny EJ, Shulman GI, Prichard JW, Shulman
168. De Stefano N, Matthews PM, Arnold DL. Reversible decreases in RG. Detection and assignment of the glucose signal in H-1 NMR difference
N-acetylaspartate after acute brain injury. Magn Reson Med 1995;34: spectra of the human brain. Magn Res Med 1992;27:183–188.
721–727. 192. Govindaraju V, Young K, Maudsley AA. Proton NMR chemical shifts and
169. Hugg JW, Kuzniecky RI, Gilliam FG, Morawetz RB, Faught RE, Hethering- coupling constants for brain metabolites. NMR Biomed 2000;13:129–153.
ton HP. Normalization of contralateral metabolic function following tem- 193. Panigrahy A, Krieger MD, Gonzalez-Gomez I, et al. Quantitative short
poral lobectomy demonstrated by H-1 magnetic resonance spectroscopic echo time 1H-MR spectroscopy of untreated pediatric brain tumors:
imaging. Ann Neurol 1997;40:236–239. preoperative diagnosis and characterization. Am J Neuroradiol 2006;
170. Vion-Dury J, Nicoli F, Salvan AM, Confort-Gouny S, Dhiver C, Cozzone 27(3):560–572.
PJ. Reversal of brain metabolic alterations with zidovudine detected by 194. Pettegrew JW, Panchalingam S, Withers G, McKeag D, Strychor S. Changes
proton localised magnetic resonance spectroscopy [letter]. Lancet 1995; in brain energy and phospholipid metabolism during development and
345:60–61. aging in the Fischer 344 rat. J Neuropathol exp Neurol 1990;49(3):237.
171. Urenjak J, Williams SR, Gadian DG, Noble M. Specific expression of 195. Azzopardi D, Edwards D. Magnetic resonance spectroscopy in neonates.
N-acetylaspartate in neurons, oligodendrocyte-type-2 astrocyte pro- Curr Opin Neurol 1995;8:145–149.
genitors, and immature oligodendrocytes in vitro. J Neurochem 1992; 196. Nioka S, Zaman A, Yoshioka H, et al. 31P magnetic resonance spectroscopy
59(1):55–61. study of cerebral metabolism in developing dog brain and its relationship
172. Blüml S, Seymour K, Ross B. Developmental changes in choline- and to neuronal function. Dev Neurosci 1991;13(2):61.
ethanolamine-containing compounds measured with proton-decoupled 197. Kiernan JA. Barr’s The Human Nervous System, 7th ed. Philadelphia:
(31)P MRS in in vivo human brain. Magn Res Med 1999;42:643–654. Lippincott-Raven, 1998.
173. Brand A, Richter-Landsberg C, Leibfritz D. Multinuclear NMR stud- 198. Choi C-G, Ko T-S, Lee HK, Lee JH, Suh DC. Localized proton MR
ies on the energy metabolism of glial and neuronal cells. Dev Neurosci spectroscopy of the allocortex and isocortex in healthy children. Am J
1993;15:289–298. Neuroradiol 2000;21:1354–1358.
174. Moore GJ. Proton magnetic resonance spectroscopy in pediatric neurora- 199. Vigneron DB, Barkovich AJ, Noworolski SM, et al. Three-dimensional
diology. Pediatr Radiol 1998;28:805–814. proton MR spectroscopic imaging of premature and term neonates. Am J
175. Govindaraju V, Basus V, Matson G, Maudsley A. Measurement of chemical Neuroradiol 2001;22:1424–1433.
shifts and coupling constants for glutamate and glutamine. Magn Res Med 200. Costa M, Lacerda M, Garcia Otaduy M, Cerri G, Da Costa Leite C.
1998;39:1011–1013. Proton magnetic resonance spectroscopy: normal findings in the cerebel-
176. Connelly A, Cross J, Gadian D, Hunter J, Kirkham F, Leonard J. Magnetic lar hemisphere in childhood. Pediatr Radiol 2002;32:787–792.
resonance spectroscopy shows increased brain glutamate in ornithine 201. Tedeschi G, Righini A, Bizzi A, Barnett A, Alger J. Cerebral white matter
carbamoyl transferase deficiency. Pediatr Res 1993;33:77–81. in the centrum semiovale exhibits a larger N-acetyl signal than does gray
177. Kreis R, Ernst T, Ross BD. Development of the human brain: in vivo matter in long echo time 1H-magnetic resonance spectroscopy imaging.
quantification of metabolite and water content with proton magnetic Magn Reson Med 1995;33:127–133.
resonance spectroscopy. Magn Res Med 1993;30:424–437. 202. van der Knaap MS, van der Grond J, van Rijen PC, Faber JAJ, Valk J,
178. Michaelis T, Merboldt K-D, Bruhn H, Hänicke W, Frahm J. Absolute Willemse K. Age-dependent changes in localized proton and phosphorus
concentrations of metabolites in the adult human brain in vivo: quantifi- MR spectroscopy of the brain. Radiology 1990;176:509–515.
cation of localized proton MR spectra. Radiology 1993;187:219–227. 203. Frahm J, Hanefeld F. Localized proton magnetic resonance spectroscopy
179. Urenjak J, Williams SR, Gadian DG, Noble M. Proton nuclear magnetic of cerebral metabolites. Neuropediatrics 1996;27:64–69.
resonance spectroscopy unambiguously identifies different neural cell 204. Kok R, van den Berg P, van den Bergh A, Nijland R, Heerschap A. Matura-
types. J Neurosci 1993;13:981–989. tion of the human fetal brain as observed by 1H MR spectroscopy. Magn
180. McGuinness GA, Weisz SC, Bell WE. CSF lactate levels in neonates. Effects Reson Med 2002;48(4):611–616.
of asphyxia, gestational age, and postnatal age. Am J Dis Child 1983; 205. Kreis R, Hofmann L, Kuhlmann B, Boesch C, Bossi E, Hüppi P. Brain
137:48–50. metabolite composition during early human brain development as
181. Fernandez F, Verdu A, Quero J, et al. Cerebrospinal fluid lactate levels in measured by quantitative in vivo 1H magnetic resonance spectroscopy.
term infants with perinatal hypoxia. Pediatr Neurol 1986;2:39–42. Magn Reson Med 2002;48:949–958.
182. Leth H, Toft PB, Pryds O, Petersen B, Lou HC, Henriksen O. Brain lactate 206. Hida K. In vivo 1H and 31P NMR spectroscopy of the developing rat brain.
in preterm and growth-retarded neonates. Acta Pediatr 1995;84:495–499. Hokkaido J Med Sci 1992;67(2):272.
183. Cady EB, Lorek A, Penrice J, et al. Detection of propan-1,2-diol in neona- 207. Hennig J, Pfister H, Ernst T, Ott D. Direct absolute quantification of
tal brain by in vivo proton magnetic resonance spectroscopy. Magn Reson metabolites in the human brain with in vivo localized proton spectroscopy.
Med 1994;32:764–767. NMR Biomed 1992;5:193–199.

Barkovich_Chap02.indd 78 5/7/2011 2:42:55 AM


Chapter 2 • Normal Development of the Neonatal and Infant Brain, Skull, and Spine 79

208. Hüppi PS, Posse S, Lazeyras F, Burri R, Bossi E, Herschkowitz N. Magnetic 230. Partridge SC, Mukherjee P, Henry R, et al. Diffusion tensor imaging:
resonance in preterm and term newborns: 1-H spectroscopy in developing serial quantitation of white matter tract maturity in premature newborns.
human brain. Pediatr Res 1991;30:574–578. NeuroImage 2004;22:1302–1314.
209. Mathur-De Vor R, Delporte C. Relaxation times of relevant nuclei in bio- 231. McGraw P, Liang L, Provenzale J. Evaluation of normal age-related changes
logical systems. In: De Certaines J, Bovee W, Podo F, eds. Magnetic Reso- in anisotropy during infancy and childhood as shown by diffusion tensor
nance Spectroscopy in Biology and Medicine. Oxford: Pergramon, 1992: imaging. Am J Roentgenol 2002;179:1515–1522.
77–96. 232. Klingberg TC, Vaidya J, Gabrieli JDE, Moseley ME, Hedehus M. Myelina-
210. Ernst T, Kreis R, Ross BD. Absolute quantitation of water and metabolites tion and organization of the frontal white matter in children: a diffusion
in the human brain. I. compartments and water. J Magn Res 1993;102:1–8. tensor MRI study. Neuroreport 1999;10:2817–2821.
211. Kugel H, Roth B, Wittsack H-J, Heindel W, Ernst S, Lackner K. Correlation 233. Nomura Y, Sakuma H, Takeda K, Tagami T, Okuda Y, Nakagawa T. Dif-
of quantitative spectroscopic data and clinical outcome in infants with fusional anisotropy of the human brain assessed with diffusion-weighted
cerebral sequelae of asphyxia or disorders of energy metabolism. Paper MR: relation with normal brain development and aging. Am J Neuroradiol
presented at: Society of Magnetic Resonance, San Francisco, CA, 1994. 1994;15:231–238.
212. Lu D, Pavlakis SG, Frank Y, et al. Proton MR spectroscopy of the basal 234. Toft PB, Leth H, Peitersen B, Lou HC, Thomsen C. The apparent diffusion
ganglia in healthy children and children with AIDS. Radiology 1996; coefficient of water in gray and white matter of the infant brain. J Comput
199:423–428. Assist Tomogr 1996;20:1006–1011.
213. Hüppi PS, Fusch C, Boesch C, et al. Regional metabolic assessment of 235. Wimberger D, Roberts TP, Kucharczyk J, Barkovich AJ, Kozniewska E,
human brain during development by proton magnetic resonance spec- Prayer LM. Diffusion-weighted imaging at 4.7T in the assessment of brain
troscopy in vivo and by high-performance liquid chromatography/gas maturation in albino rats. Paper Presented at Society of Magnetic Resonance
chromatography in autopsy tissue. Pediatr Res 1995;37:145–150. Imaging, San Francisco, 1993.
214. Stejskal EO, Tanner JE. Spin diffusion measurements: spin echoes in 236. Beaulieu C, Allen PS. Diffusional anisotropy of water in nerve cords
the presence of a time-dependent field gradient. J Chem Phys 1965;42: without myelination. Paper Presented at Society of Magnetic Resonance in
288–292. Medicine, Berlin, 1992.
215. Stejskal EO. Use of spin echo in pulsed magnetic-field gradient to 237. McKinstry R, Mathur A, Miller J, et al. Radial organization of developing
study anisotropic, restricted diffusion and flow. J Chem Phys 1965;43: preterm human cerebral cortex revealed by non-invasive water diffusion
3597–3603. anisotropy MRI. Cereb Cortex 2002;12:1237–1243.
216. Sakuma H, Nomura Y, Takeda K, et al. Adult and neonatal human brain: 238. DeIpolyi AR, Mukherjee P, Gill K, et al. Comparing microstructural and
diffusional anisotropy and myelination with diffusion-weighted MR macrostructural development of the cerebral cortex in premature new-
imaging. Radiology 1991;180:229–233. borns: diffusion tensor imaging versus cortical gyration. NeuroImage 2005;
217. Chenevert TL, Brunberg JA, Pipe JG. Anisotropic diffusion in human 27:579–586.
white matter: demonstration with MR techniques in vivo. Radiology 1990; 239. Maas L, Mukherjee P, Carballido-Gamio J, et al. Early laminar organiza-
177:401–405. tion of the human cerebrum demonstrated with diffusion tensor imaging
218. Drobyshevsky A, Song S-K, Gamkrelidze G, et al. Developmental changes in extremely premature infants. NeuroImage 2004;22:1134–1140.
in diffusion anisotropy coincide with immature oligodendrocyte pro- 240. Basser PJ, Pajevic S, Pierpaoli C, Duda J, Aldroubi A. In vivo fiber
gression and maturation of compound action potential. J Neurosci 2005; tractography using DT-MRI data. Magn Reson Med 2000;44(4):625–632.
25:5988–5997. 241. Mori S, Crain BJ, Chacko VP, Zijl PCMV. Three-dimensional tracking
219. Beaulieu C. The basis of anisotropic water diffusion in the nervous of axonal projections in the brain by magnetic resonance imaging. Ann
system – a technical review. NMR Biomed 2002;15:435–455. Neurol 1999;45(2):265–269.
220. Prayer D, Barkovich AJ, Kirschner DA, et al. Visualization of nonstructural 242. Conturo TE, Lori NF, Cull TS, et al. Tracking neuronal fiber path-
changes in early white matter development on diffusion weighted MR ways in the living human brain. Proc Natl Acad Sci USA 1999;96(18):
images: evidence supporting premyelination anisotropy. Am J Neuroradiol 10422–10427.
2001;22:1572–1576. 243. Kier EL, Staib LH, Davis LM, Bronen RA. Anatomic dissection tractogra-
221. Wimberger DM, Roberts TP, Barkovich AJ, Prayer LM, Moseley ME, phy: a new method for precise MR localization of white matter tracts. Am
Kucharczyk J. Identification of “premyelination” by diffusion-weighted J Neuroradiol 2004;25(5):670–676.
MRI. J Comput Assist Tomogr 1995;19:28–33. 244. Wakana S, Caprihan A, Panzenboeck MM, et al. Reproducibility of quanti-
222. Robertson R, Ben-Sira L, Barnes P, et al. MR line-scan diffusion-weighted tative tractography methods applied to cerebral white matter. NeuroImage
imaging of term neonates with perinatal brain ischemia. Am J Neuroradiol 2007;36(3):630–644.
1999;20:1658–1670. 245. Danielian LE, Iwata NK, Thomasson DM, Floeter MK. Reliability of fiber
223. Cowan FM, Pennock JM, Hanrahan JD, Manji KP, Edwards AD. Early tracking measurements in diffusion tensor imaging for longitudinal study.
detection of cerebral infarction and hypoxic ischemic encephalopa- NeuroImage 2010;49(2):1572–1580.
thy in neonates using diffusion weighted magnetic resonance imaging. 246. Partridge SC, Mukherjee P, Berman JI, et al. Tractography-based quan-
Neuropediatrics 1994;25:172–175. titation of diffusion tensor imaging parameters in white matter tracts of
224. Engelbrecht V, Scherer A, Rassek M, Witsack H, Modder U. Diffusion- preterm newborns. J Magn Reson Imag 2005;22(4):467–474.
weighted MR imaging in the brain in children: findings in the normal brain 247. Berman JI, Mukherjee P, Partridge SC, et al. Quantitative diffusion ten-
and in the brain with white matter diseases. Radiology 2002;222:410–418. sor MRI fiber tractography of sensorimotor white matter development in
225. Forbes K, Pipe J, Bird C. Changes in brain water diffusion during the premature infants. NeuroImage 2005;27(4):862–871.
1st year of life. Radiology 2002;222:405–409. 248. Berman JI, Glass HC, Miller SP, et al. Quantitative fiber tracking analysis
226. Neil JJ, Shiran SI, McKinstry RC, et al. Normal brain in human newborns: of the optic radiation correlated with visual performance in premature
apparent diffusion coefficient and diffusion anisotropy measured by using newborns. Am J Neuroradiol 2009;30:120–124.
diffusion tensor MR imaging. Radiology 1998;209:57–66. 249. Lebel C, Caverhill-Godkewitsch S, Beaulieu C. Age-related regional varia-
227. Papadakis NG, Xing D, Houston GC, et al. A study of rotationally invari- tions of the corpus callosum identified by diffusion tensor tractography.
ant and symmetric indices of diffusion anisotropy. Magn Reson Imaging NeuroImage 2010;52:20-31.
1999;17:881–892. 250. Westlye LT, Walhovd KB, Dale AM, et al. Differentiating maturational and
228. Morriss M, Zimmerman R, Bilaniuk L, Hunter J, Haselgrove J. Changes in aging-related changes of the cerebral cortex by use of thickness and signal
brain water during childhood. Neuroradiology 1999;41:929–934. intensity. NeuroImage 2010;52:172-85.
229. Hüppi P, Maier S, Peled S, et al. Microstructural development of human 251. Engelbrecht V, Rassek M, Preiss S, Wald C, Mödder U. Age-dependent
newborn cerebral white matter assessed in vivo by diffusion tensor changes in magnetization transfer contrast of white matter in the pediatric
magnetic resonance imaging. Pediatr Res 1998;44:584–590. brain. Am J Neuroradiol 1998;19:1923–1929.

Barkovich_Chap02.indd 79 5/7/2011 2:42:55 AM


80 Pediatric Neuroimaging

252. Ceckler TL, Wolff SD, Yip V, Simon SA, Balaban RS. Dynamic and chemical 263. Biswal B, Yetkin FZ, Haughton VM, Hyde JS. Functional connectivity in
factors affecting water proton relaxation by macromolecules. J Magn Res the motor cortex of resting human brain using echo-planar MRI. Magn
1992;98:637–645. Reson Med 1995;34(4):537–541.
253. Fralix TA, Ceckler TL, Wolff SD, Simon SA, Balaban RS. Lipid bilayer and 264. McKeown MJ, Sejnowski TJ. Independent component analysis of
water proton magnetization transfer: effect of cholesterol. Magn Res Med fMRI data: examining the assumptions. Hum Brain Mapp 1998;6(5–6):
1991;18:214–223. 368–372.
254. Dousset V, Grossman RI, Ramer KN, et al. Experimental allergic 265. Kiviniemi V, Starck T, Remes J, et al. Functional segmentation of the
encephalomyelitis and multiple sclerosis: lesion characterization with brain cortex using high model order group PICA. Hum Brain Mapp 2009;
magnetization transfer imaging. Radiology 1992;182:483–491. 30(12):3865–3886.
255. Chew WM, Rowley HA, Barkovich AJ. Magnetization transfer contrast 266. Smith JF, Pillai A, Chen K, Horwitz B. Identification and validation of
imaging in pediatric patients. Radiology 1992;185(P):281. effective connectivity networks in functional magnetic resonance imaging
256. van Buchem MA, Steens SCA, Vrooman HA, et al. Global estimation of using switching linear dynamic systems. NeuroImage 2010;52:1027-1040.
myelination in the developing brain of the basis of magnetization transfer 267. Raichle ME. Cognitive neuroscience. Bold insights. Nature 2001;412:
imaging: a preliminary study. Am J Neuroradiol 2001;22:762–766. 128–130.
257. Villringer A, Rosen RB, Belliveau JW, et al. Dynamic imaging with lan- 268. Greicius MD, Krasnow B, Reiss AL, Menon V. Functional connectivity in
thanide chelates in normal brain: contrast due to magnetic susceptibility the resting brain: A network analysis of the default mode hypothesis. Proc
effects. Magn Res Med 1988;6:164–174. Natl Acad Sci USA 2003;100(1):253–258.
258. Laswad T, Wintermark P, Alamo L, Moessinger A, Meuli R, Gudinchet F. 269. Fransson P, Skiold B, Horsch S, et al. Resting-state networks in the infant
Method for performing cerebral perfusion-weighted MRI in neonates. brain. Proc Natl Acad Sci 2007;104(39):15531–15536.
Pediatr Radiol 2009;39(3):260–264. 270. Smyser CD, Inder TE, Shimony JS, et al. Longitudinal analysis of neural net-
259. Petersen ET, Lim T, Golay X. Model-free arterial spin labeling quantification work development in preterm infants. Cereb Cortex 2010;20:2852-2862.
approach for perfusion MRI. Magn Reson Med 2006;55:219–232. 271. Gao W, Zhu H, Giovanello KS, et al. Evidence on the emergence of the
260. Biagi L, Abbruzzese A, Bianchi M, Alsop D, Del Guerra A, Tosetti M. brain’s default network from 2-week-old to 2-year-old healthy pediatric
Age dependence of cerebral perfusion assessed by magnetic resonance subjects. Proc Natl Acad Sci 2009;106(16):6790–6795.
continuous arterial spin labeling. J Magn Reson Imaging 2007;25:696–702. 272. Supekar K, Musen M, Menon V. Development of large-scale functional
261. Ogawa S, Lee TM, Kay AR, Tank DW. Brain magnetic resonance imaging brain networks in children. PLoS Biol 2009;7(7):e1000157.
with contrast dependent on blood oxygenation. Proc Natl Acad Sci USA 273. Fair DA, Cohen AL, Power JD, et al. Functional brain networks develop
1990;87:9868–9872. from a “local to distributed” organization. PLoS Comput Biol 2009;5(5):
262. Bandettini PA. Seven topics in functional magnetic resonance imaging. e1000381.
J Integr Neurosci 2009;8:371–403.

Barkovich_Chap02.indd 80 5/7/2011 2:42:55 AM


CHAPTER

Metabolic, Toxic, and


3 Inflammatory Brain Disorders
A. JAMES BARKOVICH AND ZOLTAN PATAY

Introduction Metabolic and neurodegenerative disorders primarily


Imaging techniques in metabolic disorders involving the cerebellum
A simple pattern approach to metabolic diseases Differentiation of cerebellar atrophy from hypoplasia
White matter versus gray matter Cerebellar ataxias
Gray matter disorders Cerebellar atrophy
White matter disorders Cerebellar atrophy with adolescent/young adult onset
Disorders affecting gray and white matter Cerebellar hypoplasia
Deeper analysis of toxic/metabolic/inflammatory diseases
A Few comments about classifi cations
Clinical aspects of metabolic diseases
Deeper analysis of gray and white matter diseases
Diseases primarily affecting white matter
Metabolic disorders primarily affecting gray matter
Metabolic disorders affecting both gray and white matter

diseases, and injury from radiation or chemotherapy, can be associ-


INTRODUCTION ated with similar histologic and imaging changes. These inflamma-
This chapter includes a very diverse group of brain disorders that have tory conditions are included in this chapter, as well. Infections are
a number of similarities, particularly in their imaging characteristics. discussed separately, in Chapter 11.
Inborn errors of metabolism are usually caused by a gene mutation The diagnosis of the disorders discussed in this chapter is challeng-
that results in formation of an abnormal protein, disrupting the func- ing for all concerned with the care of the affected child. The presenting
tion of one or more metabolic pathways. Some metabolic disorders signs and symptoms are usually nonspecific ones that may be acute,
have no neurological manifestations. Others present with signs and subacute, or slowly progressive; these may include hypotonia, seizures,
symptoms of central nervous system involvement exclusively; these are spasticity, ataxia, movement disorder, delay in achieving develop-
true neurometabolic disorders. Some others may have both systemic mental milestones, or focal neurologic deficits. The imaging study is
and central nervous system manifestations, and in a broader sense, if most often suggestive of a certain type of disorder—metabolic/toxic
the neurological implications of the disease are significant, these may or demyelinating—but rarely is diagnostic. Biochemical tests may be
also be referred to as neurometabolic disorders. normal or nonspecific, while genetic analyses are often unrewarding
In neurometabolic diseases, brain injury and clinical symptoms unless a specific genetic defect is suspected and sought. As a result, at
result from lack of production of a necessary biochemical or from least 60% of children with inborn metabolic errors never receive a spe-
formation of an abnormal biochemical that is, directly or indirectly, cific diagnosis despite extensive investigations (1,2). Even if a diagnosis
toxic to the brain. Sometimes the disruption of metabolism (usu- is made, the expectations for outcome may be uncertain, as the precise
ally fetal, occasionally maternal) affects in utero brain development, distinctions among, for example, the various mitochondrial disorders
resulting in a malformation (Table 3-1). Much more typically, how- and autoimmune inflammatory disorders are far from clear, especially
ever, toxic chemicals are removed and necessary chemicals supplied in children (3–9).
via the placenta; thus, brain injury does not begin until sometime In order to establish a diagnosis, some method of organizing the
after birth. Children with more profound enzymatic changes tend diseases from an imaging perspective is helpful, as narrowing the dif-
to have earlier clinical onset (neonatal, infantile), whereas those ferential diagnosis facilitates the clinical workup, saving time and
with milder biochemical phenotypes typically develop symptoms expense. In the future, all of these disorders may be diagnosed, classi-
later (juvenile, adult). Both endogenous (caused by inborn meta- fied, and treated according to the underlying genetic mutation and its
bolic errors) and exogenous (ingested, injected, or inhaled) toxins effect on metabolic pathways. However, our understanding of genet-
may interfere with normal brain metabolism in similar ways and, ics, epigenetic factors, and metabolic pathways is not yet good enough
therefore, cause similar patterns of brain damage; therefore, both are to accomplish these goals. Therefore, we are a long way from a useful
included in this chapter. Still other processes, such as autoimmune genetic classification of these diseases.

81

Barkovich_Chap03.indd 81 5/6/2011 12:07:52 AM


82 Pediatric Neuroimaging

TABLE 3-1 Malformations of the CNS Described in the Context


of Inborn Errors of Metabolism
Abnormalitya Metabolic Disorder
Cerebellar dysgenesis and 3-hydroxyisobutyric aciduria (hypoplasia)
other posterior fossa Bifunctional enzyme deficiency (dysplasia)
abnormalities Congenital disorders of glycosylation type 1a (cerebellar hypoplasia)
Folate and 5,10 methylene-tetrahydrofolate reductase deficiencies (in
maternal disease, Chiari type 2 malformation)
Fumarase deficiency (cerebellar dysplasia)
Glutaric aciduria type 2 (dysgenesis of cerebellar vermis)
Infantile Refsum disease (dysplasia)
Menkes disease (cerebellar dysplasia, vermian hypoplasia/agenesis)
Nonketotic hyperglycinemia (cerebellar hypoplasia)
Pseudoneonatal adrenoleukodystrophy (cerebellar dysplasia)
Pyruvate dehydrogenase deficiency (absence of pyramids,
heterotopia of the inferior olivary nuclei)
Respiratory chain deficiencies involving complex IV (hypoplasia
of cerebellar hemispheres with relative sparing of the vermis,
pontocerebellar hypoplasia)
Smith-Lemli-Opitz syndromeb (cerebellar hypoplasia, abnormal
foliation, small inferior cerebellar vermis)
Zellweger syndrome (cortical dysplasia)
Cerebral dysgenesis Glutaric acidemia type 2 (warty protrusion of the surface of the
cerebral cortex)
Neonatal carnitine palmitoyl-transferase 2 deficiency
Cerebral lobar hypoplasias Glutaric aciduria type 1 (frontotemporal)
Smith-Lemli-Opitz syndromeb (frontal lobe hypoplasia)
Corpus callosum abnormalityc 3-hydroxyisobutyric aciduria (agenesis)
Congenital disorders of glycosylation (dysgenesis, hypoplasia)
Dihydropyrimidine dehydrogenase deficiency (partial agenesis)
Fumarase deficiency (agenesis)
Glutaric aciduria type 2 (hypoplasia)
Infantile Refsum (dysgenesis)
Menkes disease (dysgenesis)
Neonatal lactic acidosis, complex I/IV deficiency, and fetal cerebral
disruption
Phenylketonuria (maternal, dysgenesis)
Pyruvate dehydrogenase deficiency (dysgenesis)
Smith-Lemli-Opitz syndromeb (absence, dysgenesis)
Zellweger syndrome (agenesis, partial dysgenesis)
Cortical dysplasia Peroxisomal disorders
Smith-Lemli-Opitz syndromeb
Dentate nuclei abnormalities Pyruvate dehydrogenase deficiency (dysplasia)
Zellweger syndrome (dysplasia)
Gray matter heterotopia/ Peroxisomal disorders
cortical heterotopia Carnitine palmitoyl transferase 2 deficiency
Fumarase deficiency (cerebral and cerebellar)
Glutaric acidemia type 2
Menkes disease
Neonatal lactic acidosis, complex I/IV deficiency, and fetal cerebral
disruption
Pyruvate dehydrogenase deficiency (subcortical heterotopia)
Smith-Lemli-Opitz syndromeb

Barkovich_Chap03.indd 82 5/6/2011 12:07:52 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 83

TABLE 3-1 Malformations of the CNS Described in the Context


of Inborn Errors of Metabolism (Continued)
Abnormalitya Metabolic Disorder
Holoprosencephaly Smith-Lemli-Opitz syndrome
Inferior olivary nucleus Bifunctional enzyme deficiency (dysplasia)
abnormalities Pontocerebellar hypoplasia type 2 (histologically)
Pontocerebellar hypoplasia type 4 and 5 (C-shaped inferior olivary
hypoplasia)
Pseudoneonatal adrenoleukodystrophy (dysplasia)
Pyruvate dehydrogenase deficiency (ectopic inferior olives)
Zellweger syndrome (dysplasia)
Lissencephaly 3-hydroxyisobutyric aciduria
Open opercula Fumarase deficiency
Glutaric aciduria type1
Pachygyria 3-Hydroxyisobutyric aciduria (pachygyria/lissencephaly)
Bifunctional enzyme deficiency (pachygyria/lissencephaly)
Glutaric aciduria type 2
Pyruvate dehydrogenase deficiency
Rhizomelic chondrodysplasia punctata
Smith–Lemli–Opitz syndromeb
Polymicrogyria Peroxisomal disorders
Fumarase deficiency
Smith-Lemli-Opitz syndrome
a
The number of reported cases of CNS malformations associated with metabolic disorders is small and are often
published as single case reports. Some of them, therefore, may be coincidental. As a general rule, metabolic disorders are
more likely to interfere with normal brain development if they start prenatally (e.g., peroxisomal biogenesis disorders,
nonketotic hyperglycinemia) or if they present as a maternal metabolic abnormality (e.g., hypovitaminosis).
b
Smith-Lemli-Opitz syndrome (OMIM 270400) is a recently described inborn error of cholesterol metabolism (biosyn-
thesis); indeed the first truly polymalformative metabolic disorders. The imaging diagnosis of this disease heavily relies
on the recognition of the peculiar constellation of multiple CNS malformations (see above).
c
Callosal hypoplasia nearly always accompanies prenatal, perinatal, or infantile white matter injury. Such cases are not
included.

In this chapter, the disorders are classified by the most typical MR of differential diagnoses for a particular pattern of brain involvement.
pattern of brain involvement, which frequently coincides with the ini- Please refer to Tables 3-3 to 3-5 when reading this initial section. The
tial clinical presentation. Discussion will center mainly on the pattern second part of the chapter discusses the same disorders in greater depth,
of brain involvement (“pattern recognition”), as detected by morpho- including some genetic, biochemical, and clinical information, as well
logic MRI, diffusion imaging, and proton MR spectroscopy (Table 3-2). as references for those interested in pursuing additional information
By proper neuroimaging analysis of the early pattern of brain involve- about the diseases. This second part follows the order of the outline at
ment, many disorders may be identified and differentiated from others the beginning of the book (the List of Diagnoses), not the order of Tables
with similar clinical presentations (2,10). It is important to recognize 3-3 to 3-5. Keep this in mind in order to avoid becoming confused. Also
that the patterns of injury are dynamic (they change over time). Ini- please remember that the discussions are intended to provide an over-
tial imaging findings may be very subtle or may not be detected until view of only the pertinent points of the disorders; a complete discussion
the disease progresses. Moreover, many toxic/metabolic/inflamma- of inborn errors of metabolism is well beyond the scope of this book.
tory brain disorders ultimately have a nonspecific imaging appearance Interested readers are referred to textbooks on the subject, such as those
(diffuse white matter injury or diffuse atrophy) in the late stages of the by van der Knaap and Valk (11) or Scriver et al. (12,13).
disease. Therefore, if an imaging study is to have a role in the diagnosis
of inborn metabolic errors, the study should be performed early in the IMAGING TECHNIQUES IN METABOLIC
course of the disease. Follow-up studies should be performed if the
disorder progresses.
DISORDERS
The first part of this chapter is a listing of many toxic/metabolic/ MRI is the modality that will be primarily discussed in this chapter,
inflammatory disorders based solely upon their imaging character- because the ability to detect subtle abnormalities of brain tissue is far
istics; this section should be useful for those who merely want a list superior with MRI than with CT or ultrasound. Ultrasound often

Barkovich_Chap03.indd 83 5/6/2011 12:07:52 AM


84 Pediatric Neuroimaging

TABLE 3-2 Separation of Types of White Matter Histopathology by MR


Hypomyelination Demyelination Myelin Vacuolization Cystic Degeneration Controls
MTR - - - -- NL
T2 + + ++ +++ NL
Dav NL + ++ +++ NL
FA - -- --- -- NL
MRS +Cr NL Cr --Cr --Cr NL Cr
NL NAA --NAA --NAA --NAA NL NAA
NL Ch +Ch -Ch --Ch NL Ch
++mI +++mI -mI --mI NL mI
+Lac ++Lac +Lac ++Lac No Lac
NL Glu -Glu --Glu ---Glu NL Glu
NL Gln ++Gln -Gln NL Gln NL Gln

MTR, magnetization transfer ratio; T2, T2 relaxivity; Dav, average diffusivity; FA, fractional anisotropy; MRS, MR spectroscopy; +, mildly increased; ++, moderately
increased; +++, markedly increased; -, mildly decreased; --, moderately decreased; ---, markedly decreased; Cr, Creatine; NAA, N-acetylaspartate; Ch, Choline;
mI, myo-inositol; Lac, lactate; Glu, glutamate; Gln, glutamine.
Source: After van der Voorn JP, Pouwels PJW, Hart AAM, et al. Childhood white matter disorders: quantitative MR imaging and spectroscopy. Radiology
2006;241(2):510–517.

shows abnormalities in metabolic disorders that have onset prenatally, studies for developmental delay or suspected metabolic disorder; faster is
in the neonatal period, or early in infancy. However, the findings are not always better! Most of the MR discussions in this chapter concern
nearly always nonspecific such as ventricular dilation, germinolytic findings on T2-weighted images; they apply to T2 FLAIR and RARE as
cysts, echogenic branching vessels in the basal ganglia (“lenticulostriate well as conventional spin-echo sequences.
vasculopathy”), or echogenic white matter (14), findings that are just Some authors have advocated the production of calculated T2
as likely to be seen in infection or injury. These findings are discussed maps of the brain and calculation of quantities such as relative water
when helpful. CT is sometimes useful to detect calcification and will be content and myelin water fraction as an approach to the study of white
mentioned when appropriate. matter disease (16,17). Although these methods may help determine
When performing morphologic MR scans of patients with the extent of myelin abnormality (16), they have not yet been shown
metabolic disorders, heavily T1-weighted images (inversion recov- to help in distinguishing among metabolic disorders; they are not
ery, spoiled gradient-echo images) are very useful to assess myelin discussed in this book.
injury and delayed or hypomyelination during the first year of life. In general, for morphological imaging. 3D volume acquisitions
T2-weighted images are usually much more useful than T1-weighted are useful but are somewhat hampered by inherent quality limitations.
images in the assessment of myelination and white matter injury after Best results are obtained by high in-plane resolution (512 matrix),
the first birthday; however, T1-weighted images are still useful to look thin section (3 mm) 2D sequences, especially when taking advantage
for discrepancies between the T1 and T2 appearances, which might of high field strength (3T) imaging. Susceptibility-weighted imaging
give a clue to diagnosis in hypomyelinating disorders (such as in 18q has the potential to differentiate between posthemorrhagic stigmata
deletion syndrome). T2 FLAIR images are superb for the detection of and calcifications in general, but capability needs to be yet validated
supratentorial white matter changes in children beyond the age of 1 in the case of neurometabolic diseases. In order to enhance the accu-
to 2 years (after myelination is largely completed). Conventional spin- racy of lesion pattern analysis, high spatial resolution imaging is
echo T2-weighted images are very useful on the initial study for detec- needed.
tion of subtle cortical malformations that might help in the diagnosis Diffusion imaging (and magnetization transfer) may be useful in
of a congenital infection (such as cytomegalovirus, see Chapter 11), separating regions of acute inflammation or cellular injury (reduced
cobblestone malformation (see Chapter 5), or generalized peroxisomal diffusivity) from those disorders with hypomyelination, myelin vac-
disorder; cortical malformations are more difficult to discern with T2 uolization, or cystic degeneration (increased diffusivity) and those
FLAIR because of reduced contrast between gray matter and white involved more chronically (normal or increased diffusivity, Table
matter. T2 FLAIR is also less sensitive in the detection of lesions within 3-2) (10). MR spectroscopy (MRS) may provide important diagnos-
the posterior fossa (15). Rapid acquisition relaxation-enhanced (also tic information in those disorders in which an abnormal metabolite
known as RARE, fast spin echo, and turbo spin echo) T2-weighted is present or a normal metabolite is present in abnormal quantity. We
images show cortex and white matter well but are less sensitive to the perform MRS routinely in patients with suspected metabolic disease,
presence of iron deposition or calcification; a long echo time gradient- as the combination of anatomic MRI, diffusion imaging, and MRS
echo sequence or susceptibility-weighted sequence should always be provides the best chance for making a diagnosis (10,18). See Chapter 1
added if fast spin echo or FLAIR is used as the primary T2-weighted for MRS techniques and Chapter 2 for the evolution of normal metab-
sequence. PROPELLER T2 (see Chapter 1), sometimes used to minimize olite levels during development. Single photon emission tomography
motion artifact, has poor white matter contrast and should be avoided in (SPECT) and positron emission tomography (PET) are occasionally

Barkovich_Chap03.indd 84 5/6/2011 12:07:52 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 85

useful, especially in gray matter diseases; when useful, these modalities appears abnormal, may be involved primarily or secondarily. The ven-
will be discussed. tricles and sulci are often big. The basal nuclei (the thalami and basal
For evaluation of the white matter, it may be useful to determine ganglia), brainstem, or cerebellum may be affected. We use a system-
whether the white matter abnormality represents hypomyelination, atic approach to the analysis of these disorders based upon the pattern
demyelination, myelin vacuolization, or cystic degeneration of the of cerebral involvement (Tables 3-3 to 3-5), based upon the work of
white matter (10). As the metabolic and microstructural characteris- van der Knaap and Valk (2,11). (Note that disorders predominantly
tics of toxic/metabolic/inflammatory disorders become better known, affecting the cerebellum are discussed at the end of this chapter). This
this type of differentiation will become more important because dif- approach is a simplification because many of the metabolic disorders
ferent types of metabolic disorders cause different microstructural and have a different appearance on imaging studies when imaged at differ-
biochemical disturbances to the axons, oligodendrocytes, and myelin ent stages of the disease. In addition, some disorders may have nearly
itself. Thus, by using magnetization transfer ratios, diffusion tensor identical neuroimaging features but have different ophthalmologic or
imaging, and proton MR spectroscopy, in addition to anatomic images, dermatologic features. This imaging approach is, therefore, most use-
one may be able to better specify the type of white matter abnormality ful when correlated with other clinical features and when the images
(Table 3-2). As with all MR analyses of these disorders, imaging is most are acquired and evaluated in the early stages of the disease. As men-
valuable when obtained early in the course of the disease and, if pos- tioned earlier, many diseases have very similar appearances when in
sible, during an exacerbation of disease: findings may be very different the chronic phase, with diffuse loss of brain tissue and increased water
in active phases of the disease (typically showing reduced diffusivity in the remaining tissue. Another potential source of error is that dif-
or high levels of abnormal metabolites) than in more chronic phases ferent disorders (i.e., disorders with different genotypes, resulting in
(where diffusivity is more typically increased and metabolite levels different enzyme deficiencies) can result in the accumulation of the
return to normal or lower). These characteristics are mentioned, when same biochemical(s) in the serum or cerebrospinal fluid (CSF) (simi-
known, in the following discussions. lar biochemical phenotypes); as a result, distinctly different diseases
are sometimes classified together as a single entity. It is not surpris-
ing, therefore, that some of them appear to have “atypical” patterns
A SIMPLE PATTERN APPROACH TO on imaging studies! Clinical manifestations of genetically, or even
biochemically, similar disorders may differ as to age of onset, clini-
METABOLIC DISEASE cal course, or outcome. These differing clinical phenotypes are likely
The imaging characteristics of toxic/metabolic/inflammatory brain modulated by modifier genes or environmental factors, which further
diseases can be very confusing. The white matter, which nearly always confound this complex subject. Finally, classification of many diseases

TABLE 3-3 Disorders with Exclusive or Dominant Gray Matter Involvement


A. Cortical Gray Matter d. Succinate semialdehyde dehydrogenase deficiency
1. Neuronal ceroid lipofuscinoses e. Guanidinoacetate methyltransferase deficiency
2. Mucolipidoses type I f. Isovaleric acidemia
3. Rett syndrome g. Pyruvate dehydrogenase (E2) deficiency
4. Alpers syndrome h. Beta-ketothiolase deficiency
5. Autoimmune anti-NMDA receptor encephalitis (19) 4. Prolonged (hyperintense) T2 in claustrum
B. Deep Gray Matter a. Wilson disease
1. Prolonged (hyperintense) T2 in striatum 5. Prolonged (hyperintense) T2 in subthalamic nucleus
a. Leigh syndrome (usually white matter involvement) a. Leigh syndrome
b. Alpha-ketoglutaric aciduria b. Kernicterus (chronic, with globus pallidus involvement)
c. Type I and IV 3-methylglutaconic aciduria C. Cerebellar and Brainstem
d. Juvenile Huntington disease 1. Prolonged (hyperintense) T2 in periaqueductal gray matter
e. MELAS a. Leigh syndrome
f. Wilson disease (often involves white matter, periaqueductal) b. Wernicke encephalopathy
g. Propionic acidemia (usually white matter involvement) c. Wilson disease
h. Ethylmalonic acidemia (usually white matter involvement) 2. Prolonged (hyperintense) T2 in cerebellar nuclei
i. Neuropathy/Striatal necrosis from SLC25A19 mutation (20) a. Propionic acidemia
j. Hypoxic-ischemic injury (older infants, adolescents, and adults) b. Ethylmalonic aciduria
k. Hypoglycemic injury (older infants, adolescents, and adults) c. Leigh disease
2. Short (hypointense) T2 in globus pallidus c. 3-methylglutaconic aciduria (type 1 and 4)
a. Pantothenate kinase associated neurodegeneration (formerly d. Beta-ketothiolase deficiency
Hallervorden-Spatz disease – T2 hyperintensity in center of T2 e. Biotin-responsive basal ganglia disease
hypointensity) f. Ethylmalonic aciduria
b. Oculodigital dental dysplasia g. Glutaric aciduria type 1
c. Infantile neuroaxonal dystrophy (atypical, PLA2G6 mutation) 3. Short (hypointense) T2 in cerebellar nuclei
3. Prolonged (hyperintense) T2 in globus pallidus a. Cerebrotendinous xanthomatosis
a. Methylmalonic acidemia 4. Prolonged (hyperintense) T2 in inferior olivary nucleus
b. Toxins (carbon monoxide, manganese, cyanide involves a. Dihydropyrimidine dehydrogenase deficiency
cerebellum too) b. Leigh syndrome
c. Kernicterus (chronic; subthalamic nuclei involved)

Barkovich_Chap03.indd 85 5/6/2011 12:07:52 AM


86 Pediatric Neuroimaging

is likely to change as we gain knowledge and experience. Nonetheless, If the pattern of the imaging study indicates that it is primarily one
this systematic approach will allow the user to get close to the diagnosis of cortical involvement (cortical thinning with enlarged cortical sulci),
much of the time. consideration should be given to such disorders as the neuronal ceroid
lipofuscinoses, Rett syndrome (in girls), Alpers disease, or glycogen
storage diseases (Table 3-3).
White Matter Versus Gray Matter
If only deep gray matter is involved, the signal intensity and loca-
The first important decision is whether the disease involves predomi- tion of the affected structures are pivotal (Table 3-3). Involvement
nantly gray matter (poliodystrophies), white matter (leukodystro- of the striatum (caudate and putamen) is seen in mitochondrial
phies), or both (pandystrophies). In general, disorders that primarily disorders (primarily Leigh syndrome, MELAS, and the glutaric aci-
affect cortical gray matter will cause thinning of the cortex and, thus, durias), many organic acidemias (propionic and ethylmalonic aci-
have prominent cortical sulci. In the acute phase (during metabolic demias), juvenile Huntington disease, hypoxic-ischemic injury (in
decompensation), most disorders primarily affecting deep gray mat- older infants, children, and adults, see Chapter 4), and hypoglycemia
ter will show swelling, reduced diffusivity, and either low attenuation (in older children and adults, see Chapter 4). Many of these disorders
(CT) or T1 hypointensity and T2/FLAIR hyperintensity (MRI) in the may also cause white matter injury. If involvement is restricted to the
involved structures; chronically, the structures are shrunken and have globus pallidus and consists of T2 hypointensity or T2 hypointensity
increased diffusion. The cerebral white matter will often have an abnor- with central T2 hyperintensity, the diagnosis of pantothenate kinase
mal appearance in disorders of gray matter, as Wallerian degeneration associated neurodegeneration (formerly called Hallervorden-Spatz
of axons causes diminished white matter volume and decreased white disease) or oculodental digital dysplasia can be suggested; these
matter attenuation (CT) or mildly to moderately prolonged white mat- are easily differentiated on clinical exam. If isolated globus pallidus
ter T2 relaxation time (MRI). This white matter appearance can often involvement shows T2 hyperintensity, methylmalonic acidemia, suc-
be differentiated from that of primary white matter disorders if the cinate semialdehyde dehydrogenase deficiency, guanidinoacetate
imaging study is performed early in the course of the disease. Disorders methyltransferase deficiency, poisoning (as from carbon monoxide
predominantly affecting white matter (other than hypomyelinating or cyanide), or kernicterus (see Chapter 4) should be considered.
disorders) cause marked hypoattenuation (CT) or T2 hyperintensity High signal in the globi pallidi on T1-weighted images in a neonate
(MRI) before any volume loss is apparent. Indeed, in some white mat- or young infant suggests urea cycle disorders if the insular cortex
ter diseases (such as Canavan disease), the white matter frankly enlarges is similarly involved. In infants and children, T1 hyperintensity of
in early stages because the myelin expands (spongiform myelinopathy) the globi pallidi should suggest hepatic failure. Injury to subthalamic
before it degenerates. Other white matter disorders, such as X-linked nuclei in conjunction with the globi pallidi should suggest kernict-
adrenoleukodystrophy, have an inflammatory component in the early erus, while subthalamic nuclei/putaminal injury suggests Leigh
stages. The inflammation causes edema, with accompanying mass syndrome.
effect upon adjacent sulci, and blood–brain barrier breakdown with
resultant enhancement. Moreover, many white matter disorders, such
White Matter Disorders
as infantile Alexander disease, start locally (frontal lobe in the case of
Alexander disease) and progress into adjacent areas (both white mat- If the abnormal signal intensity involves predominantly white matter,
ter and gray matter). Many white matter disorders result in devasta- determine whether the white matter is being broken down (demyeli-
tion of the involved areas, with necrosis and cavitation of the affected nation, cystic degeneration, or myelin vacuolation, see Table 3-2) or
brain and marked ex vacuo dilatation of the ventricles, whereas the whether the myelin is merely deficient or delayed (hypomyelination).
abnormal white matter secondary to gray matter disorders appears less If white matter is being broken down, the location of the abnormality
severely damaged. Finally, the clinical presentation of patients with (periventricular, deep, or subcortical white matter) and its progression
cortical gray matter disorders (seizures, visual loss, dementia in early pattern should be determined (Table 3-4). Initially, the white matter
stages) often differs from that of deep gray matter disorders (chorea, should be assessed to determine whether involvement is localized, mul-
athetosis, dystonia) and both differ from the presentation of white tifocal, or diffuse. Localized white matter involvement may represent
matter disorders (pyramidal signs such as spasticity, hyperreflexia or early multiple sclerosis (MS), acute disseminated encephalomyelitis
cerebellar signs such as ataxia). Clinical information is often critical in (ADEM), autoimmune angiitis, or radiation injury (a history of radia-
the proper evaluation of these patients, and, therefore, consultation of tion therapy should be sought). Asymmetric multifocal white matter
the imaging physician with the referring physician (geneticist or neu- involvement typically suggests autoimmune/inflammatory disorders
rologist, in most cases, who may have detected important findings on such as MS, ADEM, autoimmune angiitis, or systemic lupus erythema-
dermatologic or ophthalmologic exam) is essential for proper care of tosus (SLE), or unusual genetic disorders such as polyglucosan body
the child. disease, or hereditary diffuse leukoencephalopathy with neuroaxonal
spheroids.
If white matter involvement is symmetric, it should be analyzed
Gray Matter Disorders
to determine whether the subcortical, deep, or periventricular white
Once identified as being primarily of gray matter, the next step is to matter is initially involved and whether it starts in the frontal, pari-
determine if the disorder involves cortical or deep gray matter. Exam- etal, or occipital lobes or in the cerebellum or brainstem. An attempt
ine the deep gray nuclei for abnormal attenuation (CT) or abnormal should be made to find out whether the patient has macrocephaly. If
T2/FLAIR signal intensity (MRI). Sometimes MR spectroscopy or dif- early involvement is restricted to primarily deep and periventricular
fusion imaging is useful, although it is uncommon for diffusion to be white matter, the thalami should be specifically analyzed. High attenu-
abnormal in the setting of a normal FLAIR image. For confirmation ation (CT) or T2 hypointensity (MRI) bilaterally in the thalami sug-
of cortical involvement, a specific search for sulcal effacement, cortical gests Krabbe disease (demyelination with early involvement of internal
swelling and reduced diffusion (acute phase) or cortical thinning with capsules and cerebellar nuclei) or GM1 or GM2 gangliosidosis (demy-
sulcal enlargement (chronic phase), and abnormal signal intensity of elination with hyperintensity of deep cerebral nuclei). If the internal
the cortex may be helpful. capsules, cerebral peduncles, dorsal pons, and cerebellar white matter

Barkovich_Chap03.indd 86 5/6/2011 12:07:52 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 87

TABLE 3-4 Disorders Involving White Matter Only


A. Subcortical White Matter (U Fibers) Early
1. Large head
a. Megalencephalic leukoencephalopathy with cysts (formerly called van der Knaap disease)—subcortical cysts
b. Infantile Alexander disease—frontal involvement, extends into caudates and putamina-large myo-inositol and small NAA peak on MRS
2. Normal head size
a. Galactosemia
b. Cystic leukoencephalopathy without megalencephaly—cysts in anterior temporal lobe
3. 4-hydroxybutyric aciduria—also has cerebellar atrophy (21)
4. Aicardi-Goutières syndrome—progressive microcephaly and extensive punctate calcifications
B. Deep White Matter Early initially spares subcortical U fibers
1. Symmetric confluent white matter involvement
a. Calcium or short T1 and T2 in thalami
i. Krabbe disease (involves corticospinal tracts and cerebellar nuclei)
ii. GM1 or GM2 gangliosidoses
b. Normal thalami
i. Pons/Medulla corticospinal tract involvement
a. X-linked adrenoleukodystrophy,
b. Giant axonal neuropathy
c. Acyl CoA oxidase deficiency
ii. No specific brain stem tracts involved
a. Metachromatic leukodystrophy
b. Phenylketonuria
c. Lowe disease (cysts in deep white matter)
d. Sjögren-Larsson syndrome
e. Hyperhomocysteinemia (5, 10 methylenetetrahydrofolate reductase deficiency or errors involving cobalamin metabolism)
f. Radiation/Chemotherapy
g. Childhood ataxia with diffuse central nervous system hypomyelination and cavitation of affected white matter
(leukoencephalopathy with vanishing white matter)
h. Merosin-deficient congenital muscular dystrophy associated with small pons, sometimes small vermis
iii. Brain stem involved.
a. Maple syrup urine disease—myelinated white matter, including dorsal brain stem and corticospinal tracts
b. Dentatorubral and pallidoluysian atrophy—atrophy of dorsal brain stem and cerebellum
c. Juvenile Alexander disease
2. Multifocal white matter involvement
a. Multiple sclerosis
b. Acute disseminated encephalomyelitis
c. Polyglucosan body disease (22,23)
d. Autoimmune angiitis of the CNS (24,25)
e. Systemic lupus erythematosus
f. Hereditary diffuse leukoencephalopathy with neuroaxonal spheroids (26)
C. Hypomyelination
1. Pelizaeus-Merzbacher disease (27)
2. Pelizaeus-Merzbacher-like disease (28,29)
3. Trichothiodystrophy
4. 18q- syndrome (30,31)
5. Sialuria (32,33)
6. Cockayne syndrome type II
7. Hypomyelination with atrophy of the basal ganglia and cerebellum (34)
8. Hypomyelination with congenital cataracts (35,36)
9. 3-phosphoglycerate dehydrogenase deficiency
D. Nonspecific White Matter Pattern
1. 3-hydroxy-3-methylglutaryl-coenzyme A lyase deficiency (37)
2. Congenital and acquired infections (see Chapter 11), particularly viral encephalitides, can involve gray and white matter symmetrically or
asymmetrically and should be included in the differential diagnosis in the proper clinical setting.
(References Given for Some Disorders Not Otherwise Discussed in the Chapter)

Barkovich_Chap03.indd 87 5/6/2011 12:07:52 AM


88 Pediatric Neuroimaging

are affected in a newborn, maple syrup urine disease (myelin vacuola- A Few Comments About Classifications
tion) should be considered.
If sequential MR studies are available, the pattern of progression White matter diseases of children were traditionally divided into the
of white matter involvement may be useful. Some disorders, such as categories of dysmyelinating and myelinoclastic diseases. This is not
L-2-hydroxyglutaric aciduria and Canavan disease, have a centripetal a useful distinction unless analysis is histologic, because many abnor-
progression pattern; the subcortical white matter is involved early and malities of white matter are the result of inflammatory infiltrates or
involvement progresses centrally. In contrast, other disorders such as accumulation of toxins from inborn errors of metabolism, which do
Krabbe disease and X-linked adrenoleukodystrophy, exhibit a centrifu- not fit well into either group. Dysmyelinating diseases result from an
gal progression pattern; the periventricular white matter is involved inherited enzymatic deficiency that causes abnormal formation or
early with progression to the deep and subcortical white matter. Pro- increased breakdown of the components of myelin. Areas of abnor-
gression may also advance from anterior to posterior as in infantile mal myelination in dysmyelinating diseases are relatively symmetri-
Alexander disease or from posterior to anterior as in X-linked adreno- cal, central (sparing subcortical U fibers in early stages), affect both
leukodystrophy or Krabbe disease. cerebral and cerebellar white matter, and are poorly marginated (45).
The pattern of hypomyelination, as opposed to damaged or Myelinoclastic disorders involve destruction of intrinsically normal
destroyed myelination, is seen in the so-called hypomyelinating dis- myelin; causes include extrinsic and intrinsic toxins, infection, chemo-
orders. It can be recognized by the appearance of delayed myelination therapy, radiation, and inflammatory autoimmune disorders such as
on T2-weighted images, often accompanied by a much more normal MS. Myelinoclastic injury is characteristically sharply defined, asym-
appearance on T1-weighed images. metric, and involves the subcortical U fibers early in the course of the
Congenital and acquired infections (see Chapter 11), particularly disease (45). As discussed above, the use of these morphologic charac-
viral encephalitides, can involve gray and white matter symmetrically teristics allows a diagnosis to be established in less than half of affected
or asymmetrically and should be included in the differential diagnosis patients.
in the proper clinical setting. In many sources, disorders of white matter are classified by the
intracellular organelle in which the malfunctioning enzyme is located
(45–47). For example, metachromatic leukodystrophy, in which the
Disorders Affecting Gray and White Matter defective enzyme is situated in lysosomes, is classified as a lysosomal
disorder, and X-linked adrenoleukodystrophy, in which the defective
Exclusive damage of either gray matter or white matter structures enzyme is in the peroxisomes, is a peroxisomal disorder. However,
is rather rare in neurometabolic disorders. Most commonly both the phenotypic appearance of the brain (e.g., white matter involve-
are involved, but in variable proportions. Disorders with significant ment versus gray matter involvement) usually depends more upon
damage to both gray and white matter (Table 3-5) can be divided the specific metabolic pathway involved than the organelle in which
into those involving only the cerebral cortex and those involving the enzyme is located; indeed, some enzymes are synthesized in one
deep gray nuclei (with or without cortical involvement). Those dis- organelle but have their function in another. Therefore, classifica-
orders involving only cortical gray matter can be subdivided depend- tion by organelle is impractical from the imaging perspective. Some
ing on whether the patient has normal long bones and spinal column understanding of the organelle approach to classification is necessary,
(Table 3-5). however, if one is to read and understand the literature. Therefore, one
If deep gray matter is involved, differential diagnosis is depen- should know that lysosomes are intracellular organelles that contain
dent upon which nuclei are primarily involved. As seen in Table 3-5, lysosymes, hydrolytic enzymes that aid in the phagocytosis of unde-
various combinations of deep gray nuclei can be involved at the same sirable molecules and particles. Dysfunction of specific lysosomal
time as various parts of the white matter are affected. Close analysis enzymes results in a variety of disorders that affect the brain. Those
of the patterns of both gray matter and white matter involvement, that affect oligodendrocytes result in white matter diseases (46). It is
as well as the presence of calcification, may aid in establishing the also important to know that mitochondria are the organelles that pro-
diagnosis (38). duce ATP, the energy source for intracellular processes. As a result, dis-
orders of mitochondrial function typically result in impaired function
DEEPER ANALYSIS OF TOXIC/METABOLIC/ in organs that require high energy such as the heart, skeletal muscle,
and the brain (and within the brain, the gray matter structures) (4).
INFLAMMATORY DISEASES One should also know that peroxisomes are small organelles measur-
The following sections describe a large number of toxic, metabolic ing 0.2 to 1.0 mm. They are limited by a single membrane and contain
and autoimmune inflammatory disorders, their major clinical mani- a minimum of one oxidase, to form hydrogen peroxide, and one cata-
festations, underlying genetic and/or biochemical causes, numerical lase to decompose it (45,48–50). Although peroxisomes can contain
listing in Online Mendelian Inheritance in Man (OMIM (44) ), and many enzymes, their most important functions from the perspective
imaging characteristics. The classification of each disorder in this sec- of this chapter are synthesis of plasmalogens (ether-phospholipids), a
tion is based mainly on standard anatomic imaging characteristics. type of phospholipid that constitutes the vast majority of membrane
When diffusion MRI, proton MR spectroscopic, and positron emis- phospholipids in myelin; beta oxidation of very long chain (>26 car-
sion tomographic characteristics of disorders have been described, bons) fatty acids, dihydroxycholestanoic and trihydroxycholestanoic
these characteristics are discussed. In some disorders that have rather acid, pristanic acid, and long chain dicarboxylic acids; and cholesterol
nonspecific imaging characteristics (such as the creatine synthesis biosynthesis. The process of beta oxidation results in the synthesis of
disorders), metabolic characteristics on MR spectroscopy may be hydrogen peroxide. Plasmalogens, long chain fatty acids, and choles-
diagnostic and are discussed at greater length; more commonly, these terol are important components of myelin. If these structures are not
“functional” studies are nonspecific but may narrow the differential properly formed, they result in formation of unstable myelin that is
diagnosis. more easily broken down by normal metabolic processes of the body

Barkovich_Chap03.indd 88 5/6/2011 12:07:52 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 89

TABLE 3-5 Disorders Involving Gray Matter and White Matter


A. Cortical Gray Matter and White Matter Only (No Deep Gray Matter Involvement)
1. Normal bones
a. Cortical dysgenesis
i. Congenital infection (most commonly cytomegalovirus)
ii. Cobblestone malformations of the cerebral cortex (see Chapter 5)
b. No cortical dysgenesis
i. Alpers disease
ii. Menkes disease
2. Abnormal bones
a. Mucopolysaccharidoses
b. Lipid storage disorders
c. Peroxisomal disorders
B. Deep Gray Matter and White Matter Involvement
1. Primarily thalamic involvement
a. Krabbe disease—early corticospinal tract, cerebellar nuclear involvement
b. GM1 Gangliosidoses
c. GM2 Gangliosidoses
d. Wilson disease—also periaqueductal gray matter
e. Profound neonatal hypotensive encephalopathy
f. Many viral encephalitides
2. Primarily globus pallidus involvement
a. Canavan disease—thalami involved, large NAA peak on MRS
b. Kearns-Sayre syndrome—with subcortical white matter, often caudate
c. Methylmalonic acidemia—occasionally with deep white matter
d. Toxins (carbon monoxide and cyanide)
e. Maple syrup urine disease—with dorsal brain stem, corticospinal tracts
f. L-2-hydroxyglutaric aciduria—with subcortical white matter (centripetal progression), mild striatal involvement, and cerebellar nuclei
g. Fucosidosis (T2/FLAIR hypointensity)
h. Dentatorubral and pallidoluysian atrophy—extensive cerebellar/midbrain atrophy
i. Urea cycle disorders—with insular involvement
j. Cree leukoencephalopathy—with dorsal brain stem
3. Primarily striatal involvement
a. Leigh syndrome
b. MELAS—usually cortical involvement
c. Wilson disease
d. Alexander disease (with frontal white matter)
e. Ethylmalonic acidemia (39)
f. Propionic acidemia (40)
g. Glutaric aciduria Type I (Glutaryl-CoA dehydrogenase deficiency)—with anterior temporal lobe hypoplasia
g. 3-hydroxy-3-methylglutaryl-coenzyme A lyase deficiency (37)
h. Molybdenum co-factor deficiency
i. Mitochondrial ATP synthetase deficiency (41)
j. 3-methylglutaconic aciduria
k. Beta ketothiolase deficiency. (41)
l. Malonic acidemia (41)
m. Alpha ketoglutaric aciduria (42)
n. Biotinidase deficiencies (41)
o. Biotin responsive basal ganglia disease (43)
p. Toxins
q. Hypoxic-ischemic injury in older child or adult
r. Hypoglycemic injury in older child or adult
s. Cockayne disease (38)
t. Hypomyelination with atrophy of basal ganglia and cerebellum—with atrophic caudate, putamen, cerebellum

(References Given for Some Disorders Not Otherwise Discussed in the Chapter)

Barkovich_Chap03.indd 89 5/6/2011 12:07:52 AM


90 Pediatric Neuroimaging

(50,51). In addition, if long chain fatty acids are not properly formed,
the result is disrupted lipid packing in myelin with resultant abnormal TABLE 3-6 Most Common Devastating
T2 relaxation time of white matter (52). Unfortunately, classification
by organelle is not useful for an imaging approach, as most organ- Metabolic Diseases of the
elles contain many proteins that can be involved in many metabolic Newborn
pathways.
The use of physiologic techniques such as diffusion, magnetization Isolated sulfite oxidase deficiency
transfer, and proton spectroscopy characteristics (Table 3-2) has great
Propionic acidemia
potential in the classification of toxic and metabolic disorders. Unfor-
tunately, these characteristics are incompletely known for many toxic/ Methylmalonic acidemia
metabolic/inflammatory disorders. Therefore, at this time, a combina- Isovaleric acidemia
tion of morphologic and physiologic data (when known) seems the
HMG-CoA lyase deficiency
best method. In the following sections, disorders of white matter are
organized by the pattern of initial white matter involvement on ana- 3-methylglutaconic aciduria (some forms)
tomic MRI. Characteristics of diffusion imaging, magnetization trans- Primary lactic acidosis
fer, proton spectroscopy, and PET are discussed when they are useful in
further refining the diagnosis. Glutaric aciduria type 2
Pyroglutamic aciduria
Clinical Aspects of Metabolic Diseases Urea cycle defects (citrullinemia, ornithine transcarbamylase
deficiency)
Pattern recognition alone is often insufficient to characterize a particu-
lar disorder. The integration of clinical data (gender, age of onset, and Maple syrup urine disease
type of clinical manifestations; disease course; physical or ophthalmo- Non-ketotic hyperglycinemia
logical stigmata) and imaging abnormalities (lesion types and patterns,
Menkes disease
as well as advanced MR data) allows recognition of the more specific
clinical-radiological pattern. Therefore, knowledge of some clinical Nesidioblastosis
data is often important for the neuroradiologist involved in the diag- Peroxisomal biogenesis disorders
nostic workup and follow-up of patients with suspected or confirmed
neurometabolic disorders. The most important categories of clinically
relevant data are discussed here.

Age of Onset are useful. Diffusion-weighted imaging may be pathognomonic in


The age of onset of metabolic diseases is a very useful clinical pattern maple syrup urine disease; MR spectroscopy may be diagnostic in
element. Depending on age of onset, the following categories may be 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) lyase deficiency
used: neonatal (from birth to 1 month), early and late infantile (1–6 (unpublished observation) or suggestive in energy metabolism disor-
months and 6 months to 3 years), early and late childhood-juvenile ders (lactate) and urea cycle defects (glutamate), as well as in nonke-
(3–6 and 6–16 years), and adult (16 and over). It is important to recog- totic hyperglycinemia (glycine).
nize that the onset of the clinical signs and symptoms does not neces-
sarily correspond to the onset of the metabolic derangement. Systemic Manifestations of Metabolic Disorders
As a clear manifestation of their considerable phenotypic varia- Systemic manifestations of neurometabolic disorders may include
tions, almost all neurometabolic diseases have several clinical forms dysmorphia (e.g., coarse facial features), organomegaly, skeletal abnor-
depending on the age of onset. Within the same disease category, ear- malities, or, more generically, failure to thrive. Systemic complications
lier onsets usually suggest a more profound metabolic derangement, in metabolic disorders are common but often misleading clinically.
whereas later onset often reflects clinically milder but not necessarily Patients with metabolic diseases are prone to intercurrent infections
benign variants. Most metabolic disorders of neonatal onset fall into with or without CNS involvement, but intercurrent infectious diseases
the category of the so-called devastating metabolic diseases of the can trigger episodes of metabolic crisis in patients with neurometabolic
newborn. The underlying metabolic derangements in these diseases disorder. Acute pancreatitis is increasingly recognized as a possible
typically result in global brain toxicity (encephalopathy), leading to complication of organic- and aminoacidopathies and usually occurs
diffuse brain edema and neurological manifestations reflecting varying during an acute ketoacidotic crisis. Hematological abnormalities,
proportions of white or gray matter involvement. The term “devastat- notably anemia, neutropenia, and thrombocytopenia are often seen in
ing metabolic diseases of the newborn” actually refers to a fairly well- organic acidopathies.
defined clinical syndrome. The neonate is typically normal at birth. The
prodrome begins a few days later and is characterized by refusal to feed Neurological Abnormalities
and vomiting (occasionally misinterpreted as pyloric stenosis). This Neurometabolic diseases may present as an acute or chronic
is followed by lethargy and coma, which may clinically mimic central encephalopathy. Mixed forms can be characterized by progressive
nervous system infection. At this point, seizures and changes in muscle encephalopathy and occasional metabolic deterioration. Acute enceph-
tone (hypotonia or hypertonia) are common. If the disease is not diag- alopathy develops during acute decompensation (lactic acidosis, ketosis
nosed and treated promptly, the progressive toxicity leads to irrevers- or ketoacidosis, hyperammonemia, hypoglycemia, hyperglycinemia).
ible neurological deficit or death. Most devastating metabolic diseases Neurological manifestations include hypotonia, seizures, metabolic
of the newborn are organic or amino acidopathies (Table 3-6). stroke with rapid onset of extrapyramidal movement disorders, and
If a “devastating metabolic disease” is suspected in a newborn or coma. Other metabolic disorders may be associated with a more insidi-
young infant, in addition to anatomical MRI, advanced MR techniques ous course and result in a chronic progressive encephalopathy with

Barkovich_Chap03.indd 90 5/6/2011 12:07:53 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 91

neurological manifestations that may include seizures, pyramidal and hypertonia, presenting with opisthotonus) is typical of maple syrup
extrapyramidal movement disorders, and neurocognitive and behav- urine disease.
ioral problems.
Additional Useful Clinical Features
Seizures
Other useful features in the diagnosis of metabolic disorders include
Seizures are frequent but nonspecific complications of metabolic dis-
a characteristic odor caused by the production of abnormal metabo-
orders. Inborn errors of metabolism are a common cause (16%) of
lites, facial dysmorphia (present in peroxisomal disorders and muco-
first-time seizures in infants under the age of 6 months. Seizures reflect
polysaccharidoses), ophthalmologic and dermatologic disorders.
cerebral cortex involvement in the pathological process. In some met-
A discussion of these features is beyond the scope of this book; for
abolic diseases (e.g., fatty acid oxidation disorders, nesidioblastosis),
details, readers are referred to standard texts of metabolic disor-
especially in those with early infantile onset, seizures may be secondary
ders such as those by van der Knaap and Valk (11) or Scriver et al.
to hypoglycemia.
(12,13).
Stroke or Strokelike Clinical Presentation
While the development of neurological deficit (pyramidal signs, DEEPER ANALYSIS OF GRAY AND WHITE
extrapyramidal signs) in neurometabolic diseases is often slowly pro-
gressive or insidious, inherited metabolic disorders may also present
MATTER DISEASES
with focal neurological signs of acute onset. These clinical events may Diseases Primarily Affecting White Matter
be related to true strokes (ischemic or hemorrhagic) or stroke-like
episodes. Some organic acidopathies (e.g., propionic, isovaleric and White Matter Diseases Initially Affecting Periventricular
methylmalonic acidemias) are known to predispose to intracranial Cerebral White Matter
hemorrhage due to coagulation abnormalities related to thrombocy- Metachromatic Leukodystrophy
topenia. Acute ischemic complications (i.e. true infarctions) occur in Metachromatic leukodystrophy (OMIM 250100) causes diffusely
some aminoacidopathies, likely due to direct vessel-wall damage, and abnormal myelination of the cerebral hemispheres; it can be mani-
in lysosomal storage disorders such as Fabry disease and cystinosis. fested in a variety of forms. All forms are caused by decreased activity
In Fabry disease, both ischemic and hemorrhagic complications may of arylsulfatase-A or, very rarely, a cofactor of arylsulfatase-A (saposin
occur since endothelial damage (glycosphingolipid deposits within B, or sphingolipid activator protein B), which degrade sulfogalactosyl-
the endothelium) leads initially to occlusive arterial disease, followed ceramide into galactocerebroside and sulfate. The responsible genes are
by bleeding from overloaded collaterals, somewhat similar to child- ARSA, located at chromosome 22q13.31-qter (53), and PSAP, located
hood moyamoya syndrome. Rare other neurometabolic causes of at chromosome 10q22.1 (54). The decreased activity of arylsulfatase-A
ischemic stroke are Menkes disease, sulfite oxidase deficiency (molyb- or saposin B results in failure of myelin breakdown and reutilization
denum cofactor deficiency), and carbohydrate-deficient glycoprotein in the central and peripheral nervous systems. Furthermore, ceramide
syndrome. sulfatide accumulates and impairs function of neurons, macrophages,
Stroke-like events probably represent episodes of regional met- and Schwann cells (55).
abolic decompensation with subsequent functional disturbances The most common clinical form of metachromatic leukodystro-
within brain parenchyma, which may or may not lead to permanent phy is the late infantile variant, which typically presents with a gait
structural damage. Stroke-like episodes with hemiparesis or hemiano- disorder and strabismus early in the second year of life. Impairment
psia are characteristic in “mitochondrial disorders” such as MELAS. of speech, spasticity, and intellectual deterioration appear gradually.
Occasionally, extrapyramidal movement disorders (dystonia, chore- Progression is steady, with death occurring usually within 4 years of
oathetosis or tremor) may develop suddenly and mimic stroke, typi- the onset of symptoms (56). The juvenile form is slightly rarer. Neu-
cally in organic acidurias (e.g., glutaric aciduria type 1, methylmalonic rologic symptoms become evident between 5 and 7 years of age and
acidemia) and are usually associated with severe basal ganglia dis- progress slowly; affected patients often present with declining school
ease, typically without thalamic involvement. This can be contrasted performance (56,57). In the uncommon adult form, patients develop
with perinatal hypoxic-ischemic brain damage, in which thalami an organic mental syndrome and progressive corticospinal, corticobul-
and posterior parts of putamina are usually affected (see Chapter 4). bar, cerebellar, or extrapyramidal signs (58).
Clinically, both may present later in childhood with extrapyramidal The imaging findings in metachromatic leukodystrophy are non-
“cerebral palsy.” specific. CT scans show progressive atrophy and diffusely low attenua-
tion in the central cerebral white matter. No enhancement occurs after
Muscle Tone Changes contrast administration (58,59). MR scans show progressive symmetric
Muscle tone changes are common in neurometabolic disorders. areas of prolonged T1 and T2 relaxation times in the deep and periven-
Hypotonia is common in many metabolic disorders, especially in tricular cerebral white matter (Fig. 3-1); the subcortical white matter
those associated (directly or indirectly) with impairment of energy is spared until late in the course of the disease (Fig. 3-1A) (46,59).
metabolism (mitochondrial disorders) and peroxisomal disorders. In High-resolution images show stripes of affected and unaffected myelin
nonketotic hyperglycinemia and propionic acidemia, hypotonia may (called a “tigroid” pattern) that extend peripherally from the surface
be caused by increased blood glycine levels, since glycine is known of the lateral ventricles (Fig. 3-1A); these represent relatively spared
to have an inhibitory effect on spinal ventral motor neurons. In fatty myelin and lipid-containing glial cells (60). In my experience, these
acid oxidation disorders, and perhaps in many of the mitochondrial are rather characteristic, although van der Voorn et al. describe similar
cytopathies too, affected children are hypotonic because of associated (but less obvious) stripes in Krabbe disease and GM1 gangliosidoses
myopathy. Hypertonia is typical of Krabbe disease and with methylma- (both described later in this chapter). Eichler et al. found a charac-
lonic and isovaleric acidemia, while contractures are seen in rhizomelic teristic evolution of white matter involvement that is similar among
chondrodystrophia punctata. Alternating muscle tonus (hypotonia and the late infantile, juvenile, and adult-onset groups (61). A homogenous

Barkovich_Chap03.indd 91 5/6/2011 12:07:53 AM


92 Pediatric Neuroimaging

FIG. 3-1. Metachromatic Leukodystrophy. A. Axial T2-weighted


image shows abnormal hyperintensity in the deep and periventricu-
lar white matter; the subcortical white matter (white arrowheads) is
spared. Note the horizontal stripes of relatively spared (darker) white
matter. B. Axial diffusion weighted image shows abnormal hyper-
intensity of the white matter, indicating reduced diffusivity.C. Axial
postcontrast T1-weighted image in a different patient shows enhance-
ment (white arrowheads) of the cisternal portion of the oculomotor
nerves. D. Sagittal postcontrast T1-weighted image of the lumbar
spine shows enhancement of the cauda equina (white arrowheads).

region of T2 hyperintensity is initially found in the frontal and parietal the unaffected regions showing normal diffusion characteristics and
periventricular and deep white matter; it is faintly hyperintense in mild the affected regions exhibiting increased water motion. Proton MRS
disease and more hyperintense in more severe disease; callosal involve- shows decreased N-acetylaspartate (NAA) and elevated myo-Inositol
ment is mild. As the disease progresses, the signal abnormality extends (myo-I)(67).
toward the subcortical white matter and the tigroid pattern becomes
evident (61). Involvement of the corticospinal tracts, cerebellar white Globoid Cell Leukodystrophy (Krabbe Disease)
matter, and basal ganglia is only seen late in the course of the disease Clinical manifestations of Krabbe disease (OMIM 245200), also
(61,62), along with progressive loss of hemispheric brain tissue. An MR known as globoid cell leukodystrophy, typically begin acutely between
scoring system has been devised to assess severity of brain involvement age 3 and 6 months with restlessness, irritability, intermittent fever,
(61). If paramagnetic contrast is administered, the cranial nerves and feeding problems, poor head control, hyperactive reflexes, and delayed
cauda equina may enhance (Fig. 3-1C and D) (63); the reason for this is development (68). Optic atrophy and hyperacusis often develop, fol-
not known, but it is likely due to the inflammatory response to myelin lowed by episodes of apnea and need for nasogastric or gastrostomy
breakdown. feedings. Terminally, the infants are flaccid and develop bulbar signs;
Diffusion-weighted imaging shows decreased water diffusivity in they die within the first few years of life (56,57). The basic defect in
affected regions (64,65) early in the course of the disease (Fig. 3-1B), this condition is a deficiency of galactosylceramide beta-galactosidase,
and increased diffusivity in later phases of the disease (66). If high- a lysosomal enzyme that is a key component in metabolic pathways of
resolution diffusion-weighted images can be obtained, one can see the myelin turnover and breakdown. Deficiency of this compound results
same horizontal stripes of affected and unaffected white matter, with in the accumulation of galactosylsphingosine (psychosine), which is

Barkovich_Chap03.indd 92 5/6/2011 12:07:53 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 93

toxic to neurons, oligodendrocytes, and Schwann cells (56,69). The Few reports of proton MRS of patients with Krabbe disease have
gene for galactosylceramidase (GALC) has been mapped to chromo- been published. In infantile Krabbe disease, reports show marked ele-
some 14q31; a number of mutations in this region have been shown vation of choline and myo-I, moderate elevation of creatine, moder-
to cause clinical Krabbe disease (70). For those who evaluate adult ate to marked NAA reduction, and inconsistent lactate elevation in the
patients as well as children, it is important to note that the disease white matter (Fig. 3-2F); these changes were less marked in gray matter
may present at different ages and with different clinical and radiologic (87–89). These findings are attributed to changes in glial membrane
manifestations, depending upon the exact site and type of mutation composition and microglia activation for synthesis of phospholipid
(71–73). membranes. We have also noted increase in the size of the “macromo-
Pathologic examination shows replacement of cerebral white mat- lecular peaks,” broad peaks centered at 0.9 and 1.3 ppm (Fig. 3-2F).
ter with rubbery, hard, translucent material; pseudocysts may develop Changes in juvenile Krabbe disease were much less marked and con-
adjacent to the frontal horns and in the corpus callosum (74). Micro- sisted mainly of elevated myo-I in white mater and normal spectra of
scopically, severe neuronal loss is observed in the thalami, cerebellar gray matter (88). In adults, MRS of white matter shows mild decrease
cortex, cerebellar dentate nuclei, and inferior olivary nuclei (74). Severe in the concentration of NAA and mild elevations of creatine, choline,
destruction of myelin and axons with reduced numbers of oligoden- and myo-I (73,88).
drocytes is observed in white matter throughout the CNS, but is most
pronounced in the corona radiata, corpus callosum, and cerebellar Classic X-Linked Adrenal Leukodystrophy/
peduncles (74). Adrenomyeloneuropathy/Acyl CoA
Although diagnosis in these patients is based upon the assay of Oxidase Deficiency
beta-galactosidase from white blood cells or skin fibroblasts, the CT Classic, X-linked adrenal leukodystrophy (OMIM 300100) is the
features can occasionally be very helpful. Early in the disease, high den- result of a mutation of the ABCD1 gene, which has been mapped to
sity is seen bilaterally in the thalami (Fig. 3-2A), caudate nuclei, corona chromosome Xq28 and codes for a peroxisomal membrane protein
radiata, and cerebellar dentate nuclei. The high density may be seen of the ATPase binding cassette type (90,91). Abnormality of the pro-
before or in conjunction with the decrease in attenuation of the white tein impairs the transport of very long chain fatty acids into the per-
matter (75). As the disease progresses, diffuse white matter atrophy oxisome, where they are normally metabolized into shorter chain fatty
ensues; at this stage, the CT appearance of Krabbe disease resembles acids that can be used for the biosynthesis of complex lipids and acy-
that of the end stage of all other dysmyelinating diseases (75,76). On lated proteins in the cytoplasm (92). Thus, plasma analysis for very
T1-weighted MR images in young infants, the thalami are often abnor- long chain fatty acids is the best initial biomarker for the disease, allow-
mally bright and the normal hyperintensity of the internal capsules is ing unambiguous diagnosis in males (although false negative results
delayed in appearance (Fig. 3-2B); these findings are useful if present are found in about 20% of obligate heterozygote carriers (93,94). So
(77). On T2-weighted images, the hila of the cerebellar nuclei remain far, more than 500 different mutations of the ABCD1 gene have been
hyperintense, and the corticospinal tracts are abnormally hyperin- described (94); there is no clear correlation between location of the
tense in the internal capsules, medial medullary pyramids, and cere- mutation and phenotype (95,96). This disorder is characterized by two
bral white matter (Fig. 3-2C–E) (72); in our experience these findings major pathophysiologic components, a severe inflammatory demyeli-
strongly suggest the diagnosis. Later in the first year and in the second nation that predominates in the cerebral white matter and an axonal
year, the deep cerebral white matter shows T2 hyperintensity, particu- degeneration that predominates in the posterior fossa and spinal cord
larly in the posterior frontal/anterior parietal lobes (Fig. 3-3) (78,79) (97). The axonopathy and inflammation are both thought to result
with extension into the callosal splenium or posterior limb of the inter- from accumulation of very long chain fatty acids in the nervous tis-
nal capsule; the cerebellar white matter is more severely involved, and sue; normal cells are protected by plasmalogens (structural phospho-
significant volume loss is often present (Fig. 3-3D–G) (80,81). By the lipids in nervous tissue), but plasmalogens are also affected in many
second year, the MR appearance of Krabbe disease is that of nonspe- patients with ABCD1 mutations (98). Three different zones of white
cific T1 hypointensity and T2 hyperintensity in the cerebellar nuclei matter involvement are identified within the cerebrum; these affect the
and the deep cerebral and cerebellar white matter (72). Van der Voorn radiologic appearance. A central burned-out zone of scarring (Zone A)
et al. describe the presence of stripes of relatively spared white mat- contains only astrogliosis. Just peripheral to the central zone is an
ter representing perivenular clusters of globoid cells (Fig. 3-3G) (60), inflammatory zone (Zone B), with perivascular inflammatory cells and
but these stripes are not found as consistently in Krabbe disease as in demyelination; axons are preserved. Peripheral to the inflammation is
metachromatic leukodystrophy. The subcortical white matter is spared a zone of ongoing demyelination (Zone C), where myelin is breaking
early in the course of the disease (82). The thalami may be normal or down in the absence of inflammation. Some authors also suggest the
show T1 hyperintensity or T2 hypointensity (Fig. 3-3) (59). Enhance- presence of a zone of impending or incipient demyelination (Zone D),
ment and enlargement of the cranial nerves (83,84) and nerve roots peripheral to Zone C; a decrease in NAA (with or without increased
of the cauda equina (85) have been reported after administration of choline) can be found in this zone when proton MR spectroscopy is
paramagnetic contrast, presumably secondary to myelin breakdown performed (99).
and associated inflammatory response; such enhancement may be a Although many different clinical forms of adrenoleukodystrophy
common finding. have been described (Table 3-7) (92,100), the most common (and the
Diffusion tensor imaging may be useful in the early diagnosis of one of interest to those caring for children) is the childhood cerebral
Krabbe disease, because affected neonates have significantly lower form, which is characterized mainly by the inflammatory demyelina-
fractional anisotropy than age-matched controls (86). Diffusion- tion of the cerebrum. Affected patients are almost exclusively boys,
weighted images show reduced diffusion early in the course of the who usually present between age 5 and 12 years (mean 7.2 years) (92);
disease, particularly in the subcortical white matter, caudate head, all ethnic groups are affected with no apparent difference in frequency
and anterior limb of the internal capsule. As the disease progresses, (94). Many children present with learning difficulties at school and
the white matter begins to show uniformly increased diffusion are initially given a diagnosis of attention deficit hyperactivity disor-
(65,66). der. Other early features of the disorder include impaired visuospatial

Barkovich_Chap03.indd 93 5/6/2011 12:07:54 AM


94 Pediatric Neuroimaging

FIG. 3-2. Krabbe disease in a 4-month old infant. A. Axial noncontrast CT shows hyperdensity of the thalami (white arrows). B. Axial T1-weighted image
shows abnormal hyperintensity of the thalami and absence of the normal hyperintensity in the internal capsules. C–E. Axial T2-weighted images show
abnormal hyperintensity (white arrows) in the hila of the cerebellar nuclei (C), the posterior limbs of the internal capsules (D), and the perirolandic region
of the centrum semiovale (E). F. Single voxel short echo proton MR spectrum from frontal white matter shows abnormally elevated “macromolecular” peaks
(large arrows), a small NAA peak (small arrow), and large choline (Ch) and myo-inositol (mI) peaks for age.

Barkovich_Chap03.indd 94 5/6/2011 12:07:54 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 95

FIG. 3-3. Evolution of Krabbe disease in an infant. A. Axial T2-weighted image at age 14 months shows
abnormal hyperintensity (white arrow) in the left pontine corticospinal tract. B and C. Axial T2-weighted
images show abnormal hyperintensity (white arrows) fairly well localized to the corticospinal tracts. No
volume loss is present. Some horizontal stripes of hypointensity can be seen within the affected white
matter in (C). D. Axial T2-weighted image at age 21 months shows abnormal hypointensity of the medial
aspect of the middle cerebellar peduncles (white arrowheads) and the brainstem corticospinal tracts (black
arrows). E–G. Axial T2-weighted images show more pronounced involvement of the corticospinal tracts
(white arrows, compare with A–C), increased extent of white matter involvement in the periventricular
and deep cerebral white matter (F and G), and loss of volume (enlarging sulci and diminished white
matter volume compared to B and C). Note the horizontal stripes of hypointensity in (G). These are less
marked than in metachromatic leukodystrophy.

Barkovich_Chap03.indd 95 5/6/2011 12:07:55 AM


96 Pediatric Neuroimaging

TABLE 3-7 Phenotypes of X-ALD Carriers


Phenotype Description Estimated Relative Frequency
Male X-ALD Carriers
Asymptomatic (MRI normal) Patients up to 10 y old Increasing as diagnostic tests
improve
Asymptomatic (MRI Patients between 2 and 10 y old. Usually initially diagnosed as Increasing as diagnostic tests
abnormal) another leukodystrophy. improve
Cerebral (mild) without Patients usually between 3 and 10 y, but up to 21 y. Present with 45%
adrenomyeloneuropathy behavior changes, school failure, dementia. Typically initially
(AMN) diagnosed as ADHD, autism, or Asperger syndrome.
Cerebral (severe) without Onset between 5 y old and adulthood. Progressive behavioral, 2%–3%
AMN cognitive, and neurologic (epilepsy, visual loss, speech loss,
bulbar palsy) deficit. Often leads to total disability within
3 y. May be misdiagnosed as other neurodegenerative
diseases, tumor, or psychosis
Pure adrenomyeloneuropathy Onset 28 ± 9 y. Paraparesis, sphincter disturbances, sensory changes, 35%
(AMN) incoordination, pain, impotence. Progressive over decades.
Involves primarily spinal cord with minimal or no inflammatory
response. Often misdiagnosed as multiple sclerosis, progressive
spastic paraparesis, cervical spondylosis, amyotrophic lateral
sclerosis. ∼20% will develop brain involvement.
AMN with cerebral Onset 28 ± 9 y. Like pure AMN plus dementia, behavioral distur- 15%
involvement bances, psychosis, epilepsy, aphasia, visual loss bulbar palsy.
Cerebellar Cerebellar/brain stem involved in adolescence or adulthood. Patients 2%–3%
present with ataxia, cranial neuropathies or long tract signs
Addison only Onset usually before 10 y. Adrenal insufficiency without neurologic 20%
signs or symptoms. Rarely brain MRI abnormal.
Asymptomatic Biochemical and gene abnormality without demonstrable adrenal or Common <4 y
neurologic deficit. ∼50% will become symptomatic within 10 y. Rare >40 y
Female X-ALD Carriers
Asymptomatic No evidence of adrenal or neurologic disease ∼50%. Diminishes with age. Most
women <30 y old have this form
Mild myelopathy Increased deep tendon reflexes and distal sensory changes in lower Increases with age. ∼30% of
extremities women >40 y
Moderate to severe Resembles AMN, but later onset and milder Increases with age. ∼15% of
myeloneuropathy women >40 y
Cerebral involvement Rare in childhood. Slightly more common in middle age or later ∼2%
Clinically evident adrenal Rare at any age ∼1%
insufficiency

Source: Adapted from Moser HW, Raymond G, Dubey P. Adrenoleukodystrophy: new appraoches to a neurodegenerative disease. J Am Med Assn 2005;294:3131–3134.

acuity, a gradual disturbance in gait, and slight intellectual impairment. appear with time. Spinal cord and peripheral nerve involvement may
Abnormal skin pigmentation or other signs and symptoms of adrenal occur without central symptoms and, rarely, adrenal insufficiency may
insufficiency sometimes precede neurologic abnormalities; in other occur without neurological involvement (102–104).
cases, adrenal symptoms never develop. About 10% of patients pres- The other major form of this disorder is characterized mainly by
ent acutely with seizures, acute adrenal crisis, acute encephalopa- axonal degeneration in the brainstem and spinal cord; this form is
thy, or coma (101). Progression of the disease is usually fairly rapid. called adrenomyeloneuropathy (105). Adrenomyeloneuropathy typi-
Hypotonia, seizures, visual complaints, and difficulty in swallowing cally presents in young adults (mean age 28 years) with incontinence,

Barkovich_Chap03.indd 96 5/6/2011 12:07:57 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 97

progressive paraparesis, or progressive cerebellar dysfunction (100); Imaging studies show several characteristic patterns. About 80% of
peripheral neuropathy may be detected (94). Affected patients can affected patients with cerebral ALD have predominant posterior white
develop inflammatory demyelination in the cerebrum at any age; this matter involvement (108); the first area affected in this group is the mid-
form is referred to as “cerebral AMN” and also most commonly occurs dle of the callosal splenium. CT reveals low attenuation, starting in the
during the third and fourth decades (94,97). In addition, although splenium of the corpus callosum and extending into the central parietal
this is an X-linked disorder and presents predominantly in males, and occipital white matter (corresponding to what later becomes Zone
about 20% of heterozygous females develop neurological disability A described earlier, Fig. 3-4A); very early in the course of the disease,
that resembles adrenomyeloneuropathy but is of later onset (mean involvement may be restricted to the central splenium. The anterior
age 43 years, range 8–75 years) and somewhat milder than in affected edge of this low attenuation region often shows contrast enhancement,
males (100,106). The reasons for the differing clinical presentations of secondary to an immune-mediated inflammation (Zone B). MR scans
patients with identical mutations have not been clarified, but a likely demonstrate marked T1 hypointensity and T2/FLAIR hyperintensity
cause in women is variable X chromosome inactivation. (Fig. 3-3B), increased diffusivity, and reduced diffusion anisotropy in
The diagnosis of X-linked adrenoleukodystrophy is confirmed by Zone A (109), characteristically with enhancement of the inflamma-
chromosomal studies and by assay of plasma, red cells, or cultured skin tory leading edge of demyelination (Zone B, Fig. 3-4C) if paramagnetic
fibroblasts for the presence of increased amounts of very long chain contrast is administered. The enhancing areas typically show reduced
fatty acids (104,107). False negative results occur in about 15% to 20% diffusion (Fig. 3-4D) because of the increased cellularity. The presence
of women heterozygotes. Therefore, women suspected of this disorder of contrast enhancement seems to be associated with clinical worsening
should undergo mutation analysis (92). of the disease (110). In early phases of the disease, the subcortical white

FIG. 3-4. Adrenoleukodystrophy; classic


imaging appearance. A. Axial noncontrast
CT shows low attenuation in the callosal
splenium (s), peritrigonal white mat-
ter (p), and posterior thalami (t). B. Axial
FLAIR image shows abnormal hyperinten-
sity involving the callosal splenium, poste-
rior periventricular and deep white matter
(sparing subcortical white matter), posterior
thalami, and posterior aspect of the poste-
rior limb of the internal capsule. C. Axial
postcontrast T1-weighted image shows a
rim of enhancement (white arrowheads) in
the inflammatory zone that surrounds the
area of damaged white matter. D. Diffusion
weighted image shows that the inflamma-
tory zone is slightly hyperintense (white
arrowheads), indicating reduced diffusivity.

Barkovich_Chap03.indd 97 5/6/2011 12:07:57 AM


98 Pediatric Neuroimaging

FIG. 3-4. (Continued) E and F. Map from a two-dimensional spectroscopic imaging acquisition (E) and the spectra themselves with echo time of 144 ms
(F). The spectra show high choline, low creatine, and low NAA in the peritrigonal regions (voxels 37, 41, 44, 48) and generally low metabolite levels in the
splenium (voxels 38–40 and 45–47).

matter is spared on standard imaging sequences (Fig. 3-4) (46,47), but individuals with ALD, particularly in those with stable MRI exams
diffusion tensor imaging may show mildly increased water diffusivity and in siblings (of patients with ALD) who are known to have the
and reduced anisotropy (109). The corticopontine and corticospinal ALD mutation but are not symptomatic. Heterozygous women have
tracts in the pons and medulla often show T2 hyperintensity (Fig. 3-5) reduced NAA/Cr and NAA/Cho in the corticospinal tracts (both in
(111) and enhancement. The splenium of the corpus callosum appears the cerebral hemispheres and in the internal capsules) and reduced
with low signal intensity on the midline sagittal, T1-weighted image NAA/Cho in the parietooccipital white matter when compared to age-
(112); in chronic disease, it is small and atrophic. Less common pat- matched controls (106).
terns have been described (47,113,114), including calcification in the The imaging findings in adrenomyeloneuropathy (Fig. 3-7) differ
parietooccipital white matter. from those in classic childhood form of adrenoleukodystrophy in that
The second most common pattern is predominantly frontal the cerebrum is less commonly involved, whereas the cerebellar white
involvement (Fig. 3-6) (115), seen in about 15% of affected patients matter and corticospinal tracts (in the internal capsules and brainstem)
(108). In this pattern, the genu of the corpus callosum, frontal white are more commonly involved (Fig. 3-7) (105,119). Lesion progression
matter, anterior limbs and genus of the internal capsules, and, occasion- appears to be slower than in ALD (119). No inflammatory component
ally, the cerebellar white matter are predominantly involved. Contrast is present and, therefore, no enhancement is seen.
enhancement is seen on the peripheral margins (Zone B), as in the pos- No extensive radiologic differential diagnosis exists for the classic
terior form. Other patterns include predominantly unilateral involve- pattern of ALD. Another peroxisomal disorder, acyl CoA oxidase defi-
ment restricted to an entire hemisphere, both frontal and occipital ciency, has very similar imaging findings, with early involvement of
(Fig. 3-5) (107), and a pattern with supratentorial involvement the pontomedullary corticospinal tracts, and cerebellar white matter,
restricted to the genus of the internal capsules (111). followed by involvement of the splenium, and deep parietooccipital
Affected heterozygous women usually have normal MR scans but white matter (120). However, the clinical presentation is quite differ-
may have findings similar to those in affected boys, with involvement ent, as both boys and girls are affected, often with delayed cognitive and
of parietooccipital white matter and involvement of the corticospinal motor development, followed by developmental regression during the
tracts (106). third year of life (120).
Proton MRS shows decreased NAA; increased choline, glutamine,
and glutamate; decreased myo-I; and increased lactate resonances in Leukoencephalopathy with Vanishing White Matter
areas of active disease (116,117). In more chronic disease, or with Leukoencephalopathy with vanishing white matter (VWM, OMIM
longer echo times, findings may be limited to low NAA and elevated 603896, formerly called childhood ataxia with diffuse central nervous
choline (Figs. 3-4 and 3-6). MRS shows abnormalities before the system hypomyelination, CACH) is a disorder in which patients typi-
MRI study becomes abnormal in some children (116) and in normal cally have normal development in early childhood, then develop pro-
appearing white matter peripheral to the regions of T2 prolongation gressive ataxia and spastic diplegia with relapsing-remitting phases,
and enhancement; this area of abnormal MRS may represent Zone development of bulbar symptoms, optic atrophy, and epilepsy, leading
C. Indeed, it is suggested that reduction in the NAA/choline ratio to death in the second decade of life. Involvement of the peripheral
to below 5.0 is predictive of disease progression within the next 2 nervous system is uncommon (121). Onset occurs from childhood
to 3 years (118). Therefore, proton MRS should be performed in all to early adulthood (122) or even middle age (123); rarely patients

Barkovich_Chap03.indd 98 5/6/2011 12:07:58 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 99

FIG. 3-5. Severe ALD with atypical pattern. A. Axial T2-weighted image shows involvement of the corticospinal and corticopontine tracts in the ventral
pons (black arrows) as well as the middle cerebellar peduncles (white arrows). B. Axial T2-weighted image shows involvement of the genus of the internal
capsules (white arrows) with sparing of the more posterior aspects of the capsules. C. Axial postcontrast T1-weighted image shows enhancement of both the
genu and splenium of the corpus callosum (white arrows). D. Coronal T2-weighted image shows extension of abnormal hyperintensity from the internal
capsules (white arrowheads) inferiorly into the pontine corticospinal tracts (black arrows).

may present as neonates (124) or in infancy (or even as fetuses) and Adult variants are also described, usually presenting in the teenage
have rapidly fatal courses (125,126). Patients tend to have episodes years or 20s with occasional seizures or psychiatric symptoms or
of deterioration after trauma or infection (127–129) or sometimes dementia (136); some affected adult women have associated ovarian
fright (130). Rarely, extreme macrocephaly may develop (131,132). failure (137).
Analysis of CSF, serum, or urine may show elevation of glycine (133). The disease is familial and appears to be autosomal recessive. It can
Cree leukoencephalopathy (134), described in previous editions of be caused by mutations in any of the five genes encoding subunits of
this book as a distinct disorder in Native American populations (par- the eukaryotic translation initiation factor EIF2B: EIF2B1 at 2p23.3,
ticularly the Cree and the Chippewa), is an early-onset form of VWM, EIF2B2 at 14q24, EIF2B3 on chromosome 12, EIF2B4 at 1p34.1, or
with onset between 3 and 9 months (126). Affected children typically EIF2B5 at 3q27 (34,138). The mechanism of cell injury is incom-
present with abnormalities of tone (hypotonia or spasticity), then pletely understood, but is believed to be related to stress conditions
progress to seizures, posturing, and death within a few months (134). (heat stress, head injury) that result in a reduction in protein synthe-
Imaging findings are identical to those in early-onset VWM (135). sis within affected cells (121,136); glial cells appear to be selectively

Barkovich_Chap03.indd 99 5/6/2011 12:08:01 AM


100 Pediatric Neuroimaging

FIG. 3-6. Atypical adrenoleukodystrophy with predominant frontal involve-


ment. A. Axial T2-weighted image shows abnormal T2 hyperintensity of the
callosal genu and the surrounding frontal white matter (white arrows). B and
C. Map from a two dimensional spectroscopic imaging acquisition (B) and
the spectra themselves (C), acquired with echo time of 144 ms. The spectra are
abnormal in the callosal genu and frontal white matter (voxels 1–8), showing low
NAA and increased choline. Spectra from the region of the callosal splenium and
peritrigonal white matter (voxels 9–21) are normal.

FIG. 3-7. Adrenomyeloneuropathy. A. Axial


T2-weighted image through the mid-pons
shows abnormal hyperintensity in the lateral
pons (arrowheads) and cerebellar white mat-
ter (small arrows). B. Axial FLAIR image at
the level of the bodies of the lateral ventricles
is normal.

Barkovich_Chap03.indd 100 5/6/2011 12:08:02 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 101

TABLE 3-8 MRI Criteria for the Diagnosis of VWM


Obligatory Criteria
1. Cerebral white matter shows either diffuse or extensive signal abnormalities; the immediately subcortical white matter may be spared.
2. Part or all of the abnormal white matter has a signal intensity close to or the same as cerebrospinal fluid on proton density or FLAIR
images, suggestive of white matter rarefaction or cystic destruction.
3. If proton density and FLAIR images suggest that all cerebral white matter has disappeared, there is a fluid-filled distance between
ependymal lining and the cortex, but not a total collapse of the white matter.
4. The disappearance of the cerebral white matter occurs in a diffuse “melting away” pattern.
5. The temporal lobes are relatively spared, in the extent of the abnormal signal, degree of cystic destruction, or both.
6. Cerebellar white matter may be abnormal, but does not contain cysts.
7. There is no contrast enhancement.
Suggestive
1. Within abnormal white matter there is a pattern of radiating stripes on sagittal and coronal T1-weighted or FLAIR images; on axial images
dots and stripes are seen within the abnormal white matter as cross-sections of the stripes.
2. Lesions within the central tegmental tracts in the pontine tegmentum.
3. Involvement of the inner rim of the corpus callosum; the outer rim is spared.

Adapted from van der Knaap MS, Pronk JC, Scheper GC. Vanishing white matter disease. Lancet Neurol 2006;5:413–423, with permission.

vulnerable. The effect of the mutation on the function of the protein time of presentation (Fig. 3-8B). Degeneration to CSF intensity starts
probably determines the severity of the disease (136,139); most patients in the central white matter (Fig. 3-8) but eventually involves the entirety
have residual eIF2B activity of 30% to 80%, while carriers have normal of the white matter in the cerebrum; only the ventricular lining is
activity (140). spared at this point (Fig. 3-9E and F). Signal changes of myelination are
Diagnostic criteria, as suggested by van der Knaap et al. (122,129), seen only in the striatal region and cerebellum. Contrast enhancement
include the following: (a) Normal or mild delay of initial motor and has never been reported. Cerebellar white matter may have normal or
mental development. (b) Neurologic deterioration has a chronic pro- abnormal signal (Fig. 3-9), but cavitation is not seen in the cerebellum
gressive course, with episodes of deterioration precipitated by minor or brainstem (136). Cerebellar atrophy varies from mild to severe and
infection, minor head trauma or fright, and may lead to lethargy or primarily involves the vermis (129). In the brainstem, abnormal hyper-
coma. (c) Neurologic signs consist mainly of cerebellar ataxia and spas- intensity is seen initially in the pontine central tegmental tracts, but
ticity. Optic atrophy may develop but is not obligatory. Epilepsy may ultimately involves the ventral pons as well (122,145). Later stages of
occur, but is not the predominant sign of the disease. Mental abilities involvement are characterized by nearly complete loss of white matter,
may also be affected, but not to the same degree as the motor functions. with ex vacuo ventricular enlargement.
(d) MRI criteria as in Table 3-8. When presentation is in neonates, the initial MRI may be normal
Histopathologic analysis of affected brains shows axonal loss, (144) or the cerebral gyri may appear slightly broader than normal,
hypomyelination, demyelination, gliosis, and “foamy” oligodendro- indicating either delayed sulcation or slight white matter swelling
cytes, primarily in the deep white matter (127,141,142). The cortical (124,136); this appearance may be initially interpreted as immaturity.
gray matter, subcortical U fibers, corpus callosum, and internal cap- More commonly, the initial MRI is characterized by white matter that
sules are relatively spared (141,142). Within the ventral pons, sharply is abnormally hypointense on T1-weighted images and abnormally
demarcated symmetric areas of demyelination are seen primarily in the hyperintense on T2-weighted images (Fig. 3-9). On proton density
transverse pontine fibers, with intact pontine nuclei. The dorsal pons and FLAIR images, white matter signal intensity is lower than normal
shows a symmetric area of demyelination involving the central teg- in some regions of cerebral white matter, suggesting white matter rar-
mental tract and the superior central nucleus on both sides. Whereas efaction, and possible impending cystic degeneration. No evidence of
some studies suggest that this is a primary axonopathy, with myelin myelination is seen at term.
being secondarily affected (122), others suggest a primary disease of Proton MR spectroscopy is normal early in the disease. As rar-
oligodendrocytes, resulting in excessive oligodendroglial apoptosis efaction and cavitation ensue, white matter spectra show a progres-
with secondary axonal degeneration (121,141,142), or impaired astro- sive decrease and, ultimately, complete disappearance of all “normal”
cyte function (143). signals; some lactate and glucose remains and may become more evi-
MRI shows diffuse white matter signal abnormality (T1 hypoin- dent as the amplitude of the other signals decreases (127–129). NAA
tensity and T2 hyperintensity) in which the white matter ultimately progressively decreases, whereas Cr is decreased but stable (67). Cortex
cavitates to become CSF intensity on T1- and T2-weighted images maintains a more normal spectrum, apart from a decrease in NAA and
(Figs. 3-8 and 3-9). Diffusivity is variably reduced compared to CSF a variable elevation of lactate and glucose (122,128,146).
on diffusion-weighted images; tissue in the process of cavitating has
reduced diffusivity (144), whereas cavitated white matter has increased Giant Axonal Neuropathy
diffusivity, with signal identical to CSF (Fig. 3-9). Cavitated (cystic) Giant axonal neuropathy (OMIM 256850) is a generalized disorder of
white matter is hypointense on FLAIR and proton density images and cytoplasmic intermediate filaments that results in the accumulation
thus can be differentiated from the hyperintense rarefied (but noncavi- of ovoid aggregates in many different cell types (147). The responsible
tary) white matter (Fig. 3-9). Subcortical fibers may be spared at the gene (GAN) has been mapped to chromosome 16q24.1 and encodes

Barkovich_Chap03.indd 101 5/6/2011 12:08:04 AM


102 Pediatric Neuroimaging

FIG. 3-8. Vanishing white matter disease in a 12 month old


with developmental regression after viral illness. A. Axial
T1-weighted image shows diffuse low signal intensity in the
white matter with several areas of lower intensity (black arrows).
B. Axial T2-weighted image shows extensive hyperintensity of
the white matter; the cavitating areas are more difficult to appre-
ciate than on the T1-weighted image (A). C. Axial diffusion-
weighted image shows the cavitating areas (black arrows) as very
low signal intensity. Note the relative preservation of subcortical
white matter.

a ubiquitously expressed protein that has been named gigaxonin of the lateral ventricles is often hyperintense compared to surrounding
(148,149). Affected patients initially present in childhood with fea- white matter (11). Later in the course of the disease, the corticospinal
tures of a peripheral sensory and motor neuropathy and extremely tracts in the brainstem show T2 hyperintensity and will enhance after
kinky hair (150). However, central nervous system involvement is an paramagnetic contrast administration (11). Thus, this disease has an MR
important component of the disease (151,152). Typically, muscular appearance somewhat similar to X-linked adrenoleukodystrophy. Thal-
hypotonia is noticed early in life, along with delayed motor develop- ami may show T2 hyperintensity (153). MRS shows reduced NAA, but
ment. Walking may be eventually achieved, but is broad-based and increased choline and myo-I (154). Creatine values are roughly normal.
ataxic and then regresses until the child becomes wheel-chair bound.
Ultimately, the neurological exam shows hypotonia, muscle weakness Phenylketonuria
and wasting, absent reflexes, and contractures. Optic discs are pale and Phenylketonuria (OMIM 261600) is an autosomal recessive disorder
visual acuity is diminished. most often caused by a mutation to the PAH gene, which has been
Imaging is nonspecific, showing T1 and T2 prolongation of the mapped to chromosome 12q24.1 (155). This mutation causes phe-
white matter, with sparing of the subcortical U fibers in both the cere- nylalanine hydroxylase (PAH) deficiency, although other biochemical
brum and cerebellum. The posterior limbs of the internal capsules are defects can cause hyperphenylalaninemia and manifest similar signs
commonly involved early (Fig. 3-10A), as is the cerebellar white matter and symptoms (156,157). More than 400 mutations (318 missense
(Fig. 3-10B). On noncontrast T1-weighted images, the ependymal lining mutations) of the PAH gene have been identified (158,159), partially

Barkovich_Chap03.indd 102 5/6/2011 12:08:04 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 103

FIG. 3-9. Vanishing white matter disease in a 9-month old infant. A. Axial FLAIR image shows two levels of abnormality in the cerebral white matter. The
outer layer (o) shows abnormally high water content, while the inner layer (i) shows signal intensity identical to CSF, indicating frank cavitation. Note the
relative sparing of subcortical white matter. B. Axial T2-weighted image appears to show homogeneous signal intensity. T2-weighted images do not reliably
show cavitation. C. Coronal T2-weighted image shows nearly homogeneous hyperintensity of the posterior frontal white matter, as well as involvement
of the periventricular temporal (small white arrows) and cerebellar white matter (large white arrows). D. Coronal diffusion weighted image shows marked
hyperintensity (arrows), indicating reduced diffusion in the pre-cavitary white matter of the centrum semiovale, internal capsules, periventricular tempo-
ral white matter, and ventral cingula. E and F. Vanishing white matter with nearly complete white matter cavitation. First echo (E) and second echo (B)
T2-weighted images show nearly homogeneous isointensity of white matter with the enlarged lateral ventricles. Only the remaining ependymal (arrows) can
be seen to separate the ventricles from the liquefied surrounding white matter. (Figure 3-9E and F courtesy Dr. Jaap Valk, Amsterdam.)

Barkovich_Chap03.indd 103 5/6/2011 12:08:05 AM


104 Pediatric Neuroimaging

FIG. 3-10. Giant axonal neuropathy in a 14-year-old adolescent. Axial T2-weighted image (A) and coronal FLAIR image (B) show extensive cerebral and
cerebellar white matter hyperintensity with relative sparing of subcortical white matter (small white arrowheads) and corpus callosum (white arrows). Note
involvement of the internal capsules (large white arrowheads). (These images courtesy Dr. Susan Blaser, Toronto.)

accounting for the marked clinical, biochemical, and neuroimaging Imaging studies show primarily abnormal signal in the white mat-
variability. Most PAH missense mutations cause misfolding of the PAH ter that corresponds to delayed and defective myelination. In older
protein, resulting in increased protein turnover and loss of enzymatic patients, ventriculomegaly is found as a result of white matter degen-
function (159), with consequent production of compounds (phe- eration (161). MRI shows T2/FLAIR hyperintensity that is initially
nylpyruvic and phenylacetic acids and phenylacetylglutamine) that are seen in the periventricular white matter of the cerebral hemispheres
toxic to the developing brain (59,156). Untreated patients are charac- (Fig. 3-11) (59,162). Subcortical white matter is initially spared. Some
terized by growth retardation; global developmental delay; eczematous reports suggest that the amount of white matter disease is related to
dermatitis; hypopigmentation; and a peculiar musty odor of the urine, the adequacy of biochemical control of the disease (163). In the acute
skin, and hair (155,156). Treatment is by dietary control and, occasion- phase, diffusion is reduced in the affected white matter, possibly due to
ally, dietary supplements (156,158,160); benefit from control of diet is spongiotic changes of myelin (164); some studies suggest a relationship
variable, probably due to the heterogeneity of the disease. between the serum phenylalanine level and degree of reduced diffusion

FIG. 3-11. Phenylketonuria. A. Axial FLAIR image shows abnormal hyperintensity in the frontal and parietal white matter. Note sparing of the subcortical
white matter (white arrows). B. Axial diffusivity (Dav) map shows hypointensity (white arrows), indicating reduced diffusivity, in the affected white matter.

Barkovich_Chap03.indd 104 5/6/2011 12:08:06 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 105

(165). Contrast enhancement has not been reported. The volume of classic type, onset of clinical signs and symptoms occurs during the
the cerebral hemispheres is reduced (166). first week of life when poor feeding, vomiting, dystonia, opisthotonic
In the minority of cases caused by dihydropteridine reductase posturing, and seizures become manifest (156). If the disease is not
deficiency or defect in the synthesis of tetrahydrobiopterin, the fron- recognized and treated, infants develop signs of increased intracranial
tal cortex, occipital cortex, subcortical white matter, and putamina are pressure, become comatose, and may die in a few weeks (156,172).
affected. Putaminal and frontal/occipital subcortical calcification is The severity of neurologic sequelae is strongly correlated with the
often seen in these cases on CT (155). MRI shows cortical and subcor- duration of the acute toxic phase in the neonatal period (173). The
tical atrophy, particularly in the intervascular boundary zones (“water- less severe forms of the disease, with higher residual enzymatic activ-
shed zones”) (11). T1 shortening (T1 hyperintensity) may be present in ity, can present later in childhood with metabolic crises resulting in
damaged regions of cortex. lethargy, irritability, nausea, and vomiting that may progress to stupor
Proton MR spectroscopy shows the presence of a peak from ele- or coma (171).
vated phenylalanine at 7.37 ppm (163,167). The size of this peak may Imaging is normal in the first few days of life. Because classic
be useful for monitoring treatment, although it is currently not trivial maple syrup urine disease presents in the neonatal period, it is one of
to assess this peak on most clinical MR scanners. Other peaks seem the few metabolic diseases in which transfontanelle sonography may
to be normal. Diffusion imaging reveals reduced water motion in the play a role in diagnosis. Starting with the onset of symptoms, cranial
affected regions of the brain (164) in younger children while it shows sonography shows symmetric increase in echogenicity of periventricu-
increase in the median and minor eigenvalues and decreased fractional lar white matter, basal ganglia, and thalami in the acute stage (174).
anisotropy in the deep parietal and occipital white matter in children The CT and MR findings are quite characteristic, revealing profound
older than 3 years (168). localized edema (low density on CT, long T1, T2 on MR) in the deep
cerebellar white matter, dorsal brainstem, cerebral peduncles, poste-
Maple Syrup Urine Disease rior limb of the internal capsule, sensorimotor tracts in the centrum
Maple syrup urine disease (OMIM 248600) is a genetically heteroge- semiovale, and, sometimes, globi pallidi (Fig. 3-12A–D) (59,172).
neous disorder; the causative genes encode the catalytic proteins of The regions involved correspond to those that are myelinated or
the branched-chain alpha-ketoacid dehydrogenase complex (BCKD) myelinating at the time of birth. Generalized vasogenic edema of the
that catalyzes the oxidative decarboxylation of the branched-chain cerebral hemispheres may be superimposed on the localized abnor-
amino acids leucine, isoleucine, and valine (169). Treatment requires malities (172,175), particularly in the first weeks of life. Diffusion
lifelong dietary restriction and monitoring of branched-chain amino imaging may be very useful at this stage of the disease, as the areas
acids to avoid brain injury. Despite careful management, children with (intramyelinic (176) ) MSUD edema (cerebellar white matter,
commonly suffer from metabolic decompensation in the context of dorsal brainstem, cerebral peduncles, corticospinal tracts, and globi
catabolic stress associated with nonspecific illness (170). The mecha- pallidi) show reduced diffusivity during the acute phase of the disease
nisms underlying this decompensation and brain injury are poorly (Fig. 3-12E–G), with ADC values falling to 20% to 30% of their normal
understood. Five clinical phenotypes are described; these seem to values (177). When relapses occur after the brain has fully or nearly
correlate with the degree of residual enzymatic activity (171). In the fully myelinated, reduced diffusion will be seen in most of the white

FIG. 3-12. Maple syrup urine disease. A. Axial noncontrast CT scan shows hypodensity in the frontal white matter ( f ) as well as in the globi pallidi (white
arrows). B–D. Axial T2-weighted images show abnormal hyperintensity in the brainstem (b), cerebellar white matter (c), frontal white matter ( f ), globi
pallidi (g), posterior limbs of the internal capsules (black arrowheads), optic radiations (black arrows), and the entirety of the subcortical and deep cerebral
white matter. E–G. Axial diffusivity (Dav) maps shows reduced diffusivity in the brainstem (b), cerebellar white matter (c), globi pallidi (g), posterior limbs
of the internal capsules and lateral thalami (white arrowheads), optic radiations (white arrows), and perirolandic white matter (w). H and I. Short echo
(26 ms) and long echo (288 ms) proton MRS from the basal ganglia show the characteristic broad peak at 0.9 ppm that represent branched chain amino
acids and branched chain ketoacids (bca). This peak is differentiated from the normal macromolecular peak at 0.9 ppm by its presence on long echo spectra.
Reduced NAA and lactate are also common findings.

Barkovich_Chap03.indd 105 5/6/2011 12:08:07 AM


106 Pediatric Neuroimaging

FIG. 3-12. (Continued)

Barkovich_Chap03.indd 106 5/6/2011 12:08:08 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 107

matter of the cerebral and cerebellar hemispheres (175), supporting the disorders cause intimal irregularities with subsequent arteriosclerosis,
concept that myelinated or myelinating white matter is preferentially arterial dissection, arterial thromboembolism, and venous thrombosis
affected and that the reduced diffusion is the result of intramyelinic in children and young adults. The main factor appears to be excessive
edema. After the acute phase of the disease has resolved, the patients homocysteine, which generates superoxide and hydrogen peroxide,
are left with a variable degree of brain damage, dependent upon how changes coagulation factor levels (encouraging clot formation), pre-
quickly treatment was initiated (156,172). vents small arteries from dilating (increasing likelihood of obstruc-
In the milder (intermittent, intermediate) forms of the disease that tion), and causes proliferation of smooth muscle cells in arterial walls
present with decompensation later in infancy or in early childhood, the (181). Patients have characteristic clinical abnormalities, including dis-
main finding is a profound lack of myelination that may be superim- location of the lens, osteoporosis, and thinning and lengthening of the
posed upon damage to the dorsal brainstem, cerebellar white matter, long bones. Half of untreated patients will suffer from strokes before
internal capsules, and globi pallidi. These are the same regions affected the age of 30 years. Affected patients can also manifest mental retarda-
in the classic form of the disease during the early postnatal crisis. From tion, seizures, and dystonia. Outcome is markedly improved in vitamin
the imaging perspective, the intermittent form of maple syrup urine B6 responsive patients and nonresponsive patients who are compliant
disease may resemble early Canavan disease (including diffusion find- with treatments (182). In most cases, neuroimaging studies reveal mul-
ings, since both present with reduced water diffusion in affected white tiple small infarcts of different ages throughout the brain. Demyelina-
matter structures), but the clinical context and the laboratory findings tion, white matter vacuolization, and spongy degeneration have been
allow easy differentiation. observed only rarely (155,183).
In the acute phase of illness, proton MRS of patients with maple
syrup urine disease shows slight reduction of NAA and slight eleva- 5, 10 Methylenetetrahydrofolate Reductase Deficiency (MTH-
tion of lactate; these abnormalities disappear after treatment. More FRD) 5, 10 methylenetetrahydrofolate reductase deficiency (OMIM
importantly, a somewhat broad peak at 0.9 ppm (Fig. 3-7H and I) is 236250) causes homocystinuria and hypomethioninemia due to
also present; it is believed to represent resonances of methyl protons mutations of the 5,10 alpha methylenetetrahydrofolate reductase gene
from branched-chain amino acids and branched-chain alpha ketoac- (MTHFR at 1p36.3); at least 24 mutations have been reported to cause
ids that accumulate as a result of defective oxidative decarboxylation disease (184). There is a direct correlation between methylenetetra-
of leucine, isoleucine, and valine (178,179). It can be differentiated hydrofolate reductase activity in fibroblasts and clinical severity of
from the “macromolecule” peak typically seen at about 0.9 ppm on the disease (185). This disorder has less pronounced vascular pathol-
short TE spectra (see Chapter 1) because it is present on long echo ogy than cystathionine beta synthase deficiency and cerebral infarcts
(TE = 270–288 ms, Fig. 3-12I), as well as short echo spectra (Fig. are rare; however, children with abnormal MTHFR activity may be
3-12H) (178). This peak may not totally disappear, even in patients predisposed to cerebral infarction (186), particularly due to venous
who respond to therapy, because branched-chain amino acids are thrombosis (187). Patients with this disorder most commonly pres-
essential nutrients and, therefore, total dietary withdrawal is not ent in childhood with gait abnormalities, seizures, and psychomotor
possible. retardation of variable severity. Affected patients may also present in
infancy with hypotonia, progressive lethargy, recurrent apnea, and
Hyperhomocysteinemia (Formerly Known as Homocystinuria) seizures; progression to respiratory failure, coma, and death may
Homocysteine is the end product of the so-called transmethylation ensue (188). Certain mutations (in particular, the 677 C-T mutation
pathway, a multistep metabolic process by which methyl groups from in the 5,10 methylenetetrahydrofolate reductase gene) are risk factors
methionine are transferred to essential acceptor molecules (DNA, for neural tube defects, as they impede the metabolism of folic acid
neurotransmitters, proteins, phospholipids, polysaccharides). Homo- (see Chapter 9 for comments regarding the importance of folic acid
cysteine may either be recycled into methionine (remethylation in preventing neural tube defects). MTHFR mutations are also a risk
pathway) or catabolized into cystathionine, cysteine, and sulfate (trans- factor for stroke in childhood (see Chapter 4 for discussion of child-
sulfuration pathway). The three key enzymes involved in the pathogen- hood stroke) and for cervical arterial dissections (189). Treatment
esis of hyperhomocysteinemia are cystathionine beta-synthetase (on with betaine, folic acid, and cobalamin is recommended to prevent or
the transsulfuration pathway), methionine synthetase (also known as 5- mitigate symptoms (190).
methylhydrofolate:homocysteine methyltransferase), and 5,10-methyl- Demyelination of the white matter of the brain and spinal cord is the
ene-tetrahydrofolate reductase (on the remethylation pathway); folate, most commonly observed pathology in 5,10-methylenetetrahydrofolate
B12 (cobalamin), and B6 (pyridoxine) vitamins play a role as cofactors. reductase deficiency. The demyelination starts as perivascular foci of
Hyperhomocysteinemias may, therefore, develop in several different spongiform change, that is, separation of myelin layers. Less com-
conditions, which include cystathionine beta-synthetase enzyme defi- monly, demyelination of the dorsal and lateral columns of the spinal
ciency (“classic” homocystinuria), methionine synthetase deficiency, cord may be present.
folate deficiency, defect of folate metabolism (5,10-methylene-tetrahyd MRI shows infarctions, when they occur, in addition to nonspe-
rofolate reductase deficiency), cobalamin (vitamin B12) deficiency, and cific white matter abnormalities ranging from small foci of high signal
defects of cobalamin metabolism (methionine synthetase deficiency in intensity (predominantly medial and bifrontal) to more diffuse, gener-
isolated methylcobalamin and combined methylcobalamin and adeno- alized white matter T1 hypointensity and T2 hyperintensity (Fig. 3-13)
sylcobalamin deficiencies) (180). (155,183,188). Proton MR spectroscopy is reported to show reduced
The major clinical manifestations of hyperhomocysteinemia are NAA in the white matter (191).
ophthalmological (lens dislocation), vascular (steno-occlusive arte-
rial and venous disease), and neurological (demyelination in brain and Errors Affecting Cobalamin (Vitamin B12) Metabolism Errors
spinal cord). affecting cobalamin (vitamin B12) metabolism have been shown to
cause homocystinuria and methylmalonic aciduria (MMA/HC), a
Cystathionine Beta Synthase Deficiency Cystathionine beta syn- genetically heterogeneous disorder caused by impaired conversion
thase deficiency (OMIM 236200) is caused by mutations in the cystathi- of dietary vitamin B12 (cobalamin) to its two metabolically active
onine beta synthase (CBS) gene at 21q22.3. The resulting biochemical forms, methylcobalamin and adenosylcobalamin. The consequence

Barkovich_Chap03.indd 107 5/6/2011 12:08:10 AM


108 Pediatric Neuroimaging

FIG. 3-13. Hyperhomocysteinemia (5, 10


methylenetetrahydrofolate reductase deficiency)
in a 10-month old infant. Axial T1 (A) and T2
(B)-weighted images show marked hypomyeli-
nation throughout the brain.

of reduction of these compounds is a buildup of methylmalonic acid and smudgy T2 prolongation in the peritrigonal white matter. Proton
and homocystine and their increased excretion in the urine (192). The MRS is reported to show loss of NAA and presence of lactate in the
different forms of the disorder are classified according to complemen- basal ganglia (200).
tation groups of cells in vitro: cblC (OMIM 277400, the most com-
mon, resulting from mutations of the MMACHC gene at 1p34.1), cblD Biotinidase Deficiency
(OMIM 277410), and cblF (OMIM 277380) (193). These disorders Biotin is an imidazole derivative that acts as a cofactor for many
differ from the isolated forms of methylmalonic acidemia and homo- enzymes, including pyruvate carboxylase, propionyl CoA carboxylase,
cysteinemia, which have different biochemistry, different genetics, and and beta-methylcrotonyl-CoA carboxylase, which play a role in fatty
different clinical-neuroradiological attributes. Cerebral infarcts and acid synthesis, amino acid catabolism, and gluconeogenesis (201). It
vascular thrombosis have not been reported, although intimal thicken- has a twofold enzymatic function: catalyzing the detachment of bio-
ing and fibrosis are generally present. Not surprisingly, affected patients tin from proteins through which alimentary biotin enters the body;
have a variable clinical picture (193–196). Some present in infancy with and recycling biotin after it is released from the various carboxylase
feeding difficulties, hypotonia, failure to thrive, seizures, microcephaly, enzymes. Biotinidase deficiency (also called multiple carboxylase
megaloblastic anemia, or developmental retardation; some develop deficiency, OMIM 253260) is caused by mutation of the biotinidase
hemolytic-uremic syndrome. Others may present later in childhood gene (BTD) at chromosome 3p25; the loss of biotin function inhibits
or in adolescence or adulthood with dementia, progressive gait dis- the cleavage of biotin from a compound named biocytin, resulting
turbance, acute neurologic dysfunction, or myelopathy (155,192,197). in reduced function of these biotin-dependent carboxylases. Children
Diagnosis is made by typical metabolic profile with increased urinary with this autosomal recessive disorder can become symptomatic at
excretion of methylmalonic acid, methylcitric acid, and homocystine, any age, but typically present in early to middle infancy with acute
increased plasma methylmalonic acid and free disulfide homocys- or insidious lethargy, hypotonia, anorexia or vomiting, developmental
tine, and hypomethioninemia (193). Confirmation of an intracellu- delays, ataxia, seizures, or coma (180,201). Dermatologic complica-
lar defect of cobalamin metabolism is made by analysis of cultured tions are common, including alopecia, rashes, and mucocutaneous
fibroblasts (193) or by identification of mutation of a causative gene candidiasis. Hearing loss and optic atrophy can be long-term compli-
(198). Treatment includes hydroxycobalamin, betaine, and carnitine, cations. When presenting later in childhood or adolescence, children
but response and outcome vary (193,194). Pathologic analysis of the may complain of motor weakness or diminishing visual acuity (201).
brains of affected patients typically shows spongiform demyelination With early diagnosis, response to pharmacologic doses of biotin is
of the white matter. rapid.
Neuroimaging is variable, reflecting the heterogeneity of the dis- There are few reports of imaging studies in biotinidase deficiency.
order. It may be normal early in the course of the disease, particularly MRI shows early loss of brain volume, with increased ventricular size
when presentation is in adulthood (199). MRI shows the effects of the and increased subarachnoid spaces; subdural hygromas or hematomas
myelin destruction, with variably reduced volume of cerebral white may develop (202,203). T1- and T2-weighted images show reduced
matter and abnormal T2 hyperintensity of the deep white matter with myelination, with reduced diffusion and increased fractional anisot-
sparing of the internal capsule, peripheral white matter, and subcorti- ropy in the deep, white matter of the cerebral hemispheres (Fig. 3-14)
cal U fibers (155,183,192,193). Other reports describe hydrocephalus (204). One report suggests that, uncommonly, cortical injury may
with diffuse white matter swelling, patchy cavitation of the basal gan- occur (203). Proton MRS at long and intermediate (135 ms) echoes
glia, and loss of T2 hypointensity of white matter (200). In the sin- shows marked elevation of lactate with decreased NAA and choline
gle case of cobalamin C deficiency at our institution, the MRI shows (203). All of these abnormalities reverse rapidly if therapy is started
features of enlarged cortical sulci, mildly enlarged lateral ventricles, early (204).

Barkovich_Chap03.indd 108 5/6/2011 12:08:10 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 109

FIG. 3-14. Biotinidase deficiency; 8-week-old infant with seizures. A. Axial T1-weighted image shows delayed myelination with absence of any hyperin-
tensity in the anterior aspect of the posterior limb of the internal capsule. B. Axial diffusivity (Dav) map shows abnormally reduced diffusivity (low signal
intensity, arrows) in the deep cerebral white matter; fractional anisotropy (FA) was abnormally increased in the same areas. The Dav and FA normalized
after treatment with biotin. (These images courtesy of Dr. J Soares-Fernandes, Braga, Portugal.)

Methionine Adenosyltransferase Deficiency and MR findings of abnormal cerebral white matter (205–207). Those
Methionine adenosyltransferase (MAT) I/III (OMIM 250850),expressed with the highest plasma methionine levels also have mild to moderate
solely in liver, catalyzes the biosynthesis of S-adenosylmethionine abnormal elevations of plasma total homocysteine, which may lead to
from methionine. MAT I/III deficiency, caused by mutations in the a misdiagnosis of homocystinuria due to cystathionine beta synthase
MAT1A gene at 10q22, is characterized by persistent isolated hyper- deficiency (see IV.B.1.h) (207).
methioninemia (205–207). Clinical manifestations are variable and On imaging studies, patients with MAT I/III deficiency show abnor-
poorly understood. Patients with moderate impairments in MAT I/III mal T1 and T2 prolongation in the cerebral white matter, with sparing
activity have no clinical manifestations, but those with severe impair- of the subcortical U fibers (Fig. 3-15A). Diffusion images show reduced
ments of MAT I/III activity have neurological signs and symptoms diffusivity (Fig. 3-15B). Proton MRS findings have not been reported.

FIG. 3-15. Methionine adenosyltransferase I/III deficiency in a 3-year-old boy. A. Axial T2-weighted image shows abnormal hyperintensity in the cere-
bral white matter. The subcortical white matter (white arrows) is spared. B. Axial diffusion weighted image shows high signal intensity, indicating reduced
diffusivity, in the affected white matter. (This case courtesy Dr. Jun-ichi Takanashi, Chiba, Japan.)

Barkovich_Chap03.indd 109 5/6/2011 12:08:10 AM


110 Pediatric Neuroimaging

Oculocerebrorenal Syndrome (Lowe Syndrome) be apparent if the scan is performed prior to surgery (Fig. 3-16A).
Lowe syndrome (oculocerebrorenal syndrome, OMIM 309000) is an MRI shows three distinct lesions: (a) dilated perivascular spaces in
X-linked recessive disorder that predominantly affects males. In addi- the periventricular and deep white matter (Fig. 3-16B), (b) confluent
tion to the central nervous system, the lens and kidneys are involved. regions of T2/FLAIR hyperintensity that spare the subcortical white
The responsible gene, located at Xq26.1 is called OCRL1; it encodes a matter in the early course of the disease (Fig. 3-16D), and (c) later in the
phosphatidylinositol-4,5-biphosphate-5 phosphatase that is located in disease course the appearance of multiple small spherical cysts in deep
the Golgi complex (208–210) and is postulated to play a role in mediat- and periventricular white matter (Fig. 3-16C and D) (212,215,216).
ing trafficking of proteins from endosomes to the trans-Golgi network The cysts may be the result of progressive enlargement of the perivas-
(2,11). Primary clinical manifestations include congenital cataracts, cular spaces. Proton MRS in some patients shows a slight to moderate
glaucoma, mental retardation, renal tubular dysfunction (Fanconi syn- elevation of the peak at 3.56 ppm that is typically assigned to myo-I
drome—proteinuria, generalized amino aciduria, carnitine wasting, (217,218). This may represent accumulation of phosphatidyl inositol
and phosphaturia), and metabolic bone disease leading to arthropathy 4,5-biphosphate, which is not degraded, or may represent astrogliosis
(212,213). Mild cases have been reported with cataracts and glomerular in response to tissue injury from the disease. However, not all patients
disease, but lacking renal tubular defects and mental retardation (214). with Lowe syndrome have elevation of this peak. It has been postulated
Imaging studies are characteristic. CT shows nonspecific hypoden- that the size of the peak at 3.56 ppm is related to the severity of the
sity in the cerebral white matter; calcification of the lens (cataract) may enzymatic defect (217).

FIG. 3-16. Lowe syndrome; early and later phase images. A. Axial CT scan in early infancy shows bilateral calcifications (white arrows) in the ocular lenses,
diagnostic of congenital cataracts. B. Axial T2-weighted image at age 15 months shows myelination delay and the presence of multiple dilated perivascular
spaces. Some more rounded foci of hyperintensity (white arrows) likely represent early cyst formation. C. Axial T1-weighted image in childhood shows ven-
triculomegaly and multiple small cysts (black arrows) in periventricular and deep white matter of the cerebral hemispheres. D. Coronal FLAIR image shows
hyperintensity in the periventricular and deep white matter. Not the multiple small, hypointense cysts within the abnormal white matter.

Barkovich_Chap03.indd 110 5/6/2011 12:08:11 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 111

Merosin-Deficient Congenital Muscular Dystrophy (MDC1A) arthrogryposis, resulting from diminished movements in utero. As
The congenital muscular dystrophies (CMDs) are a heterogeneous a result of their hypotonia, deep tendon reflexes are diminished and
group of disorders characterized by hypotonia, weakness, and fre- motor milestones are delayed. Intelligence is usually normal but may
quently congenital contractures in conjunction with dystrophic changes be slightly impaired. Seizures may develop, but seem to be uncom-
of muscle as demonstrated by muscle biopsy (219). At present, CMDs mon (219,222). Serum creatine kinase is moderately elevated, usually
are separated into three groups: (a) patients with normal brain MR to levels near or above 1,000 U/L (223). Some patients, however, have
and no central neurologic signs or symptoms; (b) those with central a milder course with later onset, slowly progressive muscle weakness
neurologic signs or symptoms whose imaging studies show abnormal (designated “limb-girdle muscular dystrophy”), and lower values of
myelination but normal cerebral and cerebellar cortex (almost all of creatine kinase; in this group, mental retardation is found in only 6%
these patients have absence of merosin, a molecular component of the and seizures in 8% (224).
basement membrane of muscle fiber, on muscle biopsy (220) ); and (c) MRI is the neuroimaging study of choice. Affected patients typi-
those with central neurologic signs and symptoms and imaging studies cally have delayed myelination or hypomyelination of the central cere-
showing associated cerebral cortical malformations. The first group is bral white matter (Fig. 3-17). Mild pontine and cerebellar vermian
not considered in this book. The second is included here and the third hypoplasia is present in 10% to 20% and, when present, may be help-
is discussed in Chapter 5, in the section on malformations of cortical ful in suggesting the diagnosis (145,220); occipital agyria is present in
development secondary to abnormal neuronal migration. about 10% (Fig. 3-17). Short echo proton MRS shows mildly reduced
Merosin-deficient congenital muscular dystrophy (OMIM 607855, concentrations of most metabolites in white matter (but essentially
also called MDC1A) seems to be recessively inherited and the genetic normal myo-I and normal metabolite ratios) (225), while diffusion-
defect appears to be a result of mutation of the lamina-alpha-2 gene weighted imaging shows increased water diffusion in the affected white
(LAMA2) located at chromosome 6q22-23 (221). Most affected matter (226). Definitive diagnosis is made by a combination of muscle
patients are hypotonic and weak from birth. They may be born with biopsy, brain MR, and clinical examination.

FIG. 3-17. Merosin deficient congenital muscular dystrophy. A. Sagittal T1-weighted image shows
atrophy of the cerebellar vermis, particularly in the central portions (white arrows). B. Paras-
agittal T1-weighted image shows localized occipital agyria (black arrows). C. Axial T2-weighted
image shows abnormal hyperintensity of the periventricular and deep white matter of the cerebral
hemispheres. Note relative sparing of the subcortical white matter (white arrows).

Barkovich_Chap03.indd 111 5/6/2011 12:08:12 AM


112 Pediatric Neuroimaging

FIG. 3-18. Mucolipidosis type IV. A. Midline sagittal T1-weighted image shows a small, thin corpus callosum (white arrows); the splenium is particularly
thin. The ventral pons is small. B. Coronal T2-weighted image shows abnormal hyperintensity of the periventricular and deep white matter, with sparing
of the subcortical fibers. The cerebellar hemispheres appear atrophic with enlarged fissures.

Mucolipidosis Type IV adults. X-linked forms are often associated with mutation of the PLP1
Mucolipidosis type IV (OMIM 252650) is an autosomal recessive dis- gene; this form is discussed in the section on Pelizaeus-Merzbacher
order of lysosomal storage that occurs most commonly in the Ashke- disease later in this chapter. Autosomal recessive forms often present
nazi Jewish population. It is caused by mutation of the MCOLN1 gene in childhood and are also very heterogeneous (234). Of note, about
at chromosome 19p13.2-13.3; this gene encodes the MLN1 protein, 30% of these patients have a strikingly thin corpus callosum when
which may play a major role in calcium transport, regulating lysosomal imaged by MRI, associated with variable (but often severe) cognitive
exocytosis and potentially other phenomena related to the trafficking deficit (235). Nearly all of these patients appear to have mutations
of late endosomes and lysosomes (227,228). Typically, the first sign of the spatacsin gene (SPG11) at chromosome 15q13-15 (236), with
of disease is progressive visual impairment. Psychomotor retardation resultant impaired axonal transport in long projection neurons (237).
becomes apparent in early childhood; this includes delayed motor and A patient with spatizin (SPG15) mutation has also been reported and
speech development and may be static or progressive (69). Patients may appears to have a very similar phenotype (238). Children with spatac-
rarely develop spastic paraplegia or lack corneal or retinal abnormali- sin mutations present with slowly progressive paraparesis (clumsiness,
ties (229). Seizures are rare. Examination reveals corneal opacification, difficulty walking, or frequent falls), lower extremity hyperreflexia,
progressive retinopathy, and hypotonia, which progresses to spasticity. mental retardation of variable severity and, sometimes, upper extrem-
No organomegaly has been reported. ity signs (236,239). MRI shows slowly progressive cerebral white
Imaging studies show striking hypogenesis and hypoplasia of the matter and cortical atrophy with progressive T2 prolongation of the
corpus callosum (Fig. 3-18A). The entire corpus is dramatically thin, periventricular white matter and progressive thinning of the corpus
and the rostrum and splenium may be absent. A markedly diminished callosum (splenium and rostrum are proportional to body, Fig. 3-19)
volume of white matter is present in the cerebral hemispheres; the little (238,240,241).
white matter that is present shows central T2 prolongation (Fig. 3-18B)
with sparing of the subcortical U fibers. In older patients, cerebellar or, Sjögren-Larsson Syndrome
less commonly, cerebral atrophy may appear (Fig. 3-18B) (230). Proton Sjögren-Larsson syndrome (OMIM 270200) is an autosomal recessive
MR spectroscopy shows reduced NAA throughout most of the brain disorder characterized by congenital ichthyosis, mental retardation,
with no difference between younger and older patients, implying a and progressive spastic tetraplegia (242). The severity of the disorder
static developmental encephalopathy associated with diffuse neuronal varies markedly even within families (243). The cause of the disorder is
or axonal abnormalities (231). impaired fatty aldehyde dehydrogenase (FALDH) activity (this enzyme
catalyzes the oxidation of fatty aldehydes to their corresponding fatty
Autosomal Recessive Spastic Paraplegia acids), which results in the accumulation of long chain fatty alco-
with Thin Corpus Callosum hols (244). The gene (ALDH3A2) has been mapped to chromosome
Hereditary spastic paraplegias are neurodegenerative conditions 17p11.2 (244), but mutations of many different aldehyde dehydroge-
in which the main clinical features are progressive spasticity and nase genes can cause the syndrome (44). Diagnosis is established either
weakness of the lower limbs associated with posterior column or by finding the mutation or by finding deficient fatty alcohol-NAD+
bladder involvement (232,233). This is a heterogeneous group of dis- oxidoreductase activity in leukocytes and cultured fibroblasts. Prenatal
orders, with autosomal dominant, autosomal recessive, and X-linked diagnosis is possible by assessing enzymes in cultured amniocytes and
inheritance. Autosomal dominant forms typically present in young fetal skin (155).

Barkovich_Chap03.indd 112 5/6/2011 12:08:13 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 113

FIG. 3-19. Hereditary spastic paraplegia in an 8 year old. A. Midline sagittal T1-weighted image shows thin corpus callosum, particularly the anterior body
(white arrows), while the rostrum and splenium are relatively spared; contrast with Figure 3-18A. B. Axial FLAIR image shows markedly reduced volume of
white matter with abnormal hyperintensity of periventricular and deep white matter.

Pathologic examination of affected brains shows ballooning of matter remain incompletely myelinated for many years, even into
myelin sheaths, with lipid-laden macrophages and histiocytes in the adulthood (246). Mild cerebral atrophy is found in most patients older
areas of myelin degradation. Myelin loss is seen primarily in the deep than 10 years (246). DTI shows normal diffusivity values but reduced
white matter of the cerebrum and in the corticospinal tracts in the FA in the white matter (248), likely representing impaired formation or
brainstem and spinal cord (245). impaired maintenance of myelin.
Imaging shows delayed myelination during the first years of life, Proton MRS of the occipital white matter shows a fairly charac-
with T2/FLAIR hyperintensity (Fig. 3-20A) (246). Later MR studies teristic, narrow peaks at 0.9 and 1.3 ppm (the peak at 1.3 ppm is the
show T2 prolongation in the periventricular white matter, particularly larger and more characteristic of the two) that are seen even at longer
in the regions around the trigones and frontal horns, without a great echo times (of 288 ms, Fig. 3-20B) (247) and, therefore, can be differ-
deal of volume loss (247). Of note, small areas of subcortical white entiated from the broader “macromolecular” peaks (probably methyl

FIG. 3-20. Infant with Sjögren-Larsson syndrome. A. Axial FLAIR image shows diffuse white matter hyperintensity. No volume loss is seen at this stage.
B. Proton MRS with TE = 288 ms from frontal white matter shows normal ratios of choline (Ch), creatine (Cr), and NAA. An abnormal peak is seen at 1.3
ppm that is characteristic of the disease and is therefore labeled SL for Sjögren-Larsson. The presence of the peak with TE = 288 ms and the narrowness of
the peak differentiate it from the macromolecular peak often seen at 1.3 ppm. Lactate would be a doublet with this technique. The chemical source of the
peak is not known.

Barkovich_Chap03.indd 113 5/6/2011 12:08:14 AM


114 Pediatric Neuroimaging

and methylene proton resonances, respectively) with those chemical actually be fatal. These data need to be taken into account prior to
shifts that are often seen in infants (246,247,249). These peaks are planning therapeutic strategies in such “high-risk” patients
smaller in other regions of the brain and are not seen in gray mat-
ter (246). It has been postulated that these peaks are the result of White Matter Injury With a few exceptions, the clinical and imag-
accumulation of long chain fatty alcohols (247); the area under these ing manifestations of white matter injury secondary to radiation or
curves, particularly the one at 1.3 ppm, diminishes as the patients chemotherapy in children are not significantly different from those in
mature into adulthood, suggesting unknown pathogenic mechanisms adults. Radiation injury to the brain is most commonly divided into
to compensate for the responsible biochemical defect in this disease three major groups: (a) acute reactions (1–6 weeks after treatment),
(243). Choline, creatine, and myo-I peaks are slightly elevated in the (b) early delayed reactions (3 weeks to several months after treatment),
white matter (246). and (c) late delayed reaction (several months to years following treat-
ment) (256–258).
Brain Injury from Radiation and Chemotherapy Acute and early delayed reactions are mild, often asymptomatic,
The pathophysiology of brain injury from radiation and chemother- and self-limiting, consisting of mild localized interstitial edema (sec-
apy is not fully understood. Recent information suggests that damage ondary to transient vasodilatation with variable changes in capillary
to vascular endothelial cells is an important component of radiation permeability (259) ) or perhaps transient myelin injury due to radia-
effects and that the damage is largely ischemic (250,251); however, an tion effects on oligodendrocytes (7). They are seen on imaging studies
inflammatory component has been suggested (252). Variables relevant as subtle low density (CT) or T1 and T2 prolongation (MR) without
to damage potential include total radiation dose, size of radiation field, significant mass effect or change in contrast enhancement (256,260).
size of radiation fractions, and the number and frequency of radia- DTI studies show that Dav (average diffusivity) is increased and FA
tion doses. Other factors include duration of survival, age of patient at (fractional anisotropy) decreased in T2 hyperintense white matter but
treatment, the specific chemotherapeutic agent, and whether therapy is FA was higher in adjacent normal appearing white matter (261). On
single (radiation or chemotherapy) or combined (radiation and che- proton MRS, a decrease in the size of the NAA and Cho peaks is noted,
motherapy) (252,253). even in normal-appearing white matter, for at least 6 months after
New data is emerging regarding unusual sensitivities to radia- radiation treatment (262).
tion or chemotherapies in certain individuals. It has been known for Late delayed injury is believed to result primarily from permanent
some time that syndromes caused by impaired DNA damage repair damage to blood vessels, perhaps associated with inflammation (7). It
(e.g., Fanconi anemia, ataxia-telangiectasia, etc.) predispose to child- may be seen as early as 3 to 4 months or as late as several years after
hood brain tumors, but recent reports suggest that affected patients conclusion of therapy. Breakdown of the capillary endothelium leads
are at increased risk for treatment-related toxicities, as well (254). For to blood–brain barrier breakdown and exudation of fibrin from the
example, patients with dual mutation in breast cancer susceptibility blood vessel lumen. Eventually, endothelium becomes hyalinized and
gene (BRCA2) may develop symptomatic neurotoxicity (leukoenceph- proliferates, compromising the lumen of the vessel and decreasing local
alopathy), myelosuppression, and poor immune response after “regu- blood flow, with subsequent white matter infarction (258,259). All of
lar” therapeutic doses of chemotherapeutic agents (255). Patients with these effects seem to be exacerbated when the patient also receives
other genetically determined cancer syndromes (ataxia-telangiectasia chemotherapy, particularly methotrexate (7). Pathologically, the white
or Gorlin syndrome) show excessive sensitivity to ionizing radiation matter exhibits areas of necrosis, with rarefaction and fragmentation of
(254). If such patients develop brain tumors and require radiation myelin, and cellular disruption. Patients may develop localized neuro-
therapy, conventional radiation doses may not be tolerated and could logic deficits or obtundation.

FIG. 3-21. Localized radiation necrosis in a child treated for a frontal lobe glioma. A. Axial FLAIR image shows marked edema in the frontal lobe extending
into the internal and external capsules and basal ganglia and causing right to left subfalcine herniation. B. Axial postcontrast T1-weighted image shows rim
enhancement (black arrows) in the right frontal lobe, suggesting recurrent tumor. C. MR susceptibility weighted blood volume map shows low blood volume
(blue color, black arrows) in the location of the rim enhancement, strongly suggesting radiation necrosis, not tumor recurrence.

Barkovich_Chap03.indd 114 5/6/2011 12:08:15 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 115

FIG. 3-22. Diffuse white matter injury from radiation and chemotherapy. A. Axial FLAIR image shows hyperintense white matter in the parietal and, to a
lesser extent, frontal white matter. B. Follow-up axial FLAIR image several months later show increased white matter injury and enlargement of ventricles
and subarachnoid spaces, suggesting progression of leukoencephalopathy and volume loss. C. Axial diffusivity (Dav) image at the time of the second scan
shows reduced Dav (white arrows) in both parietal and frontal white matter, confirming tissue injury. Fractional anisotropy may also be reduced due to
radiation/chemotherapy injury.

Imaging studies show variable patterns of injury, ranging from (266). Affected children may present up to 19 years after therapy (mean
single, focal lesions (Fig. 3-21) to diffuse white matter abnormality 8.1 years, earliest 2–3 years) (267,268). Presenting symptoms may be
(Fig. 3-22). Signal abnormalities reflect the edema and loss of myelin, headaches, seizures, or focal neurologic signs and symptoms; others are
as CT shows low attenuation and MRI shows T1 hypointensity and asymptomatic and found incidentally (263,266,267). The lesions may
T2/FLAIR hyperintensity (Figs. 3-21 and 3-22) (256,260). If tissue is develop after a wide range of radiation doses; in the series of Young-
injured, diffusivity may be reduced (Fig. 3-22). Contrast enhancement Poussaint et al., the range was 1,800 to 6,000 cG (267). Although there
is variable and may be transitory. Significant mass effect can be pres- was no clear correlation between dose and development of these
ent. Central necrosis within the lesions is uncommon in children (256). lesions in the series of Koike et al. (268), Chan et al. found a corre-
Petechial hemorrhage may be present, appearing as multifocal areas of lation between the number of hemorrhages and the radiation dose
short T1 and T2 superimposed upon the edema. Hemorrhage is much (263). Pathologic examination shows locally abnormal blood vessels.
more common following radiation therapy and is uncommon if treat- Currently, the lesions are only resected if they are symptomatic or have
ment is limited to chemotherapy (263). Knowledge of radiation ports caused symptomatic hemorrhage (269,270).
or the location of interstitial implants (if used) can be extremely useful
in making the proper diagnosis, as the injury is almost always limited Occlusive Vasculopathy Secondary to Radiation Young children
to the irradiated field. are more susceptible than adults to radiation injury of the large ves-
Preliminary work suggests that diffusion tensor imaging may allow sels of the circle of Willis. Children at greatest risk are those younger
detection and quantification of treatment-induced white matter injury than 4 years old who receive large doses of radiation to the sellar/supra-
(264). Compared with age-matched controls, reduction of the frac- sellar/parasellar region for supratentorial midline tumors (271–274)
tional anisotropy by 15% to 20% (see Chapter 1) can be detected in (patients with neurofibromatosis type 1 seem particularly susceptible
children treated with chemotherapy and radiation therapy even in the (275,276) ), but vasculopathy can develop after whole brain irradiation
setting of normal-appearing white matter. These changes were found for posterior fossa tumors such as medulloblastoma or ependymoma
to be most severe in children treated at younger age (<5 years old), (277) or for CNS involvement by leukemia (274). Rarely, the vasculop-
children with longer interval since treatment (>5 years), and children athy can develop as early as 15 months after radiation (277), although
with deteriorating school performance (264). Susceptibility-weighted it is more commonly seen after many years (273).
perfusion imaging shows reduced blood volume compared to unaf- Patients develop intimal hyperplasia and fibrous thickening
fected white matter (265); this is particularly useful in differentiation of the adventitia, leading to progressive narrowing of the supracli-
between radiation injury and recurrent tumor in children treated for noid carotid artery (Fig. 3-23) and the proximal anterior and middle
CNS malignancies. cerebral arteries; this may evolve into a moyamoya vascular pattern
(see Chapter 12). Clinically, this condition is manifested as growth
Hemorrhagic Vasculopathy Secondary to Radiation Hemor- retardation, cognitive impairment, transient ischemic attacks, frank
rhagic lesions may be seen in white matter of children who have been infarction, or developmental delay secondary to chronic cerebral
irradiated (263,266,267). The pathophysiology of these lesions has not ischemia (278). Imaging shows multiple small infarctions (Fig. 3-23),
been ascertained; capillary damage is a likely candidate. The imaging which may appear, as areas of T2 prolongation or areas of frank cystic
and surgical appearance are that of occult vascular malformations or necrosis.
cavernomas: sharply marginated lesions with little or no vasogenic
edema and variable signal intensity depending upon the nature of the Neoplasia Secondary to Irradiation Occasionally, neoplasia can
hemoglobin breakdown products within the lesions (see Chapter 12) develop as a long-term consequence of irradiation; this is felt to be

Barkovich_Chap03.indd 115 5/6/2011 12:08:16 AM


116 Pediatric Neuroimaging

FIG. 3-23. Radiation-induced vasculopathy in an 18-year-old girl with history of


radiation for her suprasellar tumor. A. Axial FLAIR image shows abnormal hyperin-
tensity in the basal ganglia, and external/extreme capsules. Note also pial hyperinten-
sity (white arrows) that is often associated with supraclinoid vasculopathy. B and C.
Axial FLAIR images show multiple small foci of periventricular T2 prolongation (black
arrows) as well as small cysts (white arrows). D and E. Postcontrast T1-weighted images
show curvilinear enhancement (small black arrows in E) representing dilated lenticu-
lostriate arteries in the basal ganglia. Solid white arrows in (D) point to small cysts.
Solid white arrows in (E) point to residual craniopharyngioma. Small black arrow in
(D) points to radiation-induced meningioma. F. Arterial phase image from left internal
carotid arteriogram shows occlusion of supraclinoid left internal carotid artery with
moya-moya-like collaterals (black arrows).

Barkovich_Chap03.indd 116 5/6/2011 12:08:17 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 117

a rare occurrence and it is difficult to prove causation (260). Of radi- studies, this is most easily appreciated in the skull base and spine (see
ation-induced tumors in adults reported in the literature, 70% are Chapter 2). Actively dividing cells are more susceptible to damage by
meningiomas (Fig. 3-23D), 20% are gliomas, and 10% are sarcomas radiation than are nondividing cells. Therefore, bone marrow of chil-
(279). However, malignant gliomas seem to be much more common dren is transiently damaged by irradiation, and hematopoietic cells
than meningiomas in children, and a significant incidence of sarcomas are replaced by fat cells. This phenomenon is seen on MR as homoge-
and low-grade gliomas have been noted (280). Features associated with neous T1 shortening in the skull base and vertebral bodies (from fatty
radiation-induced tumors include high histologic grade of gliomas replacement of marrow) that develops as early as 6 weeks after irradia-
in young people, multiplicity of gliomas, earlier age at presentation, tion (286). Normal marrow signal will return in slightly more than half
and higher doses of radiation (280). The neuroimaging appearance of of patients irradiated with 16 to 36 Gy (286), and in one series, return
radiation-induced tumors does not differ from that of those not asso- of singal was reported in 90% (287). No recovery is seen in patients
ciated with radiation. The neuroimaging appearance of brain tumors treated with 50 Gy or more (288). Marrow regeneration occurs in a
is discussed in Chapter 7. “band” pattern, with the more vascularized endplates recovering first.
MRI shows the development of bands of peripheral low signal inten-
Small (“Lacunar”) Cysts Small asymptomatic cysts develop in the sity on T1-weighted images of the vertebral bodies, slowly converging
white matter of approximately 15% of children who have been irradi- toward the center of the body. This signal change has been confirmed
ated prior to the age of 6 years with doses in the range from approxi- histologically as repopulation by hematopoietic cells (289).
mately 36 to 54 Gy; all patients received chemotherapy, as well (281,282).
The cysts appear after a typical latency period of 2 to 3 years and may White Matter Injury Secondary to Chemotherapeutic
grow on subsequent scans. These cysts (Fig. 3-23B and D) resemble Agents Abnormalities of the cerebral white matter can also result
large perivascular spaces, as they are isointense to CSF on all imag- from treatment with chemotherapeutic agents (290). The drugs
ing sequences and do not enhance after administration of intravenous that most commonly cause leukoencephalopathy are temozolomide,
contrast. They may result from localized brain atrophy or disturbance methotrexate, cisplatin, 5-fluorouracil, BCNU, melphalan, fludara-
in the CSF dynamics of interstitial fluid draining into the perivascu- bine, cytarabine, levamisole, arabinosylcytosine, carmustine, aspara-
lar spaces. It has not been established whether these may develop into ginase, and thiotepa (7,290,291), with methotrexate being the most
tumefactive cysts (see next section). common and best described in children, usually those with acute
lymphocytic leukemia (292–294). Temozolomide injury is more com-
Tumefactive Cysts Sometimes, slowly growing cysts develop within mon after treatment for malignant gliomas (295). The mechanisms
the area of the radiation ports. The time at which these cysts develop are not well understood but may include direct toxic effects (on axons,
is unknown, but affected patients typically become symptomatic 2 to oligodendrocytes, and progenitor cells) as well as secondary immu-
8 years after therapy. Headache is probably the most common symp- nologic reactions, oxidative stress, and microvascular injury (7). The
tom, with other symptoms being dependent upon the location of the white matter abnormalities seen on imaging studies may be transient
cyst and the structures that are affected. Tumefactive cysts seem to be or permanent and may be present with or without associated clinical
most commonly found after radiosurgery for arteriovenous malfor- abnormalities (291,296,297). Temozolomide seems to cause necrosis
mations (283), but we have seen similar cyst formation secondary to adjacent to tumor, which is thought to be due to endothelial cell dam-
high-dose conventional radiation therapy, as well (fortunately, radia- age with consequent increased vascular permeability and some paren-
tion doses for children are much lower than in the past and these are chymal necrosis (295).
very uncommon today). The incidence seems to increase with increas- Acute injury is seen in 3% to 10% of patients treated with intrath-
ing time after therapy and may be as high as 25% to 30%. The patho- ecal methotrexate (292,298), with higher doses putting the affected
genesis is unknown. patients at higher risk for leukoencephalopathy (299). Affected patients
On imaging, the cysts are typically complex and multilocular; typically present with neurologic symptoms such as aphasia, weak-
often, one cyst is dominant and surrounded by multiple smaller ness, sensory deficits, or seizures (292,293,300). In these situations, the
cysts. The cysts may have signal intensity of CSF but often are slightly imaging findings may be quite subtle on standard sequences such as
hyperintense compared to CSF on T1-weighted images, suggesting spin echo T2 and FLAIR (Fig. 3-24). Diffusion imaging shows reduced
the presence of proteinaceous fluid. This may be useful in differen- diffusion in the affected area and may be useful in the recognition of
tiating the cyst from a tumor cyst. The wall of the cyst is thin and the abnormality (Fig. 3-24C). Typical findings are round to ovoid focal
similar in signal intensity to white matter; it enhances moderately areas of reduced diffusion in the central areas of the centrum semiovale
after intravenous administration of contrast material (283). Adjacent (deep to the central sulcus) that spare the subcortical U fibers (293,294).
tissue is typically heterogeneous, showing areas of T2 prolongation Between 8 and 20 weeks after intrathecal methotrexate treatment, pro-
that are probably the result of both surgery and radiation; heteroge- ton spectroscopy shows transient reductions in NAA, increases in cho-
neous nodular enhancement is often seen in these areas after contrast line, and transient lactate peaks (Fig. 3-24D) (301). According to Chu
administration (283). The main differential diagnosis is radiation- et al. (301), these metabolic abnormalities disappear by about 1 year
induced glioma. after treatment.
In contradistinction to the focal white matter abnormalities
Effects of Irradiation on the Pituitary Gland It is known that seen in radiation-induced leukoencephalopathy and in acute che-
hypothalamic-pituitary function may be affected by irradiation of motherapeutic injury, T1 and T2 prolongation secondary to chronic
the sella and parasellar regions (284). This is manifest on MRI as chemotherapy injury tends to be symmetric, widespread and, often,
reduced pituitary gland size (285). No precise correlations between diffuse (Figs. 3-22 and 3-25). The T2 prolongation is seen primarily
degree of size reduction and degree of dysfunction have been dem- in the central and periventricular white matter, with relative sparing
onstrated. of the subcortical U fibers (155,256,291,296,297); it is often more
severe in the prefrontal and parietal lobes, sparing the posterior fron-
Post Irradiation Changes of the Bone Marrow The bone mar- tal lobes (Figs. 3-22B and 3-25). Occasionally, foci of T2 shortening
row of young children is still actively hematopoietic. On neuroimaging (hypointensity on T2-weighted images) are seen (302). The corpus

Barkovich_Chap03.indd 117 5/6/2011 12:08:18 AM


118 Pediatric Neuroimaging

FIG. 3-24. Focal acute white matter injury from methotrexate. A. Axial T2-weighted image shows faint hyperintensity (white arrows) in the left hemi-
spheric white matter. B. Axial FLAIR shows the same left hemisphere abnormality (white arrows) and an additional small area of hyperintensity in the
right hemisphere (white arrowhead). C. Axial diffusivity (Dav) map shows reduced Dav that is more severe in the left hemispheric (white arrows) than right
hemispheric (white arrowheads) lesion. D. Single voxel proton MRS from the left hemispheric lesion shows reduced NAA (downward-pointing arrow) and
the presence of lactate (upward-pointing arrow).

callosum, anterior commissure, and hippocampal commissure are Combined Radiotherapy and Chemotherapy An important con-
most often spared (256,260). When enhancement occurs after con- cept is that the combination of chemotherapeutic agents and radio-
trast administration, it tends to be multifocal, usually deep within therapy often results in more severe injury than either chemotherapy
the centrum semiovale (256). The presence of foci of enhancement or radiotherapy alone (7,290,295,304). This combination may result
and of T2 shortening tends to be associated with development of dif- in severe edema, enhancement, and mass effect within the radiation
fuse necrotizing leukoencephalopathy (302) (see the section below). ports, typically developing 5 to 13 months after therapy (mean of 10
Proton MRS of early chemotherapy-induced white matter change is months) (290,304). Differentiation from recurrent tumor may be very
normal, suggesting that the T2 prolongation results from changes difficult, and is discussed in more detail in Chapter 7. In general, injury
in myelin, as opposed to axons (303). Hemorrhagic vasculopathy occurs later than recurring tumor, is often multiple, and is often located
is not seen in patients who have received chemotherapy without several centimeters away from the tumor resection site or even con-
radiation (263). tralaterally. The study by van Tassel et al. showed common locations

Barkovich_Chap03.indd 118 5/6/2011 12:08:18 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 119

Diffuse Necrotizing Leukoencephalopathy When patients who


have received radiation and chemotherapy begin to rapidly deterio-
rate clinically in conjunction with diffuse white matter injury, the
condition is called diffuse necrotizing leukoencephalopathy (305).
This form of leukoencephalopathy is differentiated from other forms
by the presence of more extensive areas of white matter necrosis. On
imaging, the brain shows necrosis with areas of T1 and T2 shorten-
ing (Fig. 3-27). Lesions range from small rounded foci to large con-
fluent zones that may consist of noninflammatory demyelination or,
more commonly, frank necrosis (7). Injury to the blood–brain barrier
results in marked enhancement. If the patient survives, the necrotic
areas shrink and calcify (Fig. 3-27) and the volume of white matter
diminishes (256). The appearance of the white matter on imaging
studies does not always correlate well with the severity of the clinical
syndrome.

White Matter Disorders with Dysmyelination Initially


Affecting Subcortical Cerebral White Matter
Megalencephalic Leukoencephalopathy
with Subcortical Cysts (MLC)
Megalencephalic leukoencephalopathy with subcortical cysts (MLC,
OMIM 604004), also called leukoencephalopathy with macrocephaly
and mild clinical course, is an autosomal recessive disorder with onset
in infancy. This disorder is remarkable for its relatively mild early neu-
rologic signs and symptoms in the setting of a very abnormal imaging
study. Patients may present during first year of life with macrocephaly
FIG. 3-25. Chronic methotrexate injury. Axial T2-weighted image shows
extensive white matter hyperintensity in both cerebral hemispheres. Note
or during the second year of life with delay in attaining developmental
the sparing of the subcortical white matter. milestones. A slow neurologic deterioration then ensues with gradu-
ally increasing spasticity, dysarthria, ataxia, and dementia. Head cir-
to be the corpus callosum and corticomedullary junctions (304). cumference stabilizes after the first year and then parallels the normal
Tumor was more often solitary and located immediately adjacent to curve. Most patients are wheelchair bound within 8 to 10 years (306),
the surgical site. As stated above, correlation with radiation ports is although some can walk short distances with support at age 12 to
critical to making the correct diagnosis. More chronically, the changes 13 years. Seizures, usually responsive to medication, develop in a
from combined radiotherapy and chemotherapy are widespread mul- minority of affected children (307). Slow cognitive deterioration is
tifocal or diffuse cortical injury and diffuse deep cerebral and cerebel- noted from mid-childhood in older patients. Diagnosis is based upon a
lar nuclear injury with calcification. Examination of the white matter combination of clinical and MR criteria.
shows impaired myelination, diffuse injury with punctate calcifica- About 80% of cases are caused by mutations of the MLC1 gene
tions, and volume loss (Fig. 3-26). at 22qtel; the gene encodes a plasma membrane protein of unknown

FIG. 3-26. Chronic injury from chemo/radiation therapy. A. Axial noncontrast CT scan shows calcification of the basal ganglia, deep frontal and occipital
cortex, and some volume loss in the left parietal lobe, where the subarachnoid spaces are enlarged. B. Parasagittal T1-weighted MR image shows better the
full extent of cortical calcification (white arrows). C. Axial T2-weighted image shows abnormal hyperintensity of white matter, indicating impaired myelina-
tion, enlargement of subarachnoid spaces, and focal cortical injury (white arrows) in the left parietal lobe.

Barkovich_Chap03.indd 119 5/6/2011 12:08:19 AM


120 Pediatric Neuroimaging

FIG. 3-27. Diffuse necrotizing leukoen-


cephalopathy. A. Noncontrast CT scan
soon after symptoms shows white matter
edema and multifocal cortical and subcor-
tical calcifications. B. Axial T2-weighted
image a few months later show damaged
white matter with areas of T2 hyper- and
hypointensity due to a combination of
edema, calcification, and necrosis. Mass
effect from the left hemisphere causes
midline shift. C and D. Pre- and post con-
trast T1-weighted images show marked
diffuse enhancement secondary to exten-
sive injury to the blood–brain barrier.

function that is mainly expressed in neurons and glia (308–310). How- cal white matter appears swollen, with enlargement of the gyri in
ever, 20% of patients, including a number of families, with a typical affected regions (Fig. 3-28C). A characteristic feature is the presence
MLC phenotype do not have MLC1 mutations, indicating genetic of subcortical cysts that initially develop in the anterior temporal lobes
heterogeneity of the disease. These 20% have been classified into two (Fig. 3-28B) and may eventually arise in the frontal and parietal lobes
groups, one with both clinical and MRI features identical to patients (Fig. 3-28D and E). The cerebellar white matter is less affected than the
with MLC1 mutations, and another with a less severe clinical course. cerebral white matter, but some degree of T2 prolongation is present
This second group has less pronounced white matter swelling, less in the cerebellar white matter of most affected children (Fig. 3-28B and
severe megalencephaly or normal head size, and less affected cerebellar C) (307). Mild cerebellar atrophy is usually present (306). Diffusion
white matter on initial MR scans. No subcortical cysts are found out- studies show increased diffusivity in the cerebral white matter (dark on
side of the anterior temporal lobes. diffusion-weighted images, bright on ADC maps) (313). Proton MRS
Pathology of MLC cases reveals a vacuolating myelinopathy in is remarkable for low NAA levels, manifest as decreased NAA/Cr ratios,
which the outer layers of the myelin sheaths are separated, but the although MRS performed early in the course of the disease may be
inner layers are unaffected; axons are spared, but are widely separated nearly normal (67).
from adjacent axons by the vacuoles (311,312). Group I phenotype of MLC without MLC1 mutations has imaging
Neuroimaging studies are remarkable for a nearly complete findings essentially identical to that of patients with MLC1 mutations.
absence of myelin in the subcortical white matter; some central white Group II has less subcortical swelling and better preserved cerebellar
matter is spared, particularly in the corpus callosum, internal cap- white matter on initial MR scans; subcortical cysts are limited to the
sules, brainstem, and the occipital lobes (Fig. 3-28A). The subcorti- anterior temporal lobes. On follow-up scans, MRI shows spectacular

Barkovich_Chap03.indd 120 5/6/2011 12:08:20 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 121

FIG. 3-28. Megalencephaly with leukoencephalopathy and cysts. A–C. 16-month-old girl
with megalencephaly and mild developmental delay. Axial T1-weighted image (A) shows
marked absence of hyperintense myelin in the cerebral hemispheres. Some myelination is
present in the corpus callosum (white arrows) and internal capsules (isointense to basal gan-
glia). Axial FLAIR image (B) shows cysts (white arrowheads) in the anterior temporal lobes.
Note that the cerebellar white matter (which should be dark compared to gray matter at this
age) is abnormally hyperintense (white arrows). Coronal T2-weighted image (C) shows dif-
fuse abnormal hyperintensity of the white matter with the exception of the corpus callosum
(small white arrows) and hippocampal commissure (large white arrows). Cerebellar white
matter (white arrowheads) is abnormally hyperintense. Note that the cerebral gyri appear
large and rather swollen; there is no atrophy. D and E. Megalencephaly with leukoencepha-
lopathy and cysts; more advanced imaging findings. Parasagittal T1-weighted image (D)
shows subcortical cysts in right parietal (white arrowhead) and anterior temporal (white
arrows) lobes. The parietal lesion is confirmed as a cyst by the marked hypointensity (white
arrows) on the axial FLAIR image (E).

Barkovich_Chap03.indd 121 5/6/2011 12:08:21 AM


122 Pediatric Neuroimaging

improvement over the course of a few years with progression of myeli- typical MLC without MLC1 mutations is uncertain at this time. Typi-
nation such that only a few minor signal abnormalities remain; subcor- cally, delayed motor development and subsequently delayed speech are
tical cysts in the temporal lobes regress and may disappear (personal detected in the first 18 months of life. Motor dysfunction and cogni-
communication, Dr. Marjo van der Knaap). tive delay nearly always ensue. Occasionally, nystagmus, athetoid hand
Differentiation of this disorder from Canavan disease in particular movements, or dystonia develops (314,315).
is made by clinical criteria (onset in first year for Canavan, second year MRI shows characteristic involvement of the subcortical supraten-
for MLC), lack of involvement of globus pallidus and thalamus (almost torial white matter and cystic lesions in the anteriormost part of the
always present in Canavan disease), the presence of subcortical cysts temporal lobes (Fig. 3-29). The cysts do not communicate with the
(absent in Canavan disease), proton MR spectroscopy (large NAA peak ventricular system, which is typically enlarged (314,315). In our expe-
in Canavan disease, small NAA in MLC), and the diffusion character- rience, peritrigonal white matter is also involved and this involvement
istics (reduced diffusivity in Canavan disease (11), increased diffusivity extends to the ventricular surface. Proton MRS is normal. The abnor-
in MLC (313) ). mal white matter shows increased diffusivity.

Cystic Leukoencephalopathy Without Megalencephaly Aicardi-Goutières Syndrome


This recently described disorder seems to show autosomal reces- Aicardi-Goutières syndrome (AGS) is a genetically heterogeneous
sive inheritance and has been found primarily in persons of central disorder that initially is manifested in infancy with irritability, poor
and eastern European descent (314). The genetic locus has not been feeding, progressive microcephaly, spasticity, and dystonic postur-
identified and the relationship of this disorder to those resulting in ing; profound psychomotor retardation ensues, often with death in

FIG. 3-29. Cystic leukoencephalopathy without megalencephaly. A. Parasagittal


T1-weighted image shows low signal intensity in the parietal white matter with
sparing of subcortical white matter (black arrows) and two cysts (white arrows)
anterior to the temporal horn of the lateral ventricle (white arrowhead shows
ventricular wall). B. Axial FLAIR image shows extensive hyperintensity of the
periventricular, deep, and subcortical white matter. C. Axial diffusivity (Dav) map
shows that the affected white matter has mostly increased Dav (hyperintensity).

Barkovich_Chap03.indd 122 5/6/2011 12:08:23 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 123

FIG. 3-30. Aicardi-Goutieres syn-


drome. A. Axial noncontrast CT scan
shows calcification of the basal gan-
glia (white arrows) with diffuse corti-
cal and white matter atrophy. B. Axial
T2-weighted image shows marked
ventriculomegaly and enlargement of
the subarachnoid spaces due to white
matter volume loss. The basal ganglia
show a few punctate hypointensities
(black arrows) secondary to calcifi-
cation. (This case courtesy Dr. Susan
Blaser, Toronto.)

early childhood (316). Findings of chronic CSF leukocytosis and basal pigmentary retinopathy, optic atrophy, cataracts, premature aging, and
ganglia calcifications on CT raise the possibility of congenital infection, progressive neurological dysfunction; joint contractures may develop.
but testing is negative. Recent studies have revealed that the disorder is It is caused by mutations in the ERCC8 gene encoding the group 8
caused by mutations leading to cellular and immunological responses excision-repair cross-complementing protein on chromosome 5q11
that mimic congenital infections (317,318). New evidence suggests that (325). Affected patients have impaired transcription-coupled repair
the nucleases defective in AGS are involved in removing endogenously of damaged nuclear and mitochondrial DNA, which inhibits resump-
produced nucleic acids and that failure of this removal results in activa- tion of replication and transcription (326). Affected patients typically
tion of the immune system (319). Currently, five different types of AGS present with psychomotor retardation between the age of 6 and 12
are described: AGS1 (OMIM#225750, an autosomal recessive disorder months. Both development and growth progressively fall farther and
caused by mutation of the TREX1 gene on chromosome 3p21), AGS2 farther from the norm (327,328). Patients are further characterized
(OMIM#610181, autosomal recessive caused by RNASEH2B on chro- by accentuated thoracic kyphosis, lumbar lordosis, and characteristic
mosome 13q14-21), AGS3 (OMIM#610329, autosomal recessive caused facies (160,327).
by RNASEH2C on chromosome 11q13.2, AGS4 (OMIM#606034, CT in Cockayne syndrome reveals brain calcification, most com-
autosomal recessive caused by RNASEH2A on 19p13.13), and AGS5 monly in the basal ganglia (Fig. 3-31A), cerebellar dentate nuclei,
(OMIM#610905, an autosomal dominant form caused by mutation of and subcortical white matter, as well as cerebral and cerebellar atro-
TREX1). These disorders were classified under the label of “pseudo- phy (160,329). MRI shows diffuse atrophy (involving both cere-
TORCH syndrome” in previous editions of this book. bral and cerebellar hemispheres) and T2 hyperintensity, initially in
The finding of multiple punctate basal ganglia or white matter cal- the periventricular white matter, basal ganglia, and cerebellar nuclei
cifications in the proper clinical setting should raise the possibility of (330,331). The subcortical U fibers can be involved early in the disease
AGS. The putamina are most commonly involved (Fig. 3-30A) but the but are more commonly affected only in later stages (45). Suscepti-
globi pallidi, thalami, deep and subcortical white matter and, some- bility-weighted, gradient-echo, and (sometimes) spin-echo or FLAIR
times, cerebellar dentate nuclei may also show punctate or globular (Fig. 3-31B) images will show hypointensity within calcified deep cere-
calcification (320); these may or may not be visible on MRI. The sub- bral and cerebellar nuclei (330).
cortical and deep white matter typically are abnormally hypodense on Many other disorders have radiologic findings of cerebral calci-
CT and show T1 hypointensity and T2/FLAIR hyperintensity on MRI fications and abnormal myelination. These include many mitochon-
(Fig. 3-30B); subcortical white matter is involved early in the course of drial disorders (332), GM1 gangliosidosis (333), biotinidase deficiency
the disease (321). Imaging studies also show ventriculomegaly, usually (334), pseudo-TORCH syndromes (322), AGS (320), tuberous sclerosis
progressive, secondary to progressively reduced volume of white mat- (see Chapter 6), congenital cytomegalovirus, congenital toxoplasmosis,
ter in the cerebral hemispheres (Fig. 3-30B) (321). In later stages of the congenital rubella, and congenital AIDS (discussed in Chapter 11), and
disease, white matter may have signal intensity similar to CSF, particu- many others. Some of these have a neuroimaging appearance similar
larly in the frontal lobes (321). The cerebellum and brainstem may be to those in Cockayne syndrome but without the characteristic clinical
small, (322–324) but the cerebellar white matter is often spared. It is findings. References for the other disorders are provided for interested
important to remember that some patients with imaging findings that readers (316,335–337).
are suggestive of TORCH infections have genetic/metabolic disorders.
Therefore, if TORCH screens are negative in such patients and, espe- Galactosemia
cially if there is a family history of neurodegeneration in childhood, Galactosemia (OMIM 230400) is an autosomal recessive disorder that
testing for AGS should be performed. results from defective conversion of galactose to glucose by a path-
way catalyzed by several enzymes. It is most commonly the result of
Cockayne Syndrome galactose-1-phosphate-uridyl transferase deficiency due to mutation
Cockayne syndrome (OMIM 216400) is an autosomal recessive dis- of the GALT gene at chromosome 9p13, although other causes have
order that is characterized by cutaneous photosensitivity, dwarfism, been reported (338). Affected patients present as newborns and young

Barkovich_Chap03.indd 123 5/6/2011 12:08:23 AM


124 Pediatric Neuroimaging

FIG. 3-31. Cockayne syndrome. A. Axial non-


contrast CT scan shows calcification of the basal
ganglia (white arrows) and low attenuation of
the white matter. The ventricles and sulci are
enlarged, compatible in this situation with
atrophy. B. Axial FLAIR image shows multiple
punctate areas of signal void (white arrows) in
the basal ganglia, which represent punctate cal-
cifications. Diffuse hyperintensity of the white
matter is present (black arrows).

infants with signs of increased intracranial pressure and vomiting. Proton MR spectroscopy of the brain in patients with galactosemia
If untreated, patients develop severe liver disease, profound mental is reported to be normal by some authors (342), but others report reso-
retardation, epilepsy, and choreoathetosis (339). Treatment is dietary nances at 3.67 and 3.74 ppm, corresponding to galactitol, in newborns
restriction of galactose; however, even compliant patients show some with very high urinary galactitol levels (343). These peaks are not spe-
neurologic and neuropsychologic abnormalities (340). cific for galactitol, as peaks with similar chemical shifts are present in
CT scans of patients with galactosemia are nonspecific, showing many sugars, and can represent glucose in diabetes mellitus, or arabitol
extensive low attenuation in the cerebral white matter (59). MR stud- and ribitol in errors of polyol metabolism (344). Galactitol can be dis-
ies seem somewhat more specific, showing delayed myelination (per- tinguished from myo-I, which precesses at nearly the same chemical
sistent high signal) in the subcortical white matter and consequent shift, but in the presence of J-coupling, which results in inversion of the
blurring of the cortical–white matter junction on T2-weighted images peak when using an echo time of 135 to 144 ms. Galactosemic patients
(Fig. 3-32), although the same white matter seems to mature normally who have been following a galactose restricted diet for several years do
on T1-weighted images (341,342). Cerebral and cerebellar atrophy is not have galactitol peaks (343).
seen in older patients. Focal white matter lesions are occasionally seen
and are of uncertain significance (341). Mitochondrial Disorders
Although mitochondrial encephalopathies can cause demyelination,
gray matter is almost always involved. Therefore, mitochondrial dis-
orders are discussed in the section “Metabolic diseases that affect gray
matter and white matter.”

White Matter Disorders Due to Hypomyelination


(Hypomyelinating Leukodystrophies)
A group of disorders have been described in recent years, in which
the primary white matter disorder is one of hypomyelination or,
in some cases, delayed myelination of the white matter; these have
become known as hypomyelinating leukoencephalopathies (345).
They are included as a separate group because the MR characteristics
of the white matter differ from that of myelin breakdown (demyeli-
nation), myelin vacuolization, or white matter breakdown (10). The
reduction of myelin is seen as persistent T2 hyperintensity, similar to
that normally seen in neonates and young infants. However, signal
intensity of hypomyelination may be nearly normal on T1-weighted
images, fractional anisotropy is only mildly reduced, and diffusivity
is only mildly increased compared to normal age-matched brains,
proton MRS is nearly identical, while magnetization transfer ratios
are low, similar to those of demyelination and myelin vacuolization
(10). Understanding these disorders will increase our knowledge of
FIG. 3-32. Galactosemia in a 9-year-old girl. Axial T2-weighted image
show delayed myelination in the subcortical white matter with consequent the process of myelination and, perhaps, help intervene and stimu-
blurring of the cortical-white matter junction. Note the poor differentia- late myelination in affected patients. So far, these disorders include
tion of the cortex from the underlying white matter. (This image courtesy Pelizaeus-Merzbacher disease (346), Pelizaeus-Merzbacher–like
Dr. Susan Blaser, Toronto.) disease (caused by GJA12 and MCT8 mutations and a mutation on

Barkovich_Chap03.indd 124 5/6/2011 12:08:24 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 125

Xq (28,29,347,348) ), Cockayne syndrome type II (349), sialuria more copies of the PLP1 gene, typically have a more severe form of
(Salla disease) (350), Tay syndrome (351), 18q- syndrome (30,31), the disease; misfolded proteins accumulate in the endoplasmic reticu-
leukodystrophy with trichothiodystrophy and photosensitivity lum and provoke an unfolded protein response, leading to apoptosis
(352,353), congenital cataracts with hypomyelination (36), hypomy- (360,365–367). In connatal Pelizaeus-Merzbacher disease, both PLP
elination with atrophy of the basal ganglia and cerebellum (34), and and DM20 proteins become trapped in the endoplasmic reticulum;
the very recently described cerebral hypomyelination with cerebellar thus, myelin is more severely affected (368). Another form of Pelizaeus-
atrophy and callosal hypoplasia (354). Merzbacher disease, resulting from a deletion of 19 base pairs in intron
3 of the PLP1 gene, has been described in which mild early develop-
Pelizaeus-Merzbacher Disease mental delay is followed by progressive decline of acquired motor and
Pelizaeus-Merzbacher disease (OMIM #312080) (346) is a hypomyeli- cognitive milestones (369). Still another disorder, known as spastic
nating disorder caused by mutations of the PLP1 gene at chromosome paraplegia type 2 (SPG2, OMIM #312920), is caused by null mutations
Xq22 (355,356), resulting in abnormal formation of proteolipid pro- of the PLP1 gene (360). Patients with SPG2 show normal motor devel-
tein (PLP) and its isoform DM20, two of the major structural proteins opment in the first year of life and then develop progressive weakness
of myelin (356–360). The classic form of Pelizaeus-Merzbacher disease and spasticity of the lower limbs between the age of 2 and 10 years
shows X-linked recessive inheritance. Patients present in early life with (360). There may be symptoms that overlap with PMD, such as nystag-
nystagmoid eye movements and slow motor development; they sub- mus, optic atrophy, ataxia, dysarthria, or impaired cognition, but they
sequently develop involuntary movements and spasticity. The disease are less prominent than in PMD (360).
course is protracted and can easily be misdiagnosed as static, leading to On CT scans, Pelizaeus-Merzbacher disease appears as low atten-
a diagnosis of “cerebral palsy.” Death usually ensues in or early to mid- uation in the white matter with progressive white matter atrophy; it
adulthood. Patients with the connatal form of Pelizaeus-Merzbacher is therefore indistinguishable from most other white matter diseases
disease show either autosomal or X-linked recessive inheritance; thus, (370). The MR appearance of this disorder is one of a lack of myelina-
girls as well as boys may be affected (361,362). Affected patients pres- tion, without frank evidence of white matter destruction (27,46). In the
ent at birth or in early infancy; in addition to abnormal nystagmoid connatal form of the disease (Fig. 3-33), myelin is completely absent
eye movements and extrapyramidal hyperkinesia, affected patients and never develops. In the classic form, the brain typically retains the
develop spasticity, stridor, progressive contractures of the extremities, appearance of a newborn, with high signal intensity only appearing
optic atrophy, and seizures in the early stage. Progression is relatively in the internal capsule, optic radiations, and proximal corona radi-
rapid, with death occurring in early childhood; pathologic exam shows ata on T1-weighted images and a near complete absence of low sig-
lack of myelin in the entire brain (360,363). nal intensity in the supratentorial region on the T2-weighted images
Most patients (60%–70%) with PMD have increased PLP1 due (Fig. 3-34). The amount of myelination and the volume of white matter
to duplication of the genomic fragments containing the entire PLP1 slowly diminish over time (27). The cerebellum may become markedly
gene (355). Patients with duplication of the gene typically have the atrophic (11) and cortical sulci show slowly progressive enlargement
classic form of PMD (360,364). In these children, PLP is postulated to (371,372). No relationship has been demonstrated between the degree
accumulate in the endoplasmic reticulum and lysosomes while DM20 of myelination as determined by MR and the progression of the disease
makes it to the cell surface; therefore, myelin molecules can be partly or the clinical severity of the disease (373), other than the profound
formed. Other patients, with PLP1 point mutations or with three or lack of myelin in the connatal form (366,374).

FIG. 3-33. Pelizaeus-Merzbacher disease, connatal form in a 2 month old. Axial T1 (A) and T2 (B)-weighted images show a complete absence of evidence
of myelination. In a normal infant of this age, one would expect to see T1 hyperintensity and T2 hypointensity in the posterior limb of both internal cap-
sules (see Chapter 2).

Barkovich_Chap03.indd 125 5/6/2011 12:08:24 AM


126 Pediatric Neuroimaging

FIG. 3-34. Pelizaeus-Merzbacher disease, classic form


in a 9-year-old. A. Axial T1-weighted image shows
some hyperintensity in the corpus callosum, internal
capsules, and optic radiations. However, a normal
9-year-old should have hyperintense white matter
throughout the brain. This is very delayed myelination.
B. Axial T2-weighted image shows profoundly delayed
myelination; all white matter should be hypointense
compared to gray matter at this age. C. Proton MRS
(TE = 144 ms) from the frontal white matter shows
increased NAA and creatine (Cr) compared to choline
(Ch), a common finding in the classic form of PMD
due to duplication of PLP1.

Reports of proton MRS are controversial; this is probably a result oligodendrocytes (375). Studies of patients with point mutations of
of the fact that many of the reported patients have different mutations the PLP1 genes suggest that NAA is decreased, while Cho and Cr are
of the PLP1 gene or no mutations of the PLP1 gene (and therefore have increased, using long echo proton MRS (376).
PMD-like disease) and, thus, the reported patients are heterogeneous.
However, both Takanashi et al. (365) and Hanefeld et al. (375) have Pelizaeus-Merzbacher–like Disease
recently studied patients with documented duplications of the PLP1 Some patients have been described with clinical presentation identi-
gene. Both groups have reported that NAA, Cr, Glu, and myo-I are cal to that in patients with Pelizaeus-Merzbacher disease, but without
increased compared with age-matched controls (Fig. 3-34C) in white PLP1 mutations. These patients appear to have a different, Pelizaeus-
matter voxels, while Hanefeld et al. have noted that Ch is reduced; spec- Merzbacher–like disease. An X-linked group has been reported with a
tra are normal in gray matter voxels (basal ganglia and cortex) (375). locus on Xq (347) and two autosomal recessive groups with mutations
The increase in NAA is postulated to reflect either increased number of of the GJA12 gene at 1q41-42 (29,348) and MCT8 at Xq13.2 (28).
axons per voxel (because of the lack of surrounding myelin sheaths) or GJA12 (OMIM 608804) encodes the gap junction connexin pro-
increased activity of oligodendrocyte precursors. The elevated Cr and tein (Cx46.6), which is highly expressed in oligodendrocytes and
myo-I are postulated to result from reactive astrogliosis. Reduced Ch appears to play a role in a myelinogenesis (377). MRI and MR spec-
is postulated to result from the severe reduction of choline contain- troscopy appear to be nearly identical to those in PMD due to PLP1
ing myelin proteins and lipids caused by lack of normally functioning deletions (348).

Barkovich_Chap03.indd 126 5/6/2011 12:08:25 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 127

MCT8 (OMIM 300095) encodes a thyroid hormone transporter fertility, and short stature) (352). Ventriculomegaly was present in one
gene that is implicated in X-linked mental retardation with a wide patient that we examined, possibly due to progressive atrophy of the
spectrum of clinical presentations. Mutations of this gene were found cerebrum and cerebellum (385). Osteosclerosis, when present, can be
in approximately 10% of a group of patients with clinical findings of identified by CT or MRI (381). One report described proton MRS find-
Pelizaeus-Merzbacher disease, but no mutations of PLP1 or GJA12 ings of reduced choline and elevated myo-I(386).
(28). This disorder is sometimes referred to as monocarboxylate trans-
porter 8 deficiency (378). Most affected patients have early-onset hypo- 18q- Syndrome and Other Chromosome 18 Mutations
tonia, nystagmus, ataxia, and choreoathetosis. Subsequently, spasticity The chromosome 18 q deletion (18q-) syndrome (OMIM 601808) is
and generalized dystonia develop; motor and cognitive acquisitions a recently described disorder caused by a deletion of the long arm of
are delayed or absent (28). MRI shows essentially no myelination for chromosome 18 (30). Patients show a wide range of phenotypic varia-
the first year of life (similar to connatal Pelizaeus-Merzbacher disease, tion, probably related to the extent and location of the deleted portion
Fig. 3-33), but, unlike patients with PLP1 or GJA12 mutations, myelin of the chromosome. Some relatively constant manifestations include
(T1 hyperintensity and T2 hypointensity of white matter) slowly mental retardation, developmental delay, short stature, impaired hear-
appears on MR scans after age 2 years. However, the volume of white ing from aural atresia, craniofacial dysmorphism (midface hypoplasia,
matter and the thickness of the corpus callosum remain diminished frontal bossing, carp-like mouth), limb anomalies, oculomotor disor-
(28). Experiments in mouse models suggest that the abnormal brain ders, and genital hypoplasia. Most of these characteristics are found in
development is due to deficiency to T3 hormone in the brain (379). patients with deletion of the most distal part of the long arm of the
chromosome, 18q22.3-ter (31). Neurologic manifestations may be
Leukodystrophies with Trichothiodystrophy related to the fact that one of the myelin basic protein genes is located
Trichothiodystrophy (brittle, sulfur-deficient hair) is a marker for a num- on the long arm of chromosome 18 and is almost invariably included in
ber of autosomal recessive neurocutaneous syndromes with neurologic the deleted segment. Not surprisingly, other mutations of chromosome
manifestations and mental retardation. At least six syndromes with tri- 18 have also been shown to have myelination defects (387). However,
chothiodystrophy have been described (351,352). Most affected patients individuals with deletions sparing the distal area seem to be less severely
have ichthyosis, brittle sulfur-deficient hair, impaired intelligence, affected, without abnormalities in the extremities, oculomotor defi-
decreased fertility (in some cases), and short stature (352). Other features ciencies, or short stature; their cognitive problems are milder (31).
include nail and teeth dysplasia, retinal dystrophy, cataracts, increased MRI studies of patients with 18q- syndrome show findings of
susceptibility to infections, endocrine dysfunction, cardiomyopathy, and delayed myelination, slower rate of myelination, and hypomyelination
osteosclerosis (353,380,381). Major neurological features are mental compared with age-matched controls (30,388). In most cases, poor
retardation, spasticity with pyramidal signs, hyperreflexia, spinocerebel- differentiation between gray matter and white matter in the cerebral
lar signs such as nystagmus, dysarthria, tremor, and ataxia, peripheral hemispheres is reported on T2-weighted images (Fig. 3-35) (389),
neuropathy, and neurosensory hearing impairment (351,382). probably due to abnormal myelin; this particular appearance appears
MR findings have been described in four syndromes with tri- to be associated with mutations in the 18q22.3-q23 region (31). Inter-
chothiodystrophy, and all seem to show diffuse hypomyelina- estingly, the T1-weighted images have normal appearance (Fig. 3-35A)
tion, very similar to Pelizaeus-Merzbacher syndrome (Fig. 3-34) (389) despite the fact that quantitative analyses show T1 prolongation
(351,353,380,381,383–385). The hypomyelination was confirmed by of white matter (388). Diffusion-weighted images show higher diffu-
autopsy in one patient with PIBI(D)S syndrome (photosensitivity, ich- sivity than age-matched controls (Fig. 3-35C). Small anterior lobes of
thyosis, brittle sulfur-deficient hair, impaired intelligence, decreased the pituitary gland have also been reported (390). Proton MRS at age

FIG. 3-35. Chromosome 18q deletion syndrome in a 7-year-old child. Axial T1-weighted image (A) appears normal but the T2-weighted image (B) and
diffusion weighted image (C) show indistinct separation of gray matter and white matter due to hypomyelination of the white matter. These findings are
characteristic of hypomyelination.

Barkovich_Chap03.indd 127 5/6/2011 12:08:26 AM


128 Pediatric Neuroimaging

FIG. 3-36. Sialuria (Salla disease). A. Sagittal T1-weighted image shows a markedly thin corpus callosum (white arrows), indicated markedly reduced white
matter volume. B and C. Axial T1 (B) and T2 (C)-weighted images show a small amount of T1 hyperintensity and T2 hypointensity in the posterior limbs
of the internal capsules (white arrows), suggesting the presence of some early myelination. (These images courtesy Dr. Susan Blaser, Toronto.)

30 months in a single patient revealed normal NAA, increased cho- MRS of patients with Salla disease shows elevation of the Cr and
line on a long TE sequence in the parietal white matter, and increased NAA peaks, diminution of the Ch peak in the white matter; spectra
a-glutamate and myo-I on a short TE spectrum in the parietal white in the basal ganglia show an enlarged Cr peak but normal NAA and
matter. Of interest, these values nearly normalized within a year (391). Ch peaks compared with controls (399). The reason for the increased
amplitude of the “NAA” peak appears to be that the acetyl protons from
Sialuria N-acetylneuraminic acid resonate at the same frequency as the acetyl
Sialuria, (OMIM 604369) also known as Salla disease, is an autosomal protons from N-acetyl aspartate (399).
recessive disorder caused by an error in sialic acid (N-acetylneuraminic
acid) metabolism (350); the genetic locus has been mapped to the Hypomyelination with Congenital Cataracts
SLC17A5 gene at 6q14-q15 (392). A defect in the mechanisms of sialic Hypomyelination with congenital cataract is a recently described dis-
acid transport across the lysosomal membrane causes free sialic acid order that is caused by deficiency of a membrane protein of unknown
to accumulate in the lysosomes of various tissues (393). There appear function called hyccin, which is encoded by the DRCTNNB1A gene
to be two forms of the disease, and infantile form and an adult form located at chromosome 7p15.3 (36). Affected patients are first recog-
(394); while the adult form is seen predominantly in people of Finn- nized at birth, or during the first month after birth, when they are dis-
ish descent (this is true Salla disease), the infantile form has no ethnic covered to have bilateral cataracts. Development is then normal until
prevalence (44). Sialic acid levels in the urine of affected infants are the end of the first year of life, when delayed motor development is
usually about 20 times higher than in the adult form (394,395); urine detected. Affected infants learn to “cruise” (walk with support) but
sialic acid levels may be normal in milder cases and require CSF analy- never walk without help and gradually become wheelchair-bound (35).
sis to detect sialic acid (396). Affected children typically become symp- Scoliosis slowly develops and progresses, along with hyperactive deep
tomatic between the age of 3 and 6 months, manifesting hypotonia, tendon reflexes, extensor plantar responses, dysarthria, cerebellar signs,
ataxia, and often nystagmus. Head size is normal. Subsequently, devel- and truncal ataxia (35). Cognition is impaired, with mild to moderate
opmental delay becomes apparent. The disorder is slowly progressive. impairment in all patients described to date (35).
Most learn to walk and speak a few words, only to regress beginning in No central nervous system pathology has been evaluated in these
the second and third decades (33). patients. However, sural nerve biopsy shows reduced or absent myelin
MR studies of patients with Salla disease show reduced volume as well as absence of compaction of the myelin sheaths of peripheral
of cerebral white matter and markedly diminished myelination. The nerves (35). Overall, the impression is that little myelin is actually
images resemble those of patients with Pelizaeus-Merzbacher disease. formed and that this is subsequently broken down such that the dis-
The corpus callosum is extremely thin and unmyelinated (Fig. 3-36A). ease is probably best considered a combination of hypomyelination
In less severe cases, the ventricles are of normal size and some myelin and demyelination (400).
is seen in the peritrigonal white matter and the posterior limbs of the Neuroimaging with MRI shows more severe white matter abnor-
internal capsules (Figs. 3-36B and C) (33). The subcortical white matter malities compared to other hypomyelinating disorders. On T1-weighted
never seems to myelinate. In patients with more severe symptomatol- images, cerebral and cerebellar white matter may be slightly hyperin-
ogy, enlarged ventricles and subarachnoid spaces are seen (33). In some tense, isointense, or slightly hypointense compared with gray matter;
patients, the cerebellar white matter is affected, as well (397,398). The white matter is hyperintense compared to gray matter on T2-weighted
basal ganglia appear normal (Fig. 3-36) (399). As the disorder is slowly images (35). Subcortical white matter may be partially spared early in
progressive, it is difficult to know whether the myelination abnormality the course of the disease (Fig. 3-37) (400). Some patients have more
and enlargement of the CSF spaces is progressive (33). severe T1 and T2 prolongation in the periventricular and deep white

Barkovich_Chap03.indd 128 5/6/2011 12:08:26 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 129

FIG. 3-37. Hypomyelination with congenital cataracts in a 3-year-old child. A. Parasagittal T1-weighted image shows abnormal hypointensity of the deep
white matter with normal hyperintensity (white arrows) of the subcortical white matter. B. Axial T2-weighted image shows abnormal hyperintensity of deep
white matter (white arrows) with sparing of the periventricular and subcortical white matter. Note lens replacements (black arrows) in both ocular globes.
C. Coronal T2-weighted image shows abnormal hyperintensity of deep cerebral (white arrows) and cerebellar (white arrowheads) white matter; note spar-
ing of corpus callosum and of subcortical white matter in both compartments. D. Axial diffusivity (Dav) map shows increased Dav in the affected cerebral
white matter.

matter, which may show hypointensity on FLAIR images, suggesting of the disease; metabolite quantification in a single patient showed
cavitation (400). These regions seem to start anteriorly and progress elevated Cr and myo-I, with reduced NAA (400).
posteriorly; they decrease in volume over time (35,400). There is no
enhancement after intravenous administration of paramagnetic con- Fucosidosis
trast (400). Cerebellar white matter is less severely affected than cere- Fucosidosis (OMIM #230000) is a rare autosomal recessive lysosomal
bral white matter. The pons is moderately hypoplastic, sometimes storage disease, resulting from a deficiency of alpha-L-fucosidase (401).
showing T2 or FLAIR hyperintensity in the corticospinal tracts and Affected patients variably present with progressive neurologic deterio-
transverse pontine fibers (400). ration, coarse facial features, and dysostosis multiplex (401). Abnormal
Diffusion-weighted imaging shows increased diffusivity hyperintensity is present in the white matter on T2-weighted and FLAIR
(Fig. 3-37D) in affected white matter, with slightly increased Dav values images of patients in late infancy and early childhood; this may be the only
in the unmyelinated white matter and higher values (by ∼20%) in the finding in late infancy (402). However, because cortical and basal ganglia
cavitating white matter (400). Proton MRS with short echo time may involvement is common, the more complete discussion is in a later section
show an increased myo-I/Cr ratio and Cho/Cr ratios early in the course of this chapter on disorders involving both gray and white matter.

Barkovich_Chap03.indd 129 5/6/2011 12:08:27 AM


130 Pediatric Neuroimaging

Hypomyelination with Atrophy of the Basal apnea and often death) (404,405). Those patients who recover sponta-
Ganglia and Cerebellum (HABC) neous respiration develop seizures, dystonia, and pronounced develop-
Hypomyelination with atrophy of the basal ganglia and cerebellum mental delay (404). Milder clinical presentations have been reported,
(OMIM 612438) is a hypomyelinating disorder that has involvement including a childhood form with mild mental retardation and episodes
of the basal ganglia and cerebellum as well as white matter. It is dis- of delirium, chorea, and vertical gaze palsy during febrile illness, and
cussed in the section of this chapter on Disorders that involve Gray and a late-onset form in which children present with progressive spastic
White Matter. diplegia and optic atrophy but preservation of intellectual function
(156,404,406). A transient neonatal form has also been described
White Matter Diseases with Nonspecific Patterns (407,408).
CT studies have revealed cerebral and cerebellar volume loss with
Nonketotic Hyperglycinemia (Glycine Encephalopathy) hypoattenuation of the periventricular white matter (409). Early MR
Nonketotic hyperglycinemia, also called glycine encephalopathy studies show severely delayed myelination and white matter edema
(OMIM #605899), is an autosomal recessive disorder of amino acid (Fig. 3-38A–D). Prenatal neurotoxicity may result in encephalomalacia
metabolism in which large quantities of glycine accumulate in plasma, with thinning of the corpus callosum and cerebral cortical anomalies.
urine, and CSF. It is caused by a disturbance in the breakdown of gly- Studies later in the course of the disease show a nonspecific pattern
cine, which may result from mutations of several different genes in the of decreased volume and T2 hyperintensity of the cerebral cortex and
mitochondrial glycine cleavage system; loci for these genes have been cerebral hemispheric white matter with a consequently small corpus
identified at 16q24, 9p22, 3p21, and 2p21.1 (44). In the classic form callosum (Fig. 3-38F and G) (410,411). A few studies have reported
of the disease, onset of clinical symptoms is in the neonatal period or reduced diffusion in the dorsal brainstem and corticospinal tracts
early infancy (70% in first 4 days of life (403) ) with myoclonic enceph- (posterior limbs of internal capsules, central corona radiata) on dif-
alopathy (lethargy, hypotonia, and myoclonic jerks, progressing to fusion-weighted imaging (Fig. 3-38B and D), probably reflecting the

FIG. 3-38. Nonketotic hyperg-


lycinemia in a neonate. A and B.
Axial T2-weighted and diffusion
weighted images show T2 hyperin-
tensity (A) and reduced diffusivity
(B) in the cerebellar white matter
(black arrows) and central tegmen-
tal tracts (black arrowheads). C and
D. Axial T2-weighted and diffusion
weighted images at the level of the
basal ganglia show diffuse white mat-
ter hyperintensity with reduced dif-
fusivity in the posterior limbs of the
internal capsules (black arrows), the
myelinated part of the white matter.
E. Proton MRS (TE = 144 ms) shows
abnormally elevated glycine peak
(Gly) at 3.6 ppm. Myo-Inositol also
precesses at this frequency but is not
seen at echo times this long.

Barkovich_Chap03.indd 130 5/6/2011 12:08:28 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 131

FIG. 3-38. (Continued) F and


G. Follow-up MR images at age
18 months. Sagittal T1-weighted
image (F) shows a very small
corpus callosum (white arrows),
resulting from severe loss of
cerebral white matter. Axial
T2-weighted image (G) shows
enlarged ventricles and suba-
rachnoid spaces with hypomy-
elinated white matter, indicative
of atrophy from severe cortical
and white matter injury.

known pathological finding of vacuolating myelinopathy in the acute system concentrations of uracil and/or thymine may be contributing
phase of the disorder; imaging at this stage can resemble that of maple factors (418).
syrup urine disease (section IV.B.1.g), which is also due to a vacuo- Reported brain imaging findings consist of partial agenesis of
lating myelinopathy. Anisotropy of the white matter may be normal the corpus callosum, intracerebral hamartoma, T2 prolongation of
(412,413). As atrophy develops over time, however, diffusion normal- the white matter secondary to markedly delayed myelination, slight
izes and then increases while anisotropy decreases, probably as a result white matter hyperintensity, “strong contrast between white and gray
of the known axonal degeneration (413). matter,” and diffuse cerebral atrophy (415). The rare patient with
Proton MRS studies are critical for making the diagnosis, showing an underlying DPD deficiency who undergoes cancer chemotherapy
abnormal peak at 3.56 ppm (Fig. 3-38E), believed to represent glycine with 5-fluorouracil may develop severe toxic side effects, including
(411,414). Abnormal spectra are reported in patients with normal MRI a multifocal inflammatory leukoencephalopathy with hyperintense
studies. Moreover, the clinical course of the patients seems to correlate white matter lesions on T2-weighted images (419). The T2-weighted
more closely with the cerebral glycine level, as detected by MRS, than axial images show abnormally prominent CSF spaces. Abnormal T2
to plasma or CSF concentrations of glycine (411,414). In order to sepa- prolongation is present in the periventricular cerebral white matter.
rate the glycine peak from that of myo-I, which has a similar chemical In addition, abnormal T2 prolongation may be seen in the brain-
shift, the spectra should be obtained with a long echo time (135-144 or stem, specifically in the pontomedullary portions of the medial lem-
270-288 ms). The myo-I protons with that chemical shift have a short nisci and median longitudinal fasciculi, and in the inferior olivary
T2 relaxation time and, therefore, disappear on the long echo time nuclei (420). The ventral superficial reticular area of the medulla
spectra. Therefore, MRS is the most important study for the diagnosis and the parvocellular reticular area of the pons may also be involved
and for following patients with nonketotic hyperglycinemia (411). (Fig. 3-39).

Dihydropyrimidine Dehydrogenase Deficiency 3-Hydroxy-3-Methylglutaryl-Coenzyme A Lyase Deficiency


Dihydropyrimidine dehydrogenase deficiency (DPD, OMIM 274270) HMG CoA lyase is an important compound in both the breakdown
is caused by mutation of the DPYD gene located at 1p22 (415); DPD of L-leucine and in fatty acid oxidation; the latter process is impor-
is the rate-limiting enzyme in the three-step pathway of uracil and tant in the production of acetyl CoA and acetoacetate, both of which
thymidine catabolism and in the pathway leading to the formation are important compounds in energy production within the brain.
of beta-alanine (44). It has been reported mainly in patients of Euro- In addition, excess HMG CoA inhibits the process of gluconeogen-
pean or Turkish ancestry. Affected children have had a variable clinical esis. Deficiency of HMG CoA lyase (OMIM 246450) is characterized
phenotype ranging from unaffected to severe neurologic involvement by recurrent episodes of severe metabolic derangement, including
(415,416). Because of the presence of unaffected individuals, including hypoglycemia, metabolic acidosis, and sometimes hyperammonemia
asymptomatic siblings, with biochemical and molecular findings simi- (421,422). It is caused by mutations of the HMGCL gene at chromo-
lar to affected patients, it appears that DPD deficiency is a necessary, some 1pter-p33 (423). Patients typically present with their first crisis
but not sufficient, prerequisite for the development of clinical abnor- as neonates or young infants. Diagnosis is suggested by abnormalities
malities (415,417). Seizures and mental and motor retardation are in urinary organic acids (3-hydroxyisovaleric, 3-methylglutaconic,
common features, affecting 45% of individuals (415). Ocular abnor- 3-methylglutaric acids and 3-hydroxy-3-methylglutaric acid) and con-
malities (23%), autism (18%), growth retardation (18%), microceph- firmed by demonstration of deficient activity of HMG CoA lyase in
aly (14%) and mild dysmorphic features (14%) are relatively frequent leukocytes and cultured fibroblasts (421,422).
findings (415). Ophthalmologic findings in DPD deficient patients CT of patients with HMG CoA lyase deficiency shows diffusely low
include megalocornea, optic atrophy, iris and choroidal colobomata, attenuation of the white matter (37). MRI usually reveals nonspecific
and fundus hypopigmentation (415). The mechanism of neurologic gray and white matter abnormalities. During infancy or childhood,
injury in DPD has not been clearly elucidated, although decreased mild frontal atrophy and ventricular enlargement will be noted, with
levels of the neurotransmitter-alanine and increased central nervous multiple patchy or confluent white matter lesions. Deep gray matter

Barkovich_Chap03.indd 131 5/6/2011 12:08:29 AM


132 Pediatric Neuroimaging

FIG. 3-39. Dihydropyrimidine dehydrogenase deficiency. A. Axial T2-weighted image shows abnormal hyperintensity (black arrows) in the medullary
olives/reticular activating system. B. Axial diffusion weighted image at the same level as (A) shows reduced diffusivity (white arrows) in the lesions. C. Axial
T2-weighted image at the level of the pons shows hyperintensity in the medial lemnisci and median longitudinal fasciculi. D. Axial T2-weighted image at
the level of the lateral ventricles shows enlarged ventricles and subarachnoid spaces, as well as abnormal periventricular T2 hyperintensity (white arrows).
(This case courtesy Dr. Greg Enns, Stanford, California.)

abnormalities (basal ganglia and cerebellar nuclei) are subtle early, white matter of affected patients (422). Broad peaks are consistently
becoming more obvious as myelination progresses; if the disease is seen at 1.3 and 2.4 ppm during metabolic decompensation, possibly
appropriately treated, these lesions may disappear on follow-up studies. due to accumulation of HMG CoA; however, the precise biochemical
Later, as brain myelination progresses, white matter changes become explanation is not yet clear.
obvious. In most reported cases, the subcortical white matter is spared
(Fig. 3-40), but some cases have shown involvement of the subcortical Congenital White Matter Hypoplasia/Familial
axons, as well (37,422). In patients who suffer episodes of severe hypo- Spastic Paraplegia
glycemia, the characteristic neuroimaging findings of hypoglycemic Congenital white matter hypoplasia is a term used to describe children
brain injury (caudate and putaminal injury (424) see Chapter 4) may in whom the volume of white matter in the cerebral hemispheres is
be superimposed upon the baseline findings of the HMG CoA lyase reduced without a known cause, such as white matter injury of pre-
deficiency (422). maturity, twin-twin transfusion syndrome, metabolic disease, or
Proton MR spectroscopy of patients with HMG CoA lyase defi- congenital infection (57). Affected children typically have primarily
ciency shows reduced NAA/Cr, increased Ch/Cr, and increased myo-I/Cr delayed motor development and are classified as having cerebral palsy.
ratios, suggesting enhanced membrane turnover and gliosis in the Seizures or cognitive disabilities are present in some. Congenital white

Barkovich_Chap03.indd 132 5/6/2011 12:08:30 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 133

and prognosis. Recently, clinical consensus definitions have been


developed specifically for the pediatric forms (occurring in patients
younger than 10 years) of inflammatory diseases of the CNS in an
attempt to create a uniform terminology and identify common clini-
cal and neuroimaging features associated with the proposed entities
in children (429–431).
As in adults, ADEM and MS are the most common idiopathic
inflammatory diseases of the brain in the pediatric patient popula-
tion, but their diagnosis is often complicated because no single clinical,
laboratory or imaging feature is specific to either disorder. Although
imaging findings may be suggestive, it is often impossible to confi-
dently diagnose inflammatory diseases of the CNS and in particular,
to differentiate them from many other pathological conditions of the
CNS (vascular, neoplastic, metabolic, etc.) solely by MRI.

Multiple Sclerosis
Although usually considered an adult disease, symptoms of MS
can begin during childhood. It has been estimated that up to 10%
of all patients with MS present during childhood (429,430), with
10,000 cases in the United States (432). The clinical presentation of
the disease in early childhood can range from school problems and
paresthesias to dramatic presentations suggesting diffuse encephal-
opathy with cerebral edema, meningismus, and impaired conscious-
ness (433). It is important to remember that 10% to 25% of children
initially diagnosed as having ADEM are ultimately diagnosed with MS
FIG. 3-40. 3-hydroxy-3-methylglutaryl-coenzyme A lyase deficiency. Axial (8,434). Other presenting phenotypes include optic neuritis (25%),
T2-weighted image shows abnormal hyperintensity of most of the centrum transverse myelitis (11%), and CIS with single presenting symptom
semiovale with sparing of the subcortical white matter (black arrows). other than optic neuritis, transverse myelitis, or ADEM (434). There-
fore, it is often not until the second attack of a neurologic episode that
the diagnosis of MS should be suggested. Recent studies suggest that
childhood-onset MS patients are more likely than adult-onset patients
matter hypoplasia is probably the end result of a number of processes, to have a relapsing-remitting course and have a longer course before
including genetic disorders of white matter development and prena- the onset of secondary progression; however, they reach states of irre-
tal injury/infection. Genetic causes are sometimes classified as familial versible disability at a younger age than patients with adult-onset MS
spastic paraplegia (233,241,425) (see IV.B.1.n, earlier in this chapter); (430).
many different genetic loci have been associated with these disorders New recommendations from an international panel of experts
(426) and the pathophysiology is only beginning to be worked out have emphasized the importance of MRI in the diagnosis of MS in
(233,427,428). adults (434,435). In particular, MRI is used to document dissemination
Imaging findings are similar in all cases. The volume of white mat- of lesions in space and in time. Unfortunately, these criteria are not as
ter in the cerebral hemispheres is reduced, but there is no evidence of useful in children as in adults (436). Currently, the most accurate cri-
reactive astrogliosis such as from ischemic or infectious injury. The teria for differentiating MS from other, nondemyelinating, neurologic
corpus callosum is abnormally thin but fully formed. In severe cases, disorders are based on MRI and consist of at least two of the follow-
the brainstem might be thin. Basal ganglia, cerebral cortex, and cer- ing: ≥5 lesions on T2-weighted images; ≥2 periventricular lesions; or
ebellum are normal. Diffusion imaging and proton spectroscopy are ≥1 brainstem lesion (434).
normal or show slightly reduced diffusivity and NAA, respectively.
Standard MRI in MS Imaging findings in children with MS are not
Idiopathic Inflammatory, Autoimmune, Infectious, and significantly different from those in adults (Fig. 3-41). Sharply margin-
Toxic Disorders Affecting White Matter ated lesions are generally seen in the white matter T1 hypointense with
Idiopathic inflammatory diseases of the nervous system include a T2/FLAIR hyperintense (Figs. 3-41 and 3-42); often, on T2-weighted
wide range of clinical and histopathological entities that are classified images of active plaques, a central area of very high signal intensity is
according to the pattern of involvement (monofocal or multifocal, with surrounded by a peripheral area of moderately high signal intensity
possible involvement of brain, spinal cord, and nerve roots), sever- (Figs. 3-41C and 3-42A). Plaques are common in the cerebral cortex
ity (asymptomatic, mild, and severe), and disease course (monopha- but these are small and, therefore, are not well shown by MRI at 1.5T
sic, multiphasic, relapsing-remitting, progressive, etc). Traditionally, or 3T at this time. FLAIR sequences show abnormalities in MS patients
isolated optic neuritis and transverse myelitis have been considered even when standard T2-weighted images are normal (437–439). How-
monofocal and monophasic diseases; recently, the term clinically iso- ever, FLAIR is somewhat limited in the detection of posterior fossa and
lated syndromes (CIS) has been used to describe these entities. ADEM spinal cord lesions (15,440,441); therefore, we routinely acquire both
is a multifocal, but usually monophasic and self-limiting process. MS FLAIR and T2-weighted sequences in patients with known or suspected
and neuromyelitis optica (NMO) are chronic (relapsing) multifocal MS. Enhancement (Fig. 3-41D) may or may not be seen, depending
inflammatory diseases. Although inflammatory diseases of the CNS on the acuity of the plaque; new lesions enhance for an average of 3
in adults and children have many similarities, distinct differences have weeks (median 2 weeks) (442). Overall, children with MS tend to have
been identified that have potential clinical relevance for treatment fewer lesions in general and fewer enhancing lesions than adults. In

Barkovich_Chap03.indd 133 5/6/2011 12:08:31 AM


134 Pediatric Neuroimaging

children, lesions in the juxtacortical and deep white matter are much inflammatory infiltrate; in our experience, reduced diffusivity is seen
more common than those in the periventricular white matter or cor- earlier than enhancement in acute plaques. Single voxel proton MR
pus callosum (434). In addition, the incidence of tumefactive plaques spectroscopy (Fig. 3-42D) may also be useful; short echo spectra dem-
(Figs. 3-42) (443) and posterior fossa plaques (Fig. 3-41E) (444,445) onstrate elevated glutamate and glutamine in the lesion, while both long
seems to be somewhat higher, and the disease activity (frequency of and short echo spectra show elevated choline, elevated lactate, elevated
onset of new lesions) seems to be greater (445). Tumefactive plaques lipid, and decreased NAA (447). These diffusion and MRS findings are
in the brainstem and spinal cord in children may be associated with similar to those seen in tumors; therefore, a search for other plaques,
extensive swelling and MR signal changes suggestive of neoplasm (446). serial clinical exams, and serial imaging exams may be necessary to
Tumefactive plaques can sometimes be differentiated from tumors by make a definitive diagnosis of MS in such children (446). Readers inter-
the presence of other, more typical plaques, lack of mass effect, location ested in further information concerning imaging findings in MS are
adjacent to the ventricular surface, and an incomplete rim of enhance- referred to any standard adult neurology or neuroradiology text.
ment (Fig. 3-42C) as contrasted with a complete ring of enhancement The differential diagnosis for multiple white matter regions of T2/
in tumors. FLAIR hyperintensity, some of which may or may not enhance, include
Diffusion-weighted images (Fig. 3-42B) may show reduced dif- multiple small infarctions (as from antiphospholipid antibody syn-
fusivity in the regions of active inflammation, resulting from the drome, SLE, Takayasu disease, or even arterial dissection), monophasic

FIG. 3-41. Multiple sclerosis in a 15-year-old. Often, lesions in childhood are very similar to those in adults. A. Axial T2-weighted image shows multiple
lesions in both cerebral hemispheres (small white arrows), involving the periventricular, deep, and subcortical white matter. The large lesion in the middle
of the right hemisphere (large white arrow) is very hyperintense and is surrounded by edema (more moderate hyperintensity). B. Axial reformation of volu-
metric FLAIR sequence at the same level as (A) demonstrates that more subtle lesions are detectable than on T2-weighted images. C. Coronal T2-weighted
image shows the same large hyperintense, edematous lesion (large white arrows) in addition to a subcortical plaque (white arrowhead) and a plaque in the
left midbrain-pons junction (small white arrows). D. Coronal T1-weighted image after intravenous injection of paramagnetic contrast shows rim enhance-
ment of the large right hemispheric lesion (white arrows) and the subcortical plaque (white arrowhead). E. Coronal T2-weighted image shows bilateral cer-
ebellar lesions (white arrows) in addition to supratentorial lesions (white arrowheads). F. Sagittal T2-weighted image of the cervical spine shows a lentiform
hyperintense plaque (black arrows) in the dorsal spinal cord at the C1 level. Note that the dorsal surface of the cord bulges slightly outward, indicating mass
effect from an acute lesion.

Barkovich_Chap03.indd 134 5/6/2011 12:08:31 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 135

FIG. 3-41. (Continued) G. Axial T2-weighted image


through the C1 level shows the plaque (black arrows)
involving the dorsal columns of the spinal cord. H. Sagit-
tal T1-weighted image of the cervical spine after intra-
venous administration of paramagnetic contrast shows
enhancement (white arrows) of the plaque.

FIG. 3-42. Tumefactive multiple sclero-


sis. A. Axial T2-weighted image shows an
area of T2 hyperintensity (white arrows)
in the right parietal lobe. Note that the
lesion has little mass effect, is located
adjacent to a lateral ventricle, and has an
area of marked hyperintensity centrally.
B. Axial diffusivity (Dav) image shows
reduced Dav in the medial aspect; this
likely corresponds to the location of the
inflammatory infiltrate (white arrows).
C. Axial T1 postcontrast image shows an
incomplete rim of enhancement (white
arrows) at the posteromedial aspect of
the lesion. The dense enhancement of
the more anterior part is atypical. Biopsy
showed demyelination. D. Single voxel
proton MRS (TE = 144 ms) shows ele-
vated choline, low NAA and high lactate,
typical findings for a tumefactive demy-
elinating plaque.

Barkovich_Chap03.indd 135 5/6/2011 12:08:33 AM


136 Pediatric Neuroimaging

demyelinating diseases (such as ADEM or acute transverse myelitis), lesions have decreased choline and decreased NAA (indicating reso-
migraine, and infections/inflammatory disorders such as Lyme dis- lution of inflammation but loss of neurons or axons) (463). Reduced
ease or neurosarcoidosis (431). When only a single lesion is identified, NAA (464) and elevated lactate (464,465) may also be seen in acute
tumor is the most concerning differential diagnosis; dynamic perfu- plaques (Fig. 3-42). The decreased NAA correlates with axonal damage,
sion imaging is the most useful technique in differentiating these two whereas the increase in choline correlates with both local inflammation
entities (448) (see the beginning of Chapter 7). A search should also be and increased myo-I with reactive astrogliosis, while the lactate is likely
made for lesions within the cerebral cortex, or juxtacortical lesions at secondary to the inflammation (465). It should be noted that MRS of
the border between the cortex and the subcortical white matter, which a tumefactive MS plaque may be nearly identical to that of tumor, so
are becoming recognized as an important component of this disease anatomic characteristics (see the section in the beginning of Chapter 7)
(434,449,450). and other methods, such as dynamic perfusion imaging (which shows
increased blood volume in high-grade intraparenchymal tumors but
More Advanced MR Techniques in MS Although precontrast reduced blood volume in demyelinating plaques (448) ), are needed to
and postcontrast T1, T2, FLAIR, and diffusion imaging are stan- establish the diagnosis. The fact that decrease in NAA correlates with
dard sequences for the evaluation of MS, many other MR techniques axonal damage has led to the use of whole brain (“global”) proton MRS
may be useful in the evaluation of patients with MS, particularly in to quantify whole brain NAA as a marker of total axonal/neuronal dys-
the research setting. The use of magnetization transfer techniques function. Such global analysis allows inclusion of disease in normal-
improves visibility of enhancing MS lesions by suppressing the T1 appearing white matter that might otherwise be missed (454).
shortening effects of myelin. However, because some MS lesions have
high intensity on precontrast T1-weighted images, it is necessary to Neuromyelitis Optica (Devic Disease)
obtain precontrast, as well as postcontrast, T1-weighted images if NMO has been historically characterized by sequential or concomitant
magnetization transfer is used (437). Quantification of white mat- optic neuritis and transverse myelitis; however, the spectrum of the
ter injury can be accomplished via the calculation of magnetization disease has recently been expanded to include patients with encephal-
transfer ratios and diffusion tensor imaging (see Chapters 1 and 2) opathy, seizures, intractable vomiting, and brainstem-mediated hiccups
(451–454). Although FLAIR is somewhat limited in the detection of (466). Indeed, in early stages, it may be clinically difficult to differenti-
posterior fossa and spinal cord lesions (15,440,441), double inversion ate from MS(466,467). Although it is most common in adults, it may
recovery, in which the signals of both CSF and white matter are sup- be encountered in children (age of onset 14–65 years). The disorder is
pressed through the use of two separate inversion pulses (455), seems more common in women and in nonwhite populations (466–468). A
to be useful in these regions (456). viral prodrome is present in up to 84% of patients. CSF studies reveal
The ability of diffusion tensor imaging techniques to assess dam- intrathecal oligoclonal bands in 20% of patients, but, more importantly,
age to the microstructure of the brain makes it potentially the most recently developed blood test may reveal the presence of a highly spe-
useful technique for assessing brain injury in this disorder (magneti- cific serum antibody, the so-called neuromyelitis optica-immunoglob-
zation transfer is more specific for myelin damage), as axonal injury ulin G (NMO-IgG) (466–468). Prognosis is better in children (complete
results in reduced average diffusivity and reduced fractional anisotropy recovery in 77%) than in adults (18%). The disease, although initially
(454). Although these findings seem to be less severe in pediatric-onset believed to be monophasic, may be multiphasic, with recurrences within
disease than in adult-onset MS (457), early results suggest that disease 6 months to 5 years (466–468). In the acute phase, swelling of optic
in white matter pathways is widespread, with increased diffusivity and nerves and the affected part of the spinal cord are seen, but ultimately
reduced FA, even in normal-appearing white matter (458). Within the optic nerve atrophy and necrosis within the spinal cord develop.
plaques themselves, highest diffusivity values are found in T1 hypoin- Identification of lesions within the optic nerves or suprasellar
tense lesions, representing long standing destructive damage. Dif- optic pathways may be challenging and requires high-resolution imag-
fusivity values in enhancing and nonenhancing plaques are variable, ing, including STIR sequence, which appears to be the most sensitive
although anisotropy is always lower in enhancing than in nonenhanc- technique to detect optic neuritis. If lesions associated with swelling
ing plaques, suggesting that the inflammatory effects have a more vari- are located at the level of the proximal optic pathways, optic glioma
able impact on diffusivity than on anisotropy and that blood–brain may be a differential diagnostic consideration, but differences in clini-
barrier breakdown probably varies during these active periods (454). cal presentation and evolution usually allow a fairly confident differ-
Magnetization transfer techniques are also useful in differentiating entiation. Detection of spinal cord lesions is generally easy by MRI.
the inflammation associated with MS (little reduction of magnetiza- The T2 hyperintense lesion, often occupying almost the entire cross-
tion transfer) from actual demyelination (more marked reduction of sectional area of the cord, is usually more extensive (3–17 segments)
magnetization transfer) (459). Magnetization transfer ratios dimin- than MS-related cord lesions, and signal enhancement after intra-
ish more rapidly in the acute phase and are overall lower in secondary venous gadolinium injection may be seen in 20% of patients (469).
progressive MS than in relapsing-remittingMS; this observation may Lesions may be detected in brain parenchyma during the course of the
indicate a difference in the nature of these forms of the disease (460). disease in up to 60% of patients (469,470). Although this contradicts
Detection of small lesions of the cerebral cortex can be improved by the initial presumption that the disease is limited to the spinal cord
increasing signal-to-noise with parallel imaging or higher field strength and optic pathways, it does not rule out the diagnosis of the disease if
(461), or by the use of sequences that suppress both the signal of white the currently proposed diagnostic criteria are otherwise met. Until a
matter and that of CSF (462). few years ago, the diagnostic criteria of NMO required (beyond optic
Localized proton spectroscopy from white matter plaques of chil- neuritis and acute myelitis) at least two of three supportive criteria, two
dren with MS shows a decrease in NAA and creatine and an increase of which relied on MRI, notably the presence of a contiguous spinal
in choline and myo-I relative to age-matched controls (117). Adja- cord lesion and a brain MRI study not satisfying the criteria of MS.
cent white matter is normal, but adjacent cortical gray matter shows More recently, brain imaging findings have been dropped from the set
reduced NAA. This data is supported by reports in the adult litera- of supportive criteria (466–469). As spinal cord lesions may “split up”
ture suggesting that acute MS plaques have increased choline and nor- into several, apparently distinct, smaller lesions during the recovery
mal NAA (indicating inflammation but no cell loss), whereas chronic period and brain lesions may increase in number with time (ultimately

Barkovich_Chap03.indd 136 5/6/2011 12:08:34 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 137

fulfilling the diagnostic imaging criteria for MS), specificity of the MRI gray matter are involved, but to a lesser extent than white matter. In the
is highest if it is performed as early as possible after the initial clinical spine, both gray and white matter may be affected.
presentation. On imaging studies, moderate to large areas of demyelination
(low density on CT, T1 hypointensity and T2/FLAIR hyperintensity on
Acute Disseminated Encephalomyelitis (ADEM) MRI) are seen in the subcortical and deep white matter of one or both
Children may develop acute encephalitis late in the course of a viral ill- hemispheres, usually asymmetrically (Fig. 3-43) (481,482). CT, which
ness or after a vaccination; less commonly, encephalitis may occur after is often the first imaging modality used in the emergency diagnostic
a bacterial infection (particularly mycoplasma) or drug ingestion or workup of a child presenting with a severe acute neurological syndrome,
with no history of a preceding event (471,472). This disorder is called is often negative in patients who are later diagnosed to have ADEM.
acute disseminated encephalomyelitis (ADEM) or parainfectious MR examination of the brain is usually positive, but lesion patterns are
encephalomyelitis. The most common clinical presentation is the onset highly variable. Periventricular white matter is involved in about half
of focal neurological signs a few days to several weeks after the clini- of patients with ADEM, in contrast to greater than 90% involvement
cal onset of the viral infection. These are usually severe and indicate in MS and the corpus callosum is much more commonly involved in
multifocal involvement of the CNS. Headache of rapid onset is com- MS than in ADEM; these criteria might be useful in differentiating
mon, followed by vomiting (32%–35%), impairment of consciousness ADEM from an initial episode of MS(9,434,477). Other factors that
(45%–69%), and seizures (13%–35%). Nuchal rigidity is often present. favor a diagnosis of MS are absence of a diffuse bilateral pattern and
Encephalopathy exhibiting behavioral changes (confusion, excessive the presence of black holes (sharply marginated regions of T1 hypoin-
irritability) and/or alteration of consciousness is a virtually, “sine qua tensity) (434). MRI reveals abnormalities of the deep cerebral nuclei
non” diagnostic criterion. Sometimes the disease presents with isolated (Fig. 3-43B) in approximately 50% of affected pediatric patients (477,
acute psychosis. Focal neurological signs include long tract signs (85%, 483–485). The brainstem (Fig. 3-43A), spinal cord (Fig. 3-43D), and
with acute hemiparesis in 23%–77%), cerebellar ataxia (50%–65%), cerebellar white matter (Fig. 3-43A) are each involved in about 30%
cranial nerve palsy (44%–45%), and visual loss (13%–23%). Spinal to 50% of affected patients (477,483,485); spinal cord lesions often
cord involvement is present in approximately 24% of patients (473). are located in the central gray matter (Fig. 3-43E). Petechial hemor-
CSF cultures are repeatedly negative for bacteria, fungi, or viruses and rhage may be present within the lesions; this is best demonstrated by
typically (although not always) no oligoclonal bands are found; eleva- T2-weighted gradient-echo images or susceptibility-weighted images.
tion of proteins and white cell count (pleocytosis) are frequent (473). In the subacute phase, the lesions will show various patterns of enhance-
Measles, mumps, chickenpox, rubella, pertussis, mycoplasma, herpes, ment (nodular, diffuse, a complete ring, or a partial ring) after infusion
cytomegalovirus, Campylobacter jejuni, and Group A streptococcus of contrast (484,486). Diffusion images show increased diffusivity in
are the organisms most often implicated in this disease; however, many the foci of demyelination in both the acute and the subacute phases,
cases seem to occur “spontaneously,” or follow nonspecific infections a factor that is useful in differentiating ADEM from vasculitis (lesions
(474–476). Patients usually respond well to steroid therapy, and the from vasculitis show reduced diffusion in the acute phase). Perfusion
neurological findings evolve and then resolve over a period of weeks imaging will show normal or reduced blood volume. An important
(477). The majority of individuals make a complete recovery with no point is that the T2 changes in the brain evolve over time, as does the
neurological sequelae; 10% to 30% will have some permanent neuro- clinical syndrome. As a result, follow-up MR studies may show an
logical damage (474,475). apparent worsening in spite of clinical improvement (487). Although
Sometimes the disorder waxes and wanes over a period of months. ADEM may recur, as mentioned earlier, under such circumstances the
The International Pediatric MS Study Group has proposed operational clinical exam and CSF analyses are more indicative of the course of the
definitions to address this phenomenon (478). Monophasic ADEM is disease than the MR results.
defined as a multifocal clinical syndrome in patients without a prior Other disorders may present with neurologic and imaging findings
demyelinating event. Symptoms may evolve over a period of 3 months that resemble those of ADEM; these disorders include viral encephali-
after the clinical onset. Recurrent ADEM is characterized by a second tis, collagen vascular diseases, Whipple disease, neurosarcoidosis, and
attack, involving the same anatomic areas, more than 3 months after MS(431,446,488–490). These alternative diagnoses must always be
the initial event. Multiphasic ADEM is characterized by a second event kept in mind when patients present with acute neurologic deficit and
in a different anatomic area (a new focal neurologic deficit or a new imaging findings compatible with ADEM.
lesion on MRI) more than 3 months after the initial event. A significant
number of patients who receive an initial diagnosis of ADEM eventu- Acute Hemorrhagic Encephalomyelitis
ally receive a diagnosis of MS; this number could be as high as 25% Acute hemorrhagic encephalomyelitis is a rare (2%), hyperacute variant
(5,434,479). The chances of a subsequent attack of demyelination is of ADEM in which the involved regions of brain undergo hemorrhagic
higher in patients who are 10 years old or greater at the time of the necrosis. It has been associated with herpes simplex and Epstein-Barr
initial attack, have a pattern suggestive of MS (see paragraph on imag- virus infections, although the inciting infection is often not identified
ing findings below) on the initial MRI, or have an optic nerve lesion. (491,492). It is rarely iatrogenic (induced by melarsoprol, an organic
Chances of a second attack are lower in patients with myelitis (symp- drug containing arsenic that is used to treat trypanosomiasis). At
toms of spinal cord involvement) or mental status changes (encephal- autopsy, the brain is markedly swollen and numerous hemorrhagic
opathy) (480). foci are seen in cerebral and cerebellar white matter and the brainstem
ADEM is likely caused by an autoimmune response; it is postu- (pons). Microscopically, fibrinoid necrosis of vessel walls (both arter-
lated that the precipitating illness induces a host-antibody response ies and veins) is found in conjunction with perivascular tissue necro-
against a central nervous system antigen. This postulate is supported sis and hemorrhages. Mixed perivascular infiltrates (neutrophils and
by the fact that the gross pathological and histological manifestations mononuclear cells) are also seen, with evidence of demyelination and
of ADEM closely resemble those of experimental allergic encephalitis, axonal fragmentation. Patients often progress rapidly into delirium
an experimentally induced autoimmune disease caused by antibodies and coma and most die within the first few days to a week of onset
to myelin. Pathologically, a diffuse perivenous inflammatory process (474,493). The few survivors have serious neurologic sequelae (494).
causes confluent areas of demyelination. In the brain, cortical and deep Imaging findings are of multiple or large unilateral or bilateral lesions

Barkovich_Chap03.indd 137 5/6/2011 12:08:34 AM


138 Pediatric Neuroimaging

FIG. 3-43. Acute disseminated encephalomyelitis in a 2-year-old.


A. Axial T2-weighted image shows abnormal hyperintensity in the
left medullary pyramid (black arrow) and in the white matter of
both cerebellar hemispheres (white arrows). B. Axial T2-weighted
image at the level of the basal ganglia shows multiple lesions of vari-
able size involving the thalami and lentiform nuclei (white arrows),
as well as the cerebral white matter. C. Axial T2-weighted image at
the level of the bodies of the lateral ventricles shows asymmetrical
areas of hyperintensity of varying size in the deep and subcortical
white matter. D. Sagittal T2-weighted image shows multiple areas
of involvement (white arrows) in the thoracic and lumbar spinal
cord. E. Axial T2-weighted image through an involved area of the
thoracic spinal cord shows that the lesion (white arrows) is in the
central gray matter of the cord.

of the cerebral white matter, most commonly involving the frontal and diagnosis (503). Therefore, the diagnosis may not be known at the time
parietal lobes. The basal ganglia, thalami, brainstem, cerebellum, and of the initial neuroimaging study (488). Neurologic signs and symp-
spinal cord may be involved. Overall, the lesions are similar to those in toms include neuropsychiatric difficulties (most commonly depression;
ADEM except that the lesions are larger, hemorrhagic, and have more also concentration or memory problems, or frank psychosis) in 40% to
associated edema and mass effect (11,160,495). 48%, headache (22%–64%), seizures (usually generalized tonic-clonic
seizures, 20%), ischemic events (15%–28%), chorea (3%–28%), visual
Collagen Vascular Diseases/Systemic Lupus Erythematosus loss (12%), peripheral neuropathy (5%), cranial neuropathy (8%),
Collagen vascular diseases are immunologically mediated syndromes vertigo (4%), and myelopathy (4%) (488,499,503). Neurologic events
in which antigen-antibody complexes are found in various body tis- commonly occur in SLE, potentially related to therapy, infection, com-
sues, resulting in multiorgan dysfunction. The most common of these plications, or to functional problems related to the underlying disor-
disorders presenting with neurologic signs and symptoms in children der; the specific cause is, therefore, often difficult to pinpoint (502,504).
and adolescents is systemic lupus erythematosus (SLE). A variety of pathologic abnormalities have been identified in the brains
The average age of patients at the time of diagnosis of childhood of patients with SLE, including cerebral atrophy, cerebral infarcts
SLE is 13 years but patients may present as young as age 8 years. Males due to emboli, vasculitis (often associated with the antiphospholipid
are more commonly affected in childhood (496); in adults, females antibody syndrome (505,506) ), petechial hemorrhages, and areas of
are more commonly affected with a ratio of approximately 3:1. The demyelination; these are attributed to angiopathy, direct autoimmune
average age at the onset of neurologic manifestations, which develop damage to brain, demyelination, and ischemia from thromboembo-
in 13% to 45% of patients with SLE, is 14 years (488,497–502). In 32% lism (507), all of which are believed to result from the action of cyto-
to 47% of affected patients, the neurologic manifestations either pre- toxic antibodies that vary in their composition and antigenicity and,
cede the diagnosis of SLE or are coincident with the diagnosis; another thus, attack various vulnerable parts of the central nervous system
30% manifest neurologic signs or symptoms within 1 year of time of (501–504,506).

Barkovich_Chap03.indd 138 5/6/2011 12:08:34 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 139

FIG. 3-44. Systemic lupus erythematosus vasculitis in


a 10-year-old girl. A. Axial T2-weighted image shows
abnormal hyperintensity in the caudate nuclei, anterior
lentiform nuclei, anterior limbs of the internal capsules,
and right posterior lentiform nucleus (white arrow).
These regions reverted to normal on subsequent scans.
B. Postcontrast T1-weighted image shows some promi-
nent curvilinear enhancement, presumed to be vessels.
C. Collapsed view from a 3D time of flight MRA of the
circle of Willis shows several areas of narrowing (white
arrows) of the M1 segments of the middle cerebral
arteries.

Imaging studies may show a variety of findings that vary with the the lesions are paramedian in location, it is not clear whether they rep-
indication for imaging. In patients with neuropsychiatric disorders, CT resent areas affected by SLE (vasculitis or cerebritis) or those that are
and MRI are usually negative or show large CSF spaces (488). In patients abnormal because of recent hypertension or seizure, both common
with seizures, MRI/CT may show focal lesions (25%) or may be nor- in SLE, with consequent cortical and subjacent white matter changes
mal. The focal lesions are typically areas of low attenuation (CT) or T1 (510,511) (these changes have, unfortunately, been given the name
hypointensity and T2/FLAIR hyperintensity (MRI) in the white mat- “posterior reversible encephalopathy syndrome” (512) ). In general,
ter of the cerebrum, cerebellum, brainstem, or spinal cord (Figs. 3-44 abnormalities secondary to seizures or hypertension are bilateral, sym-
and 3-45) (508). The basal ganglia may be involved; if so, angiogra- metrical, paramedian, involve the cortex and subcortical white matter,
phy should be performed to look for vasculitis (Fig. 3-44). Spinal cord and are more prevalent in the posterior aspect of the brain (occipital
lesions may have moderate associated mass effect and edema (490). and parietal lobes more than frontal lobes) (510,513). Although theses
The MR lesions are believed to represent either autoimmune cerebritis/ abnormalities are most often reversible, they can sometimes result in
myelitis or parenchymal involvement secondary to small vessel vasculi- permanent brain injury (510,514). It is important to perform diffu-
tis. Depending on the underlying process, diffusion imaging may show sion imaging in such cases; those areas with normal or increased diffu-
normal or reduced diffusivity. Some peripheral enhancement after sion will completely reverse, while those areas with reduced diffusion
intravenous administration of paramagnetic contrast may be present have suffered permanent injury and will atrophy (513). Other findings
in the subacute phase. In adults, transmural angiitis may result in vas- in patients with SLE include infarcts, which are typically the result of
cular rupture and hemorrhage (509), but this has not been reported in vasculitis, and may involve both superficial and deep gray matter. They
children. When lesions are in the cerebral hemispheres, affected areas are typically asymmetric, which helps differentiate them from changes
tend to involve subcortical white matter, with or without overlying cor- that result from seizures or hypertension, and have reduced diffusion in
tex (Fig. 3-45), and may revert to normal on subsequent scans. When the acute phase. Vascular imaging may be helpful, showing angiopathy

Barkovich_Chap03.indd 139 5/6/2011 12:08:35 AM


140 Pediatric Neuroimaging

FIG. 3-45. Multiple lesions


due to antiphospholipid anti-
body syndrome in a 14 years
old with systemic lupus ery-
thematosus. A. Axial FLAIR
image shows bilateral cerebel-
lar (white arrows) and temporal
lobe (white arrowheads) lesions
in a teenager who presented
with seizures. B. Coronal FLAIR
image shows that the cerebral
lesions (white arrowheads) are
subcortical in location.

(irregularity of arterial lumens), arterial occlusions, or venous occlu- cause and pathogenesis of this disorder have not been determined
sions, as well as focal infarcts. Infarcted tissue will, obviously, not return (517,518); but current theory suggests a hyperosmotically induced
to a normal appearance. Proton spectroscopy has been shown to reveal demyelination process resulting from rapid intracellular/extracellu-
decreased NAA/Cr ratios in the basal ganglia of patients with major lar to intravascular water shifts producing relative glial dehydration,
symptoms, such as psychosis or seizures, as compared to those patients myelin degradation, and/or oligodendroglial apoptosis (519). In chil-
with less severe signs and symptoms (515). The NAA/Cr ratios appeared dren, central pontine myelinolysis has been reported to occur with
to correlate with the course of the disease in one study (516). various disorders including fulminant hepatitis, liver transplantation,
diabetic ketoacidosis, chronic debilitation, treatment of hyperam-
Osmotic Myelinolysis in Childhood monemia and craniopharyngioma, and malnutrition often associated
In adults, osmotic myelinolysis is seen primarily in the pons (central with severe fluid and electrolyte disturbances (520–525).
pontine myelinolysis) and most commonly occurs among chronic alco- The imaging appearance of childhood myelinolysis is identical to
holics in poor nutritional states. Both the poor nutritional condition that in adults, with a focus of low attenuation (CT) or prolonged T1
and an overly rapid correction of severe hyponatremic dehydration and T2 relaxation times (MRI) in the central pons (Fig. 3-46A); the
seem to predispose them to the development of osmotic myelinolysis corticospinal tracts are characteristically spared. Diffusion is reduced
in the central pontine region. Other patients with severe nutritional in the affected area(s) (Fig. 3-46B) (526). Both the patient and the MR
disturbances and fluid/electrolyte imbalances are also at risk. The exact scan may return to normal after slow treatment of nutritional and

FIG. 3-46. Central pontine myelinol-


ysis in a child with lymphoma.
A. Axial SE 2500/30 image shows T2
prolongation (arrow) in central pons.
B. Average diffusivity (Dav) image shows
reduced Dav, indicating acute cellular
injury (arrow).

Barkovich_Chap03.indd 140 5/6/2011 12:08:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 141

electrolyte abnormalities (525,527). It is important to remember that mild behavioral change to frank obtundation, with occasional ataxia,
osmotic myelinolysis can develop in other areas, including the basal aphasia, or psychological disturbance (534,541). The most common
ganglia (528), thalami (529), cerebral cortex (usually at the cortical- imaging finding is edema (swelling and T2/FLAIR hyperintensity) in
white matter junction) (519,530), and middle cerebellar peduncles the external and extreme capsules (Fig. 3-48) (542). Rarely, lead intoxi-
(531). (Relatively few disorders cause abnormalities of the middle cere- cation may lead to localized edema in the cerebellum causing hydro-
bellar peduncles. In addition to Wallerian degeneration due to pontine cephalus and presentation as a posterior fossa mass (541,543). CT and
injury, it is seen in spinocerebellar ataxias types 2 and 3, Wilson disease, MRI in these cases show localized cerebellar edema with mass effect
and fragile X permutation (532,533) ). and mild enhancement after intravenous contrast infusion (541,543).
The appearance is identical to that of acute cerebellitis from viral ill-
Toxins nesses (see Chapter 11).
Several exogenous toxins can cause demyelination. Many of these
intoxications result in a spongiform leukoencephalopathy that initially Solvent Abuse
affects the peripheral white matter, including the subcortical U fibers. Solvent abuse is a serious problem in some cultures, particularly in
Among the toxic substances that cause demyelination are triethyl tin, adolescents and young adults. Toluene and other forms of organic
hexachlorophene, cuprizone, actinomycin D, and isoniazid (160,534– materials (trichloroethane, methylchloroform) are inhaled and rap-
536). A similar pattern is described after ingestion of toxic heroin idly absorbed into the blood stream, from where they can cross the
(Fig. 3-47) (537–540). All of these disorders cause bilateral symmetric blood–brain barrier and involve the central nervous system. Patients
white matter injury that involves the subcortical U fibers early in the may present with acute encephalopathy or with chronic mental and
course of the clinical illness. neurologic deterioration. Insomnia, forgetfulness, anosmia, and tin-
nitus are reported to be common (544). Neurologic exam reveals
Lead Encephalopathy cerebellar ataxia (50%), tremors of the extremities (35%), increased
Special mention must be made of lead encephalopathy. The typi- deep tendon reflexes (25%), and rigidity (25%) (544). Pathologic stud-
cal clinical presentation of lead intoxication is abdominal cramping, ies of the brains of affected patients show neuronal and axonal loss
nausea, and vomiting. Neurologic signs and symptoms vary from with associated demyelination: diffuse demyelination of the cerebral

FIG. 3-47. Acute leukoencephalopathy


from toxic heroin ingestion. A and B. Axial
T2-weighted images show abnormal hyper-
intensity in the pons (p), middle cerebellar
peduncles (m), and cerebral white matter
(w). Note that some of the subcortical white
matter is affected. C. Coronal FLAIR image
shows involvement of the corpus callosum
(white arrows) but sparing of the basal gan-
glia. D. Average diffusivity map shows mildly
reduced diffusivity in the deep cerebral white
matter. E. Proton MRS (TE = 26 ms) from
the cerebral white matter shows low NAA
due to neuronal injury, broad macromolecu-
lar peaks at 0.9 and 1.3 ppm from damage to
membranes and release of lipids, and a small
amount of lactate (arrows point to the dou-
blet at 1.3 ppm) due to impaired mitochon-
drial production of ATP.

Barkovich_Chap03.indd 141 5/6/2011 12:08:37 AM


142 Pediatric Neuroimaging

to normal white matter. The T2 prolongation of white matter in gray


matter disease, however, is not as marked as in the leukodystrophies
(e.g., see Figs. 3-1 to 3-3), presumably because the white matter abnor-
mality results from Wallerian degeneration of the axons, rather than
from inflammation or destruction of oligodendrocytes or myelin. In
some instances, patchy, focal areas of white matter abnormalities may
also be present (in a primarily and dominantly gray matter disease);
the exact nature and pathogenesis of these findings are not yet fully
understood. In this section, gray matter disorders that have distinctive
abnormalities on neuroimaging studies are discussed.

Gray Matter Disorders Primarily Involving


the Deep Gray Cerebral Nuclei
Pantothenate kinase–Associated Neuropathy
(Neurodegeneration with Brain Iron Accumulation 1, Formerly
Hallervorden-Spatz Disease)
Pantothenate kinase–associated neuropathy (PKAN, also called neuro-
degeneration with brain iron accumulation 1 or NBIA1, OMIM 234200)
is a rare metabolic disorder that is characterized clinically by progressive
gait impairment, gradually increasing rigidity of all limbs, slowing of
FIG. 3-48. Lead encephalopathy in a 9-year-old child. Axial T2-weighted voluntary movements, choreoathetotic or tremulous movement disor-
image shows hyperintense edema (black arrows) in the external and extreme der, dysarthria, and mental deterioration (548,549). Optic atrophy or
capsules. retinitis pigmentosa may develop (549). Although the age of onset var-
ies considerably, most patients begin to show some neurological dete-
rioration during the last half of the first decade or first half of the second
and cerebellar white matter; degeneration and gliosis of ascending and decade of life (550); however, the age of onset can vary greatly (550). In
descending long fiber tracts, peripheral nerves, and axons of the corpus addition, all patients with PKAN do not have all of the manifestations of
callosum; and generalized cerebral and cerebellar atrophy (545,546). the disorder, probably because more than one genetic disorder underlies
MRI shows loss of gray-white discrimination, diffuse mild T2 pro- this disease. A number of families of diverse ethnic backgrounds have a
longation of the cerebral and cerebellar white matter, often some T2 mutation on chromosome 20p12.3-p13 (551); this has been found to be
shortening of the putamina, and generalized atrophy of cerebrum and a novel pantothenate kinase gene (PANK2) (552), the protein product
cerebellum (540,545,546). In severe cases (associated with a longer of which localizes to mitochondria in animal models (553).
duration of abuse (544) ), T2 shortening may be seen in the thalami, Two forms of PKAN have been defined (554). The classic form is
substantia nigra, and cerebral cortex (547). characterized by disease onset before the age of 10 years (mean 3.4 ±
3.0 years), manifest as extrapyramidal signs (dystonia, dysarthria, and
Progressive Multifocal Leukoencephalitis
choreoathetosis, seen in >95%), pyramidal signs (spasticity, hyperre-
Progressive multifocal leukoencephalitis is a rare demyelinating disease flexia, and extensor toe signs, seen in 25%), cognitive decline (29%),
caused by a papovavirus; it is seen almost exclusively in immunocom- and retinopathy (68%); most of these patients have mutations of
promised patients. It causes a combination of isolated and confluent PANK2 (555). The atypical form is characterized by a slightly older
areas of T2 prolongation, primarily in the white matter. Mass effect age of onset (13.7 ± 5.9 years), less common (73%), less severe, and
and contrast enhancement are minimal, if present at all. This disease is more slowly progressive extrapyramidal signs, and uncommon retin-
more fully discussed in Chapter 11. opathy (20%); pyramidal signs are about equally common (18%) as in
the classic form (555); these patients are less likely to have mutations
Subacute Sclerosing Panencephalitis
of PANK2 and are referred to as having Neurodegeneration with brain
Subacute sclerosing panencephalitis is a slow virus infection of the CNS iron accumulation (554).
that develops secondary to measles infection. Imaging studies show a The striking pathologic finding in affected patients is rust-brown
nonspecific pattern of increased water in white matter with localized pigmentation of the globus pallidus and zona reticulata of the substan-
cortical lesions in some cases. This disease is discussed more fully in tial nigra. Iron is concentrated in these regions, where it is found in large
Chapter 11. astrocytes, microglia, and neurons. Presumably as a result of the iron
deposition, one finds also symmetrical destruction of the globus pal-
Metabolic Disorders Primarily Affecting lidus (particularly the internal segment) and substantia nigra (556).
Imaging studies reflect the underlying pathology. CT scans may
Gray Matter
show low- or high-density foci in the globi pallidi. Although the cause of
A large number of conditions affecting the pediatric central nervous this variability has not been proven pathologically, it is likely that the low
system affect the gray matter, including ischemia, inflammation, infec- density lesions reflect tissue destruction, whereas the high-density foci
tions, malformations, and injuries, in addition to inborn errors of are a result of subsequent dystrophic calcification. MRI shows hypoin-
metabolism. Many metabolic gray matter disorders have nonspecific tensity in the globus pallidus on T2-weighted images (Fig. 3-49, more
imaging findings. In some disorders, such as some mucolipidoses, pronounced hypointensity than is normally seen in the second decade,
Gaucher disease, and some forms of Niemann-Pick disease, the brain see Chapter 2), resulting from iron deposition (557–559); this finding
appears normal. In others, the cerebral cortex may be thin; the cere- may appear before clinical symptoms develop (560). The globus pallidus
bral white matter is often diminished in volume and has low attenua- hypointensity may not be apparent on low field strength scanners, even
tion (CT) and prolonged T1 and T2 relaxation times (MRI) compared if gradient-echo techniques are used; on higher field strength scanners

Barkovich_Chap03.indd 142 5/6/2011 12:08:38 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 143

(3 T), the sign will appear on both T2-weighted spin echo and on FLAIR
images. Foci of T2 hyperintensity of variable size are typically present
within the area of T2 hypointensity (Fig. 3-49); these foci presumably
represent pallidal destruction and gliosis (558). This imaging sign, called
the “eye of the tiger sign” seems to be much more common in those cases
with PANK2 mutations (555) (but it is not always present (561,562) and
one report describes this characteristic sign disappearing during the
course of the disease (563) ). Patients without PANK2 mutations (i.e.,
those having neurodegeneration with brain iron accumulation (554) )
also have T2 hypointensity in the globi pallidi but are less likely to have
the central T2 hyperintensity (562). Although oculodigital dental dys-
plasia (564) has been shown to have similar globus pallidus changes to
those of PKAN, the associated white matter changes in that disorder and
the other obvious physical manifestations allow easy distinction of the
disorders. Therefore, the finding of T2 shortening in the globi pallidi during
the second decade should raise strong suspicion for the diagnosis of PKAN
rather than one of the many other disorders that affect the basal ganglia
FIG. 3-49. Pantothenate kinase-associated neuropathy in a 14-year-old in children (Tables 3-9 to 3-11). Diffusion imaging shows increased FA
with dystonia. Axial T2-weighted image shows the classic “eye of the tiger” and mean diffusivity in the globi pallidi and substantia nigra of affected
sign with T2 hyperintensity central to T2 hypointensity in the globi pallidi. patients compared with age-matched controls (565).

TABLE 3-9 Childhood Diseases Involving the Deep Cerebral Nuclei


Acute Ethylmalonic acidemia L-2-hydroxyglutaric aciduria
Hypoxia Propionic academia Maple syrup urine disease
Hypoglycemia Succinic semialdehyde dehydrogenase Wilson disease
Toxins (carbon monoxide, cyanide) deficiency 3-Hydroxyisobutyric aciduria
Hemolytic-uremic syndrome Guanidinoacetate methyltransferase Hallervorden-Spatz disease
Osmotic myelinolysis deficiency Dentatorubral and pallidoluysian atrophy
Encephalitis Biotin dependent encephalopathy Degenerative Diseases
Parainfectious Encephalomyelitis Isovaleric acidemia Juvenile Huntington disease
Tegretol Toxicity Molybdenum co-factor deficiency Sequelae of acute insults
Chronic Mitochondrial ATP synthetase deficiency Basal ganglia calcification syndromes
Inborn errors of metabolism 3-Methylglutaconic aciduria (i.e., Cockayne syndrome, Fried syndrome)
Mitochondrial disorders Malonic acidemia Other Diseases
Canavan disease Alpha ketoglutaric aciduria Neurofibromatosis type 1
Glutaric aciduria types I and II 3-Ketothiolase deficiency Chronic liver disease of any cause
Methylmalonic acidemia Biotinidase deficiencies Aicardi-Goutières syndrome

TABLE 3-10 Disorders Resulting in Deep Cerebral Nuclear Calcification


Endocrine Organic acidurias Measles
Hypoparathyroidism Congenital or Developmental Chicken pox
Pseudohypoparathyroidism Familial idiopathic symmetric basal Pertussis
Pseudopseudohypoparathyroidism ganglia calcification Coxsackie B virus
Hyperparathyroidism Hastings-James syndrome Cysticercosis
Hypothyroidism Cockayne syndrome Systemic lupus erythematosus
Metabolic Lipoid proteinosis (hyalinosis cutis [568]) Acquired immunodeficiency syndrome
Mitochondrial disorders Neurofibromatosis Toxic
Aicardi-Goutières syndrome Tuberous sclerosis Hypoxia
Fahr disease (familial cerebrovascular Oculocraniosomatic disease Cardiovascular event
ferrocalcinosis) Methemoglobinopathy Carbon monoxide intoxication
Pantothenate kinase associated neuropathy Down syndrome Lead intoxication
Carbonic anhydrase deficiency type II Inflammatory Radiation therapy
Wernicke encephalopathy Toxoplasmosis Methotrexate therapy
Griscelli disease (566) Congenital rubella Nephrotic syndrome
Fried syndrome (567) Cytomegalovirus

Barkovich_Chap03.indd 143 5/6/2011 12:08:39 AM


144 Pediatric Neuroimaging

TABLE 3-11 Anatomic Distribution of Some Diseases Affecting Basal Ganglia


Diagnosis Globus Pallidus Caudate Putamen White Matter
Acute Disorders
Hypoxia/ischemia-neonate + − + ±
Hypoxia/ischemia-older child + ++ ++ +
Hypoglycemia-neonate ± − − ++
Hypoglycemia-older child + + + +
Cyanide intoxication ++ + + −
Carbon monoxide intoxication ++ + + +
Hemolytic-uremic syndrome + + +
Osmotic myelinolysis + + + ++ pons
Viral Encephalitis + + + +
Chronic Disorders (with Possible Acute
Presentation During Metabolic Decompensation)
Leigh syndrome + + ++ +
Canavan disease ++ − − ++
GM2 Gangliosidoses − ++ − +
Juvenile Huntington Dz. + ++ ++ +
Wilson disease ++ + ++ +
Fried syndrome + ++ +
Glutaric aciduria type I − ++ ++ ++
Glutaric aciduria type II − + + +
Ethylmalonic encephalopathy + ++ ++ −
Molybdenum co-factor def. − ++ ++ +
Methylmalonic acidemia + − − +
Pantothenate kinase associated
neurodegeneration (Hallervorden-Spatz disease) ++ − − −
Propionic acidemia − ++ ++ +
Succinic semialdehyde dehydrogenase deficiency ++ − − −
Guanidinoacetate methyltransferase deficiency ++ − − −
Isovaleric acidemia ++ − − −
L-2-hydroxyglutaric aciduria ++ + + ++
Chronic liver disease ++ − − −
Kearns-Sayre syndrome ++ − − ++
Pyruvate dehydrogenase (E2) deficiency ++ − − −
3-ketothiolase deficiency − − ++ +
Alpha ketoglutaric aciduria + + ++ −
3-methylglutaconic aciduria (type 1 and 4) ++ ++ ++ +
HMG coenzyme A lyase deficiency − + + ++
Aicardi-Goutieres syndrome + + ++ ++
3-Hydroxyisobutyric aciduria + ++ ++ −
Dentatorubropallidoluysian atrophy ± − − +
++, characteristic lesion pattern element; +, occasional lesion pattern element; −, lack of involvement.

Barkovich_Chap03.indd 144 5/6/2011 12:08:39 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 145

FIG. 3-50. Juvenile Huntington disease in a


7-year-old. Axial T2-weighted (A) and FLAIR
(B) images show small, hyperintense caudate
heads and putamina. Hyperintensity of the
caudates is more easily seen on the FLAIR
image. Early in the course of the disease, the
hyperintensity might be the only abnormality.

Juvenile Huntington Disease Isovaleric Acidemia


Huntington disease (OMIM 143100) is a chronic progressive degenera- Isovaleric acidemia (also known as isovaleric acid CoA dehydrogenase
tive disease characterized by choreiform movements, mental deteriora- deficiency, OMIM 243500) is an autosomal recessive disorder caused by
tion, and an autosomal dominant transmission. The genetic defect is an mutation of the isovaleryl CoA dehydrogenase gene (IVD), located at
expanded, unstable segment of DNA with a repeating CAG segment in chromosome 15q14-15 (578,579). Two clinical forms, possibly allelic,
the first exon of the HTT gene on chromosome 4p16.3 (569). The CAG are recognized: the acute neonatal form, leading to massive metabolic
stretch on the gene is unstable when transmitted to the next generation, acidosis characterized by vomiting, dehydration, and restlessness from
with a propensity toward further expansion, particularly during sper- the first days of life and rapid death, and a chronic form in which peri-
matogenesis (570). Larger segments of CAG repeats are associated with odic attacks of severe ketoacidosis (typically provoked by infection)
earlier onset; juvenile Huntington disease is usually associated with 80 occur with asymptomatic intervening periods (580). The abnormali-
to 100 CAG repeats (571). Less than 6% of patients present prior to ties result from an accumulation of isovaleric acid, which is toxic to the
the age of 14 years (572). The clinical presentation in children varies central nervous system.
with the age of presentation. Children presenting before the age of 10 Limited reports suggest that neuroimaging in the chronic form
years most frequently manifest cognitive decline and seizures, whereas reveals low attenuation on CT scans with both T1 hypointensity and
those presenting at age 10 years or older most commonly present with T2/FLAIR hyperintensity on MRI in the bilateral globi pallidi (581).
oropharyngeal dysfunction (573). Chorea was not a presenting sign
but developed later in the course of the disease and, with dystonia, was
more prevalent in the early age at onset group, whereas rigidity and Succinic Semialdehyde Dehydrogenase Deficiency
bradykinesia were more prevalent in the older age at onset group (573). Also known as 4-hydroxybutyric aciduria, succinate semialdehyde
Patients in both groups developed gait, cognitive, and behavioral dis- dehydrogenase deficiency (OMIM 271980) is a rare autosomal reces-
orders at some point during the course of the disease. Furthermore, a sive disorder in the degradation pathway of gamma-amino butyric
slow and steady decline in IQ was observed on serial neuropsychologic acid (GABA) (582); the causative gene ALDH5A1 has been mapped
testing in patients from both groups (573). Presentation with progres- to chromosome 6p22 (583,584). The result is an increase in GABA
sive myoclonus epilepsy has been described (574). in the CSF and of 4-hydroxybutyric acid in the urine. Affected
Imaging studies are normal early in the course of the disease. As patients manifest a slowly progressive encephalopathy character-
the disease progresses, the striatum atrophies and develops T2 hyperin- ized by delayed motor function, delayed to absent language develop-
tensity; atrophy of the caudate heads results in a characteristic enlarge- ment, and mental retardation. These are variably accompanied by
ment of the frontal horns of the lateral ventricles, which become ataxia, seizures, hypotonia, behavioral problems, autistic features,
convex laterally (Fig. 3-50) (572). Atrophy in the putamen, seen bet- hallucinations, and choreoathetosis (582,585,586). Rarely, presen-
ter by MRI than by CT, is equal to or more severe that in the caudate tation may be in infancy with hypotonia, psychomotor delay, and
(573,575). Cortical atrophy is most prominent in the frontal lobes. As strabismus (587). EEG shows generalized and focal epileptiform dis-
in many other metabolic diseases, F-18 fluorodeoxyglucose PET scan- charges, photosensitivity, and background slowing (586). MRI char-
ning is sensitive to decreased glucose uptake in the striatum before CT acteristically shows T2 prolongation in the globi pallidi (Fig. 3-51A)
or MR scans become positive (576). Comparisons of the sensitivity of and, sometimes, dentate nuclei of the cerebellum (585,588). With
PET to that of MRI have not been published. A single report of proton disease progression, diffuse cerebral cortical, basal ganglia, and thal-
MRS found reduced NAA and increased myo-I on short echo (35 ms) amic atrophy ensues (589). MRS shows normal choline, creatine,
spectra in a voxel including the caudate head and anterior putamen; and NAA (Fig. 3-51B). It will be interesting to see whether MRS
the patient already had substantial atrophy of the corpus striatum at sequences looking at GABA, glutamate, and glutamine show any
the time the spectrum was obtained (577). abnormalities.

Barkovich_Chap03.indd 145 5/6/2011 12:08:39 AM


146 Pediatric Neuroimaging

FIG. 3-51. Succinate semialdehyde dehydrogenase deficiency in a 2-year-old with language delay. Axial T2-weighted image (A) shows bilateral T2
hyperintensity (white arrows) of the globi pallidi. Proton MR spectrum (TE = 288 ms) of the basal ganglia (B) is normal.

Creatine Deficiency Syndromes with reduced diffusivity of the globi pallidi (599) and mild myelina-
MR spectroscopy has allowed the discovery of several syndromes in tion delay or T2/FLAIR hyperintensity of the cerebral white matter
which the concentration of creatine is reduced within the brain. The (Fig. 3-52A) (590,592–594,598).
first of these syndromes to be described was guanidinoacetate methyl- The X-linked mental retardation syndrome (OMIM 300352) is
transferase (GAMT) deficiency (OMIM 612736), an autosomal reces- another creatine deficiency syndrome in which MRS shows absence of
sive inborn error of creatine synthesis (590) caused by mutations of or severe reduction of the creatine peak in developmentally impaired
the GAMT gene at chromosome 19p13.3. The other creatine deficiency children (600,601). It is caused by a defect in formation of a creatine
syndrome is an autosomal recessive disorder caused by a deficiency of transporter protein due to mutations of the SLC6A8 gene at chro-
L-arginine:glycine amidinotransferase (AGAT, OMIM 612718), another
mosome Xq28 (601). Affected children present with mild to moder-
enzyme important in creatine biosynthesis (591), caused by mutation ate global developmental delay and severe developmental language
of the GATM gene at chromosome 15q15.3. Creatine and creatine impairment (602); hypotonia and seizures may be present (603).
phosphate are essential for the storage and transmission of phosphate- Mutations of SLC6A8 have been found in up to 1% of patients with
bound energy in muscle and brain. They are catabolized to creatinine. mental retardation (604) and those with autistic spectrum disorders
For maintenance of the body pool of creatine, the daily urinary loss (605). This disorder does not respond to creatine supplementation. It
of creatinine must be balanced by endogenous synthesis and dietary can be separated from GAMT deficiency and AGAT deficiency by the
intake of creatine (592). GAMT and AGAT deficiencies reduce creatine elevated plasma and urine creatine levels and normal guanidinoacetate
synthesis. In both disorders, the severe depletion of creatine/phospho- levels (601).
creatine causes developmental delay, followed by regression, muscle
Wernicke Encephalopathy
hypotonia, extrapyramidal movement abnormalities, and intractable
epilepsy (590–595). GAMT deficiency is also characterized by severe Wernicke encephalopathy (OMIM 277730, also called Gayet-Wernicke
expressive language delay (596) and hyperactivity (593). These symp- encephalopathy and alcohol-induced encephalopathy) is a degenera-
toms can be partially or completely reversed by dietary creatine supple- tive condition caused by a chronic deficiency of thiamine (vitamin B1).
mentation (597,598), so it is crucial to make the diagnosis in affected Although a classical clinical triad of oculomotor abnormalities, men-
patients. Indeed, we have seen a patient with AGAT deficiency improve tal status changes, and ataxia has been described, few patients present
remarkably, initially manifesting moderate to severe delay but improv- with the full triad. In pediatric patients, the most common presenta-
ing to well above the mean 18 months after beginning creatine supple- tion, seen in more than 80%, is one of altered mental status (confusion,
mentation. Supplementation may work better in AGAT than in GAMT somnolence, stupor, or coma) (606). Ocular signs (ophthalmoplegia or
deficiency (597). nystagmus) are present in about two thirds of patients at presentation,
The combination of MRI and, particularly, proton MR spectros- but ataxia is uncommon (~20%) (606). Affected patients are typically
copy can be used to strongly suggest this diagnosis. MRS shows a mark- malnourished, most commonly from malignancy, gastrointestinal dis-
edly reduced or completely absent creatine peak; this is most easily ease, or food allergy. A recent history of carbohydrate loading or par-
seen on long TE spectra (Fig. 3-52). In addition, short TE sequences enteral nutrition is often obtained (606).
show a broad guanidinoacetate peak at 3.78 parts per million in chil- Pathologic examination shows symmetric hemorrhage secondary
dren with GAMT deficiencies. The creatine peak will slowly reappear to proliferation and dilation of capillaries in the gray matter around
and the other peaks disappear after creatine supplementation is insti- the third ventricle, sylvian aqueduct, and fourth ventricle. In adults, the
tuted. MRI is typically normal in patients with AGAT deficiency, but most common MRI findings are T2/FLAIR hyperintensity and atrophy
in many cases of GAMT deficiency shows bilateral T2 prolongation of the medial thalami and mamillary bodies, with the periaqueductal

Barkovich_Chap03.indd 146 5/6/2011 12:08:39 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 147

FIG. 3-52. Guanidinoacetate methyltransferase deficiency in an


8-month-old child. A. Axial T2-weighted image shows bilateral T2
prolongation (white arrows) of the globi pallidi. B. Single voxel
proton MRS (TE = 288 ms) of the basal ganglia shows normal
choline (Cho) and N-acetylaspartate (NAA) peaks but absence of
the creatine peak. C. Single voxel proton MRS (TE = 26 ms) of the
basal ganglia also shows absence of the creatine peak. The find-
ing is more subtle on the short echo time scan because of the less
stable baseline and the increased number of peaks. An abnormal
broad guanidinoacetate peak is present at 3.78 ppm.

area involved in about two thirds; about half show enhancement after pontine myelinolysis (612). Affected children may manifest a variety
administration of intravenous contrast (540,607–609). Increased dif- of neurologic signs and symptoms, including akinesis, ataxia, catato-
fusivity is seen in the affected areas (610). The few reported cases of nia, choreoathetosis, rigidity, dysarthria, dystonia, emotional lability,
pediatric Wernicke encephalopathy show involvement (low attenuation mutism, myoclonus, myokymia, parkinsonism, and tremor (612). The
on CT, T2/FLAIR hyperintensity on MRI) of the medial thalami (most imaging abnormality consists of T2 prolongation in the claustrum and
common finding, Fig. 3-53), putamina, caudate nuclei, and frontotem- putamen in children (Fig. 3-54), with the hippocampus sometimes
poral cerebral cortex (606,608,611). Rapid atrophy may ensue (606). being involved (613); diffusion is reduced in the affected area(s) (526).
Extrapontine myelinolysis may occur with or without associated cen-
Extrapontine Myelinolysis tral pontine myelinolysis (612).
The term extrapontine myelinolysis refers to osmotic myelinolysis in
regions other than the pons. Patients become symptomatic after rapid Hemolytic-Uremic Syndrome
shifts in fluids, usually the result of overly rapid treatment of hypona- The hemolytic-uremic syndrome (HUS) is a multisystemic dis-
tremia; the causes are almost certainly similar to those of central order, characterized by renal failure, thrombocytopenia, and a

Barkovich_Chap03.indd 147 5/6/2011 12:08:40 AM


148 Pediatric Neuroimaging

consequence of infection by Gram-negative organisms (most com-


monly verotoxin-producing E. coli). Histological studies have shown
that verotoxin produced by the bacteria binds to the endothelium of
small vessels that leads to thrombosis of vessels in affected organs
(616,617). The mutation of several genes that regulate the comple-
ment alternative pathway causes a variant of the disease called atypi-
cal hemolytic-uremic syndrome (OMIM 235400) (618), in which the
prodrome of enterocolitis and diarrhea is absent. Young children and
adolescents are principally affected; most patients are less than 5 years
old. Pathologic studies have found edema, hemorrhage, and isch-
emic injury in the basal ganglia, thalami, hippocampi, and cerebral
cortex.
On imaging studies, the deep cerebral nuclei are most commonly
affected, but the cerebral cortex is involved in severe cases. CT shows
abnormal attenuation (low or high) in the basal ganglia; cortical
hypointensity may be present. FLAIR and T2-weighted MR images
show hyperintensity in the affected areas of the deep gray cerebral
nuclei, particularly thalami, putamina, brainstem, cerebellar vermis,
and cerebral cortex (Fig. 3-55) (619,620). The adjacent white matter,
particularly the internal and external capsules in the case of putami-
nal involvement and the subcortical white matter in cases of cortical
involvement, may be involved (Fig. 3-55B) (617,620,621). T1-weighted
MR images may show hyperintensity in the basal ganglia (Fig. 3-55),
FIG. 3-53. Pediatric Wernicke encephalopathy in an anorexic 12-year-old.
resulting from coagulative necrosis secondary to microthrombosis
Axial T2-weighted image shows abnormal T2 hyperintensity (white arrows)
of the medial thalami. without hemorrhage or calcification (617). These areas of T1 hyperin-
tensity may persist on follow-up imaging (Fig. 3-55E) (620) and may
continue to enhance for many weeks (Fig. 3-55F). When the cerebral
microangiopathic hemolytic anemia that follow a prodrome of acute cortex is involved, similar signal characteristics are seen (617). Dif-
afebrile gastroenteritis, often with bloody stools. The central nervous fusion images show reduced diffusivity in affected areas in the acute
system is affected in 20% to 50% of patients; altered mental status, phase and increased diffusivity in the subacute and chronic stages of
personality changes, seizures, long tract signs, and loss of vision are the disorder (Fig. 3-55C) (620).
the most common manifestations (614,615). HUS is thought to be a
Sydenham Chorea
Sydenham chorea, also called chorea minor, St. Vitus dance, or rheu-
matic encephalitis, is a presumed autoimmune phenomenon that results
from streptococcal infections. Most common in children between age
of 7 and 12 years, it may be isolated (20%–30%) or be associated with
other manifestations of rheumatic fever (carditis, migratory polyar-
thritis, subcutaneous nodules) (622,623). Affected patients develop a
hypotonic, hyperkinetic syndrome that is characterized clinically by
involuntary and uncoordinated movements, muscular weakness, fre-
quent falls, dysarthria, difficulty in concentrating and writing, slurred
speech, and emotional lability. In most patients, the symptoms resolve
within 2 years, but recurrences may occur. Psychiatric problems, such
as poor attention span (ADHD), aggressive behavior, obsessive-com-
pulsive disorder, anxiety, and depression may develop, as well (623).
Neuropathological studies have revealed degeneration of neurons and
alteration of blood vessels in the cerebral cortex, basal ganglia, and
cerebellum (624), as well as antibodies reactive against neurons of the
caudate nucleus (623).
Imaging findings may correlate with outcome. The basal ganglia
are by far the region in which abnormalities are most commonly rec-
ognized, in particular the caudate nuclei, putamina, and cerebral white
matter. Localized low attenuation is seen on CT, while T2 prolongation
is seen on MRI (625–627). These changes are rarely permanent; they
generally resolve within weeks or months (627). When MR changes are
permanent, they seem to be associated with prolonged clinical course
of disease and recurrent episodes of neurological impairment (625).
FIG. 3-54. Extrapontine myelinolysis in a 3-year-old girl. Axial Reduced volume of the basal ganglia can be detected on volumetric
T2-weighted image shows T2 prolongation in bilateral claustra and lateral studies, even in patients with apparently normal MRI studies (628).
putamina. These areas reverted to normal after slow correction of electro- Proton MR spectroscopy shows elevated choline/creatine and reduced
lyte abnormalities. NAA/creatine (629).

Barkovich_Chap03.indd 148 5/6/2011 12:08:40 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 149

FIG. 3-55. Hemolytic-uremic syndrome, early (A–C) and late (D–F) subacute phases. A and B. Axial FLAIR image shows abnormal hyperintensity in the dorsal
brain stem (white arrowheads), cerebellar vermis (white arrow), external/extreme capsules (black arrowheads), internal capsules (black arrows) and globi pallidi (g).
C. Axial diffusivity (Dav) maps shows increased Dav in the external/extreme capsules (black arrows) and the internal capsules (black arrowheads). D. Axial FLAIR
image 2 weeks later shows residual hyperintensity (white arrows) in the putamina. E and F. Axial pre- (E) and postcontrast (F) images show persistent T1 hyperin-
tensity (white arrows) in the posterior putamina and globi pallidi. Contrast enhancement in (F) shows that the blood–brain barrier injury has not fully healed.

Chronic Liver Disease have been identified (Table 3-12): infantile (Santavuori-Haltia disease,
As in adults, chronic liver disease causes T1 shortening in the globi CLN1), late infantile (a genetically heterogeneous group that includes
pallidi, cerebral peduncles, superior cerebellar peduncles, and pituitary classical Jansky-Bielschowsky (CLN2), variant (CLN6), Finnish vari-
gland. This appears to result from just about any cause of hepatic fail- ant (CLN5), and Turkish variant (CLN7) forms), juvenile (Batten or
ure including Crigler-Najjar disease type 2, congenital hepatic fibro- Spielmeyer-Vogt disease, CLN3), early juvenile (CLN9), adult domi-
sis, viral hepatitis, sclerosing cholangitis, Byler disease, biliary atresia, nant (Kufs disease, CLN4), adult recessive, progressive epilepsy with
Alagille syndrome, Wilson disease, or copper toxicosis (630). mental retardation (northern epilepsy syndrome, CLN8), and cathep-
sin D deficient (CLN10) types (631,632).
Gray Matter Disorders Primarily Clearly, the different types of NCL represent different disease enti-
Involving Cortex ties; they are grouped together because they have similar pathologic
Neuronal Ceroid Lipofuscinosis findings and similar clinical presentations. In addition, studies with
animal models suggest that these disorders are due to mutations of
The neuronal ceroid lipofuscinoses (NCL) are among the most com-
membrane proteins, and that at least some of the affected proteins are
mon progressive encephalopathies of childhood; they occur all over the
involved in synaptic vesicle function (641).
world with an incidence of about 1 in 25,000 live births. Ten main types

Barkovich_Chap03.indd 149 5/6/2011 12:08:41 AM


150 Pediatric Neuroimaging

TABLE 3-12 Classification of Neuronal Ceroid Lipofuscinosis


Age at Onset Designation Chromosome Location Gene Product
Congenital or later CLN10 11p15 (633) Cathepsin D
Infantile or later CLN1 (Classic) 1p32 (634) PPT1
Late infantile or later CLN2 (Classic) 11p15.5 (635) TPP1
CLN5 (Finnish) 13q22 (636) partially soluble protein
CLN6 (variant) 15q21 (637) Membrane protein
CLN7 (Turkish) 4q28 (638) Membrane protein
CLN8 8p23 (639) Membrane protein
Juvenile CLN3 (Classic) 16p12 (640) Membrane protein
CLN9 unknown unknown
Adult CLN4 (Kufs) unknown unknown
PPT1, palmitoylprotein thioesterase 1; TPP1, tripeptidylpeptidase 1

Clinical presentation depends upon the type of NCL and the age and cerebral atrophy seems to progress at similar rates. In the juvenile
of presentation (642,643). Infants with the congenital form are charac- form, MRI is typically normal before the age of 10 years and, therefore,
terized by primary microcephaly, neonatal (or prenatal) epilepsy, and is not useful for diagnosis. Variable atrophy and T2 hypointensity of
death in early infancy (644). Patients with infantile forms typically are the thalami may be seen after the age of 14 years (647,651).
brought to attention because of deceleration of head growth and mus- Proton MRS in the infantile form of the disease shows a complete
cular hypotonia after the age of 6 months. Ataxia and motor clumsiness loss of the NAA peak, marked reduction of creatine and choline, and
develop together with irritability, sleep disturbance, and visual failure elevation of myo-I and lactate in gray and white matter (650,655). In
leading to a rapid developmental deterioration during the second year the late infantile form, reduced NAA has been noted in gray and white
of life. In late infantile forms, the first symptoms begin around the matter; myo-I, creatine, and choline are elevated in white matter (Fig.
third year of life, typically an unexpected delay of psychomotor devel- 3-57D) (653,655). On short TE spectra, the level of myo-I elevation is
opment or epilepsy of sudden onset. Seizures are generalized tonic- reportedly concordant with the severity of disease (653). Lactate eleva-
clonic or partial, frequently of a severe myoclonic type; they may soon tion is reportedly seen in some patients (655). In juvenile NCL, spectra
become resistant to drug treatment. The first symptom of patients with are reportedly normal (655). However, the patients reported thus far
juvenile forms is usually failure of vision, followed after several years by were at different stages of their disease at the time their spectra were
progressive dementia, seizures, and progressive impairment of speech performed. Thus, the MRS differences may have reflected differences in
and motor function (645,646). Definitive diagnosis is made by genetic the stage of the disease (amount of brain destruction) at the time of the
testing or by electron microscopic analysis of lipopigments from the spectra more than differences in the diseases themselves.
skin, which shows characteristic ultrastructral characteristics (646). PET and SPECT may be useful in establishing diagnosis in NCL. 18
For neonates with microcephaly and epilepsy, testing for deficiency of FDG PET studies show severe reduction of metabolism in all the corti-
cathepsin D may be an economical approach to diagnosis. Similarly, in cal and subcortical structures. Regional analysis shows marked bilat-
young children with unexplained epilepsy and acute cessation of devel- eral hypometabolism, particularly in calcarine, lateral occipital, and
opment, analysis for PPT1 and TPP1 activity (see Table 3-12) has been temporal cortices and in thalami (656). In infantile NCL, SPECT using
recommended (643). 99m-Tc HMPAO showed bilateral anterior frontal, posterior tem-
Findings on imaging studies (Figs. 3-56 and 3-57) are nonspecific; poroparietal, and occipital hypoperfusion. Initially, hypoperfusion was
therefore, imaging is best used to rule malformations, infection, or other localized and symmetric, whereas atrophy on MRI was more general-
causes of encephalopathy. In the infantile form, the major findings are ized. Reduction in cerebellar perfusion and onset of cerebellar atrophy
variable cerebral and cerebellar atrophy associated with T2 hyperin- occurred later. Perfusion of deep gray matter nuclei is well preserved
tense rims around the ventricles (Fig. 3-56) and T2 hypointensity in up to terminal stage in spite of apparent atrophy seen on MRI (657).
the thalami and globi pallidi (80,647–650). The T2 changes appear to SPECT in juvenile NCL shows diffuse hypoperfusion, most severe in
correlate with loss of myelin and gliosis of unknown origin (651); thal- the temporal lobes, and less so in the parietal, occipital, and cerebellar
amic atrophy develops, suggesting that the thalamic T2 changes may regions. Interesting, epilepsy does not correlate well with involvement
be primary, rather than secondary. Cortical hypoperfusion is shown by of any region by SPECT (658).
SPECT (650). Atrophy of both cortex and deep structures appears to
develop more rapidly in the infantile form (Fig. 3-56) than in the late Aspartylglucosaminuria
infantile form and earlier in the late infantile form than in the juvenile Aspartylglucosaminuria (OMIM 208400) is a hereditary lysosomal
form (647,652). In the late infantile and progressive epilepsy with men- storage disorder caused by defective activity of the enzyme aspartyl gly-
tal retardation forms, cerebellar atrophy is an early finding (Fig. 3-57; cosiminidase in various body tissues and fluids (659). The AGU gene
indeed, these patients often present with motor delay secondary to cer- on chromosome 4q32-33 has been cloned and several mutations caus-
ebellar atrophy (632,653,654) ), whereas in the other forms, cerebellar ing aspartylglucosaminuria have been identified (660). Characteristic

Barkovich_Chap03.indd 150 5/6/2011 12:08:42 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 151

FIG. 3-56. Infantile neuronal ceroid lipofuscinosis; sequential imaging studies showing progressive atrophy. A. Sagittal T1-weighted image at age 8 months
shows slightly thin corpus callosum but is otherwise normal. B. Axial FLAIR image shows mild enlargement of the frontal horns. C and D. Follow-up sagittal
T1-weighted and axial FLAIR images 1 month later shows larger cerebellar vermian fissures and thinning of the corpus callosum (C) with enlarging ventricles
and cortical sulci (D), indicating interval volume loss. Rims of high signal have developed around the lateral ventricles. E and F. Six months later, the sagittal
T1-weighted image (E) shows marked thinning of the corpus callosum with volume loss of the brain stem and cerebellar vermis, while the axial FLAIR image
(F) shows marked enlargement of ventricles and sulci. (This case courtesy Dr. Lara Brandao, Rio de Janeiro, Brazil.)

Barkovich_Chap03.indd 151 5/6/2011 12:08:42 AM


152 Pediatric Neuroimaging

FIG. 3-57. Late infantile neuronal ceroid lipofuscinosis, advanced disease. A. Sagittal T1-weighted image shows thinning of the corpus callosum and mod-
erate to severe cerebellar atrophy. B. Axial T2-weighted image shows large cerebral ventricles and sulci and slight diffuse hyperintensity of the cerebral white
matter. C. Coronal T1-weighted reformation from volumetric data set shows marked prominence of CSF spaces and profound cerebellar atrophy. D. Single
voxel proton MR spectroscopy (TE = 25 ms) of the frontal white matter shows a small NAA peak and an abnormally large myo-inositol peak (arrow).

neurological manifestations are mildly dysmorphic facial features, abnormality may be reversed by bone marrow transplant (664). As the
slowly progressive mental retardation, delayed speech, hyperkine- patients mature, cortical–white matter differentiation improves, but
sia, motor clumsiness, and slight truncal ataxia (660–662). A mental cortical atrophy develops, manifested as shrunken gyri and enlarged
decline typically begins during the second decade of life, and death sulci (661).
generally occurs during the fourth decade.
MRI studies of the brain show bilateral T2 hypointensity and T1 Anti–N-Methyl-d-Aspartate Receptor (NMDAR) Encephalitis in
hyperintensity in the posterior thalami (pulvinar region) (663) and Children and Adolescents
poor differentiation between cortex and underlying white matter that Anti–N-methyl-D-aspartate receptor (NMDAR) encephalitis is a
appears to result from increased signal intensity in the subcortical white recently described disorder with a well-defined set of clinical features
matter on T2-weighted images (661). It seems that this white matter (19,665). Initially described in young women with teratomas of the

Barkovich_Chap03.indd 152 5/6/2011 12:08:44 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 153

FIG. 3-58. N-methyl-D-aspartate receptor (NMDAR) encephalitis. A. Coronal


FLAIR image shows subtle hyperintensity (white arrows) of the right frontal cortex,
insula, and hippocampus when compared to the left hemisphere. B. Image from
pelvic ultrasound shows a hypoechogenic mass (white arrows) with a solid central
component (white arrowhead). C. Coronal postcontrast CT reformation shows the
low attenuation right pelvic mass (black arrows) with central calcification (white
arrowhead). (These images courtesy Dr. Ken Martin, Oakland.)

ovary who developed changes of mood, behavior, and personality, Neuroimaging may be normal or may show subtle T2/FLAIR
resembling acute psychosis, it was soon found that the disorder also hyperintensity in the hippocampi, cerebral cortex, or cerebellum (Fig.
develops in girls and women without teratomas, as well as in children 3-58); these areas may show reduced diffusivity if diffusion-weighted
and men. This disorder is now recognized as a surprisingly common imaging is performed early in the course of the disease. Enhancement
cause of a syndrome of neuropsychiatric symptoms (agitation, para- of the leptomeninges or multifocal parenchymal enhancement may
noia, disinhibition, focal seizures, speech dysfunction), movement be detected after intravenous injection of paramagnetic contrast,
abnormalities (orofacial dyskinesias, dystonia, choreoathetoid, rigid- depending upon the stage of the disease at the time of imaging (19).
ity), and autonomic dysfunction (hypertension, tachycardia, hyper- Prolonged disease without therapy results in atrophy. Results of pro-
tension, hyperthermia, sleep dysfunction, dilated pupils) in children ton MR spectroscopy have not been reported. If a pelvic teratoma is
(19,666). Most affected patients are initially diagnosed as having viral present, it will usually appear as a cystic and solid mass with calcifica-
meningitis. The diagnosis is made by CSF analysis and detection of the tion (Fig. 3-58B and C).
NR1 subunit of the NMDAR. Current recommendations are screening
of the abdomen and pelvis with ultrasound and MRI to look for a tera- Infantile Neuroaxonal Dystrophy
toma for at least 2 years (19). Treatment is by removal of the teratoma Patients with the infantile form of neuroaxonal dystrophy (INAD,
(if present) or immunotherapy (665,667). OMIM 256600, also called Seitelberger disease) usually present during

Barkovich_Chap03.indd 153 5/6/2011 12:08:45 AM


154 Pediatric Neuroimaging

FIG. 3-59. Infantile neuroaxonal dystrophy. A. Sagittal T1-weighted image shows moderate cerebellar cortical atrophy, with shrunken folia and enlarged
fissures. B. Coronal T2-weighted image shows hyperintensity of the shrunken cerebellar cortex and enlarged cerebellar fissures (white arrows). (These
images courtesy Dr. Susan Blaser, Toronto.)

the end of the first year or the second year of life; the disease progresses Niemann-Pick Disease
throughout childhood with death ensuing late in the first decade or Niemann-Pick disease is an autosomal recessive disorder that is charac-
early in the second decade (668). Most cases of the disorder appear to terized by progressive hepatosplenomegaly, lymphadenopathy, edema,
be caused by mutations of the PLA2G6 gene at chromosome 22q13.1 and, in some patients, neurologic degeneration. The underlying prob-
that encodes a calcium-independent phospholipase A2 enzyme that lem is a deficiency of sphingomyelinase; as a result of the deficiency,
catalyzes the hydrolysis of glycerophospholipids (669). Affected lipids (sphingomyelin, cholesterol, and lysobisphosphatidic acid)
patients develop regression of cognition and motor skills, followed accumulate throughout the reticuloendothelial system and, in some
by nystagmus and progressive deterioration of posture, gait, speech forms of the disease, in the brain. Six clinical forms (A through F) of
(if developed), and visual acuity; nystagmus and strabismus are com- the disease are described. Three of these forms, A (OMIM 257200),
mon (668,670). About 30% present following an intercurrent illness C1 (OMIM 257220), and C2 (OMIM 607625) have neurologic mani-
(668). Physical exam reveals axial hypotonia, four limb spasticity and festations, which consist primarily of loss of motor and intellectual
bulbar signs. Patients eventually develop ataxia, dystonia, optic atrophy function. Onset in early infancy and rapid deterioration are charac-
and blindness, muscular atrophy, severe dementia, and complete lack teristic of Type A variant, which is caused by mutations of the SMPD1
of motor function. Epilepsy develops in some patients (668,670,671). gene at chromosome 11p15.1-4 (674). Onset in early childhood with
An atypical form of neuroaxonal dystrophy, also caused by mutations slower progression is seen in Type C1 (242), caused by mutation of the
of PLA2G6, has been described with later age of onset (average 4.4 NPC1 gene at chromosome 18q11-12 (675) and Type C2, caused by
years) and slower progression; details of this form of the disorder are mutation of NPC2 at 14q24.3, which have similar phenotypes (676).
still being elucidated (670). Some authors consider INAD to be part of These children come to attention in the latter half of the first decade
a group of disorders called neurodegeneration with brain iron accu- due to clumsiness and developing ataxia; as the disease progresses they
mulation, along with pantothenate kinase associated degeneration develop dysarthria, dysphagia, dystonia, dementia, and seizures (676).
(discussed above in section IV.C.1.a), aceruloplasminemia, and neuro- Imaging findings (Fig. 3-60) reflect the pathologic descriptions of
ferritinopathy (670). nonspecific progressive gray matter atrophy. The cortical sulci and ven-
Pathology shows atrophy of the cerebellar and cerebral cortices, tricles are enlarged. The corpus callosum is thin. The underlying white
with marked astrogliosis, extensive axonal swelling and spheroids in matter shows diffuse, hazy T2 hyperintensity and, to a lesser extent,
the axoplasm of affected regions (671). MRI shows prominent sulci T1 hypointensity. Proton MRS shows progressive diminution in the
in the cerebral and cerebellar cortices, diminished cerebral white mat- amount of NAA.
ter, and marked T2 prolongation of the cerebellar cortex (Fig. 3-59).
Sequential scans will show progressive cerebellar atrophy in most Rett Syndrome
patients (668,672). Some patients show thinning of the optic nerves Rett syndrome (OMIM 312750 (677) ) was established as an inter-
and chiasm (672). Infants with mutations of the PLA2G6 gene often nationally defined syndrome in 1983 (678). It is a progressive neu-
show T2 hypointensity (corresponding to iron deposition) in the globi rodevelopmental disorder that occurs sporadically, with prevalence
pallidi (668); this is most notable in children with the atypical form of in girls reported at about 1/10,000. Recent work has shown that the
the disease. Proton MRS shows progressive loss of NAA and slightly disorder is caused by mutation of the MECP2 gene on chromosome
elevated lactate in the basal ganglia (673). Xq28 (679,680). Although the mutation is usually fatal in boys, some

Barkovich_Chap03.indd 154 5/6/2011 12:08:46 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 155

FIG. 3-60. Niemann-Pick disease,


type C in an 18-year-old. Axial
T2-weighted images (A and B) show
enlarged ventricles and subarachnoid
spaces and diffuse haziness of the cere-
bral white matter. These are character-
istic findings of a gray matter disease
involving primarily cortex.

boys have been reported to have Rett syndrome as a result of somatic with acute neurological symptoms and bilateral, symmetric gray mat-
mosaicism, a condition in which only some cells have the mutated gene ter involvement. Carbon monoxide poisoning (Fig. 3-61), cyanide
(681), or an extra X chromosome (682). poisoning (Fig. 3-62), intoxication with drugs such as ecstasy (695),
Typically, affected patients have a normal prenatal and perinatal and exposure to organic solvents (540,695) can all result in damage
history but have onset of progressive deterioration that begins in the to the deep gray matter (CO to the globi pallidi and substantia nigra
latter half of the first year of life. This is usually manifested as regres- (696), cyanide to the globi pallidi and striatum) and, sometimes, to
sion of purposeful hand use, cognitive functions, acquired language, the cerebral or cerebellar (particularly with cyanide, Fig. 3-62) cortex
and social skills. Decline in growth rate of the head begins at age 2 (540,697,698). Delayed injury may develop in the cerebral white mat-
to 4 months, dropping to less than 3rd percentile by age 4 years. By ter (“postanoxic encephalopathy”) by mechanisms that are unclear
age 3, profound intellectual disability is evident and patients develop (699,700). Most of these injuries will show reduced diffusion if MRI is
stereotyped repetitive hand movements with fixed position of the performed in the acute phase (696).
hands. Subsequently, intellectual disability remains relatively static
while motor functions deteriorate (680). Seizures develop in up to 80% Progressive Cerebral Poliodystrophy (Alpers Disease)
of affected patients (683,684). Pathology shows global decrease in the Alpers disease is discussed in the Mitochondrial Disorders section.
size of neurons with increased packing density, decreased melanin in
the large neurons of the pars compacta of the substantia nigra (685). Trichopoliodystrophy (Menkes Disease)
Rett syndrome is extremely uncommon in boys. When boys are Menkes disease is discussed in the Mitochondrial Disorders section .
affected, they may have a typical presentation of a Rett Syndrome phe-
notype (seen with somatic mosaicism (686) or with X-inactivation in
an XXY karyotype (682,687,688) ). Alternatively, they may have severe Metabolic Disorders That Affect Both Gray And
neonatal encephalopathy (687,689) or severe mental retardation with White Matter
progressive spasticity (690). Autopsy of one patient with a small dele-
tion in MECP2 who presented with neonatal encephalopathy showed Canavan Disease (Aspartoacylase Deficiency,
bilateral perisylvian polymicrogyria (691). Spongiform Leukodystrophy)
Although qualitative neuroimaging studies of girls with Rett Syn- Canavan disease (OMIM 271900) is an autosomal recessive disor-
drome are usually normal or show mild diffuse atrophy, quantitative der, most common in Ashkenazi Jews, that is caused by a deficiency
neuroimaging studies show global reduction in gray and white matter of aspartoacylase. The biochemical disorder results in accumulation
volumes, with the largest reductions in the prefrontal, posterior frontal, of NAA in the brain (701) and N-acetyl aspartic aciduria (702). The
and anterior temporal cortices and the caudate nucleus. White mat- ASPA gene has been cloned and localized to the short arm of chro-
ter volumes are reduced uniformly through the brain (692). Proton mosome 17 (17pter-p13) (703). In rodents, aspartoacylase activity is
MRS shows decreased NAA in older patients, more pronounced in gray expressed only in oligodendrocytes (704), where it catalyzes the hydro-
matter than in white, with reduction of the glutamate/glutamine ratio lysis of neuronally derived NAA to acetate and aspartic acid, so it may
(693). Frontal and parietal lobes and insular cortex seem predomi- be postulated that deficiency of the enzyme results in accumulation of
nantly involved (694). NAA in oligodendrocytes with consequent intramyelinic edema, vac-
uolization, and oligodendrocyte failure in humans as well. However,
Toxins the biochemistry of NAA is complex (701) and is still being elucidated
Certain toxins affect primarily gray matter. Therefore, toxic exposure (705). It has been suggested that myelin damage in Canavan disease is
should always be considered in children, particularly adolescents, related to a profound impairment of water homeostasis of brain result-

Barkovich_Chap03.indd 155 5/6/2011 12:08:46 AM


156 Pediatric Neuroimaging

FIG. 3-61. Carbon monoxide poisoning. A. Axial T2-weighted image shows hyperintensity in bilateral globi pallidi (large white arrows) and occipital
cortex (small white arrows). B. Coronal FLAIR image shows swelling and hyperintensity (arrows) of the globi pallidi and hippocampi.

ing in fluid imbalance between intracellular (axon-glial) and extracel- of the sealed interlamellar spaces and, hence, intramyelinic edema,
lular (interlamellar) spaces within myelinated white matter. In brain, with resultant demyelination and loss of glial cells (706).
NAA is synthesized in neurons and it is one of the most abundant low In the first few weeks of life, affected infants may show marked
molecular-weight parenchymal cellular metabolites, believed to have hypotonia, with severe head lag. Soon, they may develop macroceph-
a role in the molecular efflux water pump system. Indeed, NAA may aly and seizures. Psychomotor development is delayed. With further
function as a water transporter and since the synthesis of NAA is intact progression of the disease, spasticity, intellectual failure, and optic
in Canavan disease, “overproduction” of NAA within neurons may lead atrophy develop. Death usually occurs in the second year of life (707).
to increased water migration from the axon into the periaxonal space. Some patients have a more protracted clinical course with less severe
Normally, hydrolysis of NAA takes place in this space by aspartoacy- impairment of psychomotor development (708,709); indeed, some
lase. If NAA is not eliminated, excess water builds up in the periaxonal may have developmental delay and problems with social interaction
space, leading to increased osmolar pressure, probably causing rupture but a normal neurologic exam (710). Presumably the mutations of

FIG. 3-62. Cyanide poisoning. Axial T2-


weighted image (A) shows abnormal hyper-
intensity (white arrows) of the globi pallidi.
Coronal FLAIR image (B) shows diffuse severe
swelling and hyperintensity of the cerebellar
cortex.

Barkovich_Chap03.indd 156 5/6/2011 12:08:47 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 157

ASPA in these children have a less pronounced effect on the function strongly suggestive of this disorder. The NAA is less elevated in milder
of aspartoacylase (710,711). Laboratory diagnosis of Canavan disease cases (717). Reduced glutamate/glutamine is found on short echo pro-
is based on demonstration of increased urinary excretion of NAA. ton MRS (718).
In the newborn, imaging studies of the brain may be normal; how-
ever, proton MR spectra are already abnormal (see later comments Fibrinoid Leukodystrophy (Alexander Disease)
on proton MRS). In typical infantile onset patients, imaging studies Alexander disease (OMIM 203450), also known as fibrinoid leu-
reveal diffuse, symmetric abnormalities of the cerebral white matter kodystrophy, was originally described as a disorder of infants, who
without any focal predominance. When transfontanelle sonography present during the first year of life (sometimes as early as the first few
is performed with high frequency transducers in neonates, it shows weeks of life) with macrocephaly, failure to attain developmental mile-
hyperechogenicity of the white matter, causing a reversal of the normal stones, and progression to psychomotor retardation with death usu-
pattern of cortical and white matter echogenicity (712). CT shows a ally ensuing in infancy or early childhood (57,707,719,720). In the late
diffuse low density in the cerebral and cerebellar white matter and in 1990s, it was discovered that the disease is caused by mutations causing
the globi pallidi. MRI reveals diffuse T1 hypointensity and T2/FLAIR gain of function of the gene for glial fibrillary acidic protein (GFAP),
hyperintensity in the cerebral white matter (Fig. 3-63) (713). The an intermediate filament protein that is highly specific for cells of
subcortical white matter is preferentially affected early in the course astroglial lineage and is located at chromosome 17q21 (721,722). Since
of the disease (distinguishing this disorder from Krabbe disease and the discovery of GFAP dysfunction as the cause, it has been shown that
metachromatic leukodystrophy) and may appear swollen (45,46). Less Alexander disease has a large phenotypic spectrum. Typical infantile
severe cases with milder clinical courses may have nearly normal cere- Alexander disease, with onset before age 2 years, has onset in the first
bral white matter, with T2/FLAIR hyperintensity being seen only in year of life and rapid lethal course, as described above. The juvenile
the subcortical U fibers, corpus striatum, cerebellar dentate nuclei, form of Alexander disease is characterized by onset between age 2 and
dorsal pons, and portions of the cerebellar peduncles (709,711). The 12 years; head size is often normal. Affected children typically present
globi pallidi are nearly always affected (Figs. 3-63 and 3-64), with spar- with ataxia, bulbar signs (sometimes recurrent vomiting) and spastic-
ing of the adjacent putamen; thalami are frequently involved, as well, ity with a more slowly progressive course. Rarely, presentation may be
especially in more advanced stages (Fig. 3-64) (45,155,306,714). The one of spinal cord dysfunction (gladder and gait disturbances (723) )
cerebellar dentate nuclei may be affected (714) and I have seen a case or weight loss and vomiting (724). The adult-onset form of Alexander
involving the midbrain and dorsal pons. Contrast enhancement is not disease has predominantly symptoms of brainstem involvement,
reported. As the disease progresses, diffuse atrophy of the white matter beginning during adolescence or later (after age 12) with dysarthria,
and, subsequently, cerebral cortex is seen (Fig. 3-64) (59). Diffusion- dysphonia, dysphagia, pyramidal signs and ataxia; palatal myoclonus is
weighted imaging shows characteristic reduced diffusion (compared common. Many of these patients survive into adulthood (724–728).
to normal age-matched white matter, Fig. 3-63B) in the entirety of The disease is caused by physiologic disruptions resulting from
the affected white matter due to the myelin vacuolization (10,715); mutation of GFAP proteins. It is speculated that the leukodystrophy of
the reduced diffusion is quite prolonged and may be detectable over Alexander disease is a secondary effect of impaired astrocyte functions
multiple follow-up studies. DTI shows markedly reduced fractional leading to deregulation of astrocyte-derived myelination signals (722).
anisotropy (10). Mutant GFAP precipitates in astrocytes in aggregates, together with
Patients with less severe mutations have fewer findings on MRI, heat-shock proteins and alpha-B-crystallin, to form Rosenthal fibers
limited to some T2 hyperintensity in the basal ganglia (710) or even (eosinophilic inclusions in astrocyte cytoplasm), which disrupt astro-
a completely normal study. In such case, MR diagnosis is dependent cyte function (729). In addition, astrocytic density is increased and a
upon proton MR spectroscopy. paucity of myelin is seen (730). No genotype-phenotype correlation
Proton spectroscopy (Fig. 3-64C) reveals elevated levels of the has been established and, indeed, some mutations have been associated
NAA peak in both mild and severe cases (709,716), a finding that is with all three forms of Alexander disease (728). It appears, therefore,

FIG. 3-63. Canavan disease, early MRI


findings. A. Axial T2-weighted image
shows diffuse white matter hyperin-
tensity, absence of any myelination in
the internal capsules, and abnormal
hyperintensity of the globi pallidi (white
arrows). The white matter volume is
normal; no atrophy is seen. B. Axial dif-
fusivity (Dav) image shows reduced Dav
diffusely in the cerebral white matter
and in the globi pallidi (white arrows).

Barkovich_Chap03.indd 157 5/6/2011 12:08:48 AM


158 Pediatric Neuroimaging

FIG. 3-64. Canavan disease, more advanced MRI find-


ings. A and B. Axial T2-weighted images shows diffusely
hyperintense white matter (isointense to the lateral ven-
tricles), with hyperintensity of the globi pallidi (g) and
thalami (t). Note the enlarged subarachnoid spaces and
cortical sulci, indicating more advanced white matter dis-
ease with volume loss. C. Single voxel proton MRS (TE =
288 ms) from the frontal white matter shows markedly
reduced choline (Ch) and elevated NAA. Creatine (Cr)
appears normal.

that a molecular understanding awaits an understanding of the effects early in the course of the disease. A rim of short T1 and short T2 relax-
of the specific mutation on the three-dimensional structure of the ation times typically persists immediately around the lateral ventri-
GFAP molecule and its functions, as well as the effects of other genetic cles (Fig. 3-65A), particularly the frontal horns; this has been called a
or environmental factors. The diagnosis can be strongly suggested by periventricular “garland.”(723) The ependyma of the trigones may be
imaging findings (725,728), and confirmed by increased levels of GFAP hyperintense on T1-weighted images; the relationship of this hyperin-
in the CSF (731) or analysis of the GFAP gene. tensity to the “garland” is not understood. The basal ganglia are typi-
CT shows low density in the frontal white matter that typically cally involved, particularly the caudate heads, and anterior putamina,
extends posteriorly into the parietal region and internal capsule. Low den- which are usually edematous early in the course of the disease and
sity may be seen in the caudate heads. Contrast-enhancement is often seen may enhance (Fig. 3-65B). The globi pallidi and thalami are less com-
near the tips of the frontal horns early in the disease (732,733). An expanded monly affected. In later stages of the disease and, particularly in the
cavum septi pellucidi is often seen and is of uncertain significance. juvenile and adult forms, the brainstem, particularly the periaqueduc-
van der Knaap et al. have described five MRI criteria by which tal region and the medulla, commonly show T2 prolongation and may
the diagnosis of infantile Alexander disease can be made: (a) exten- enhance after administration of contrast. In addition, cavities/cysts
sive cerebral white matter changes with frontal predominance, (b) a may develop in affected regions of the brain as the disorder progresses
periventricular rim with high signal on T1-weighted images and low (155,725,734). Diffusion imaging shows increased diffusivity in the
signal on T2-weighted images, (c) abnormalities of the basal ganglia affected regions (dark on diffusion-weighted images (Fig. 3-65D),
and thalami, (d) brainstem abnormalities, and (e) contrast enhance- bright on ADC images). Proton MR spectroscopy shows reduced NAA
ment of periventricular regions and lower brainstem (725). The MR in affected areas and, sometimes, increased myo-Inositol (Fig. 3-65E).
findings in the infantile form of the disease are those of T1 hypointen- Patients with the juvenile form of Alexander disease typically
sity and T2 hyperintensity involving most of the frontal white matter have regions of T1 and T2 prolongation in the medulla, pons, and
and extending posteriorly to the parietal white matter and the internal middle cerebellar peduncles; enhancement is intense after intravenous
and external capsules (Fig. 3-65). Subcortical white matter is affected administration of paramagnetic contrast (726). Involvement of

Barkovich_Chap03.indd 158 5/6/2011 12:08:48 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 159

FIG. 3-65. Alexander disease, infantile form. A. Axial T1-weighted image shows low intensity in the frontal white matter, extending into the caudate
heads and the external and extreme capsules. Hyperintensity in and adjacent to the walls of the lateral ventricles (black arrows) is often present. B. Axial
T2-weighted image (different patient) shows marked hyperintensity in the frontal white matter, caudates and putamina, external and extreme capsules.
Note that the abnormal signal involves the subcortical white matter (small black arrows). C. Coronal FLAIR image shows involvement of subcortical parietal
white matter (small black arrows) and cerebellar white matter (large black arrows). D. Axial diffusion weighted image shows low diffusivity (hypointensity)
in the frontal lobes, characteristic of Alexander disease. E. Proton single voxel spectrum (TE = 26 ms) from frontal white matter of an affected 10 months
old shows low NAA, elevated choline (Ch) and elevated myo-inositol (myo-I).

supratentorial (predominantly frontal) white matter and basal ganglia cles (inferior, middle, and superior), midbrain tegmentum, hila of the
often develops a few years after presentation (Fig. 3-66). “Garlands” of T1 deep cerebellar nuclei, and central spinal cord (735). Patchy enhance-
hyperintensity/T2 hypointensity are often seen around the lateral ven- ment is inconsistently seen in these regions. Most patients will also have
tricles (Fig. 3-66) (723). Of interest, the subcortical white matter may not some cerebral periventricular white matter T2 prolongation (735).
be involved in this form of the disease (726). Proton MRS of the cerebral Diffusivity in the supratentorial white matter appears to be generally
white matter shows marked reduction of NAA with elevation of myo-I, increased. Spectroscopy of the supratentorial white matter typically
choline, and lactate (730). DTI of affected white matter shows increased shows increased amplitude of the myo-I peak, with normal amplitudes
diffusivity and decreased FA; magnetization transfer is reduced (730). of other metabolites (728,735).
Patients with the adult-onset form of the disease are found to
have atrophy and abnormal signal intensity in the medulla oblongata, Mucopolysaccharidoses
extending caudally to the C1-2 levels of the spinal cord (728,735). The The mucopolysaccharidoses are lysosomal storage disorders that
signal changes are typically T1 and T2 prolongation; these may be dif- result from deficiency of specific lysosomal enzymes involved in the
fuse, or may specifically affect the medial lemniscus and corticospinal degradation of mucopolysaccharides (glycosaminoglycans). Muco-
tracts. Other areas that may be involved include the cerebellar pedun- polysaccharide deposits in the lysosomes interfere with the degradation

Barkovich_Chap03.indd 159 5/6/2011 12:08:49 AM


160 Pediatric Neuroimaging

FIG. 3-66. Alexander disease, juvenile


form. A. Sagittal T2-weighted image shows
abnormal hyperintensity in the dorsal
medulla (black arrows) and cervical spinal
cord (white arrow) as well as the anterior
portion of the very thin corpus callosum.
B. Parasagittal T1-weighted image shows
abnormal hypointensity of the frontal white
matter (f) with more normal hyperinten-
sity of the parietal (p) and temporal white
matter. C and D. Axial T1 (C) and T2 (D)-
weighted images show the “garland” of
abnormal signal intensity (arrows) around
the lateral ventricles, hyperintense on the
T1-weighted image and hypointense on the
T2-weighted image.

of other macromolecules, resulting in the intralysosomal accumulation flow of interstitial fluid from the parenchyma. Enlarged perivascular
of other materials in addition to mucopolysaccharides. Incompletely spaces are therefore filled with interstitial fluid (and mucopolysac-
degraded mucopolysaccharides accumulate in the tissues and are charides), as described in pathology texts (48). Cerebral white matter
excreted in the urine as dermatan sulfate, heparan sulfate and kera- lesions may represent demyelination secondary to accumulation of
tan sulfate. Diagnosis is made by combining the clinical picture with macromolecules (mucopolysaccharides and others) within neurons
characteristic urinary mucopolysaccharides (736,737). Table 3-13 sum- and oligodendrocytes. If the patient presents with macrocephaly due
marizes the main genetic and biochemical aspects of the various muco- to hydrocephalus, interstitial edema (see Chapter 8) may also play a
polysaccharidoses. The clinical manifestations of these disorders vary. role in the development of white matter signal abnormalities.
Mucopolysaccharidoses are multisystemic diseases. Involvement Indirect CNS damage in mucopolysaccharidoses often results from
of the skeletal system (dwarfism, skull base abnormalities, bone and hydrocephalus or compression of the upper cervical spinal cord in the
joint dysplasias), liver, spleen (hepatosplenomegaly), heart (thickening cranial-cervical junction area. Hydrocephalus in mucopolysacchari-
of the valves), eye (corneal opacities), and central nervous system (pri- doses is likely a result of impaired CSF resorption due to meningeal
mary and secondary involvement) are characteristic (676). Obviously, mucopolysaccharide deposits and, possibly, increased venous pressure
for the neuroradiologist central nervous system involvement is in the due to bony overgrowth of the skull base and narrowing of venous
focus of the diagnostic imaging workup. Involvement of the CNS may outflow pathways. As a result of hydrocephalus, most patients with
be direct or indirect. mucopolysaccharidoses have macrocephaly. Narrowing of the foramen
The most frequent imaging substrates of direct central nervous magnum-upper cervical spinal canal area is due to combined effects of
system involvement are enlarged perivascular spaces and white mat- atlantoaxial instability (odontoid dysplasia in conjunction with liga-
ter abnormalities. Enlargement of perivascular spaces is likely related ment laxity) and mucopolysaccharide deposits within synovial and
to mucopolysaccharide deposition in leptomeninges, impairing out- dural structures. Ligamentous hypertrophy may develop in response to

Barkovich_Chap03.indd 160 5/6/2011 12:08:50 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 161

TABLE 3-13 Mucopolysaccharidoses


Gene Type OMIM#/Eponym Inheritance Enzyme Deficiency Urinary GAG Neurologic Signs
IDUA 4p16.3 IH 607014/Hurler AR Alpha-L-Iduronidase Dermatan sulfate Marked
IDUA 4p16.3 IS 607016/Scheie AR Alpha-L-Iduronidase Heparan sulfate None
IDUA 4p16.3 IH/S 607015/ AR Alpha-L-Iduronidase Dermatan or Intermediate
Hurler-Scheie Heparan sulfate between IH and IS
Genetic Heterogeneity. II 309900/Hunter X-Linked Iduronate sulfatase Dermatan or Mild to moderate
Locus at Xq28 recessive heparan sulfate
SGSH 17q25.3 IIIA 252900/ AR Heparan Heparan sulfate Mental deterioration
Sanfilippo A N-sulfatase
MPS3B 17q21 IIIB 252920/ AR Alpha-N-acetyl- Heparan sulfate Mental deterioration
Sanfilippo B glucosaminidase
HGSNAT 8p11.1 IIIC 252930/ AR Acetyl CoA: alpha- Heparan sulfate Mental deterioration
Sanfilippo C glucosaminide
acetyltransferase
GNS 12q14 IIID 252940/ AR N-acetylglu- Heparan sulfate Mental deterioration
Sanfilippo D cosamine
6-sulfatase
GALNS 16q24.3 IVA 253000/ AR N-acetylgalac- Keratan sulfate and Secondary to skeletal
Morquio A tosamine-6- chondroitin- changes
sulfate sulfatase 6-sulfate
GLB1 3p21.33 IVB 253010/ AR Beta-galactosidase Keratan sulfate Secondary to skeletal
Morquio B changes
ARSB 15q11–13 VI 253200/ AR Arylsulfatase B Chondroitin Secondary to skeletal
Maroteaux-Lamy sulfate changes
GUSB 7q21.11 VII 253220/Sly AR Beta-Glucuronidase Dermatan sulfate Variable
Heparan sulfate
OMIM#, Online Mendelian Inheritance in Man disease number; AR, Autosomal recessive inheritance; GAG, glycosaminoglycan.

chronic subluxation at the C1-2 level, causing additional compression as diffuse areas of T1 hypointensity and T2 hyperintensity on MR
on the upper cervical spinal cord. studies (Figs. 3-67 to 3-70). In Hurler syndrome and, to a lesser extent
Despite many similarities in the pathophysiologic processes, the in Hunter (type II), Sanfilippo (type IIIa), and Maroteaux-Lamy
actual clinical manifestations of mucopolysaccharidoses vary as a (type IV) syndromes, sharply defined enlarged perivascular spaces are
function of the different subtypes. In Hurler (MPS-I-H) and Hunter commonly present in the corpus callosum and basal ganglia, as well as
(MPS-II) diseases, the clinical picture is usually dominated by cen- the cerebral white matter (Figs. 3-68 to 3-70) (59). With progression
tral nervous system involvement, including mental retardation. Some of the disease, the lesions become larger and more diffuse and resem-
degree of intellectual deficit may be present in Scheie (MPS-I-S) and ble a leukodystrophy (Fig. 3-67) (745), reflecting the development of
Hurler-Scheie (MPS-I-HS) diseases. Sanfilippo (MPS-III) disease pres- infarcts and demyelination; if treated early, many of the lesions regress
ents with neurological abnormalities only (hepatomegaly or dysostosis after bone marrow transplantation. Diffuse white matter haziness is
are lacking). Skeletal involvement with secondary CNS manifestations seen on T2-weighted images in mucopolysaccharidoses types IH, II,
dominate the clinical picture in Morquio (MPS-IV) and Maroteaux- and III, resulting in diminished contrast between cortex and underly-
Lamy (MPS-VI) diseases, mainly as a result of vertebral subluxation, ing white matter (Fig. 3-70) (743,746). The atrophy and white matter
which occurs most frequently at the atlantoaxial joint (676,736,737) changes occur earliest in types I, II, III, and VII, usually becoming vis-
with resultant stenosis, cord compression and myelopathy. ible during the first few years of life. In mucopolysaccharidoses types
Imaging studies of the brain in the mucopolysaccharidoses are IV and VI, the white matter changes and atrophy may not become
usually ordered when secondary CNS involvement due to hydroceph- apparent until the second decade of life (736,742–744); these patients
alus, craniocervical instability, or spinal cord compression is suspected typically have normal intelligence (747) and present for medical
(738–741). CT and MRI usually reveal delayed myelination, atrophy, care with reduced exercise tolerance and myelopathy (748). Affected
varying degrees of hydrocephalus, enlarged perivascular spaces, and patients are commonly macrocephalic, probably from a combina-
white matter changes (742–744). In all types of mucopolysacchari- tion of hydrocephalus and mucopolysaccharide deposition within the
doses, the white matter abnormalities are manifested as diffuse low brain, meninges, and skull. This communicating hydrocephalus may
attenuation within the cerebral hemispheric white matter on CT and cause enlargement of subarachnoid spaces and optic nerve sheaths; if

Barkovich_Chap03.indd 161 5/6/2011 12:08:51 AM


162 Pediatric Neuroimaging

FIG. 3-67. Mucopolysaccharidosis 1H, Hurler syndrome. A. Sagittal T1-weighted image shows marked ventriculomegaly and frontal bossing (large white
arrows) due to hydrocephalus. The sphenoid bone is small, leading to a large, deep sella turcica. The basi-occiput (small white arrow) is small, as is the
odontoid process of C2 (white arrowhead). B. Axial T2-weighted image shows ventriculomegaly with diffusely hyperintense white matter due to myelina-
tion delay and interstitial edema. Mildly enlarged perivascular spaces are seen in the thalami and external capsules. C. Coronal reformat of T1-weighted
volumetric acquisition shows subcortical myelination (white arrowheads) and multiple slightly enlarged perivascular spaces (white arrows). D. Sagittal
T2-weighted image of the spine shows irregular endplates of the vertebral bodies and a gibbus deformity (focal kyphus) at T1 (white arrow) due to narrow-
ing of the anterior aspect of the vertebral body.

longstanding, the latter may result in optic nerve atrophy (Fig. 3-70E). ligament. Ligamentous hypertrophy (Fig. 3-70A and D) may develop
A high incidence of intracranial arachnoid cysts may also contribute in response to chronic subluxation at the C1-2 level, causing additional
to the macrocephaly (742,749). compression on the upper cervical spinal cord. MRI of the craniocer-
The spine abnormalities in the mucopolysaccharidoses con- vical junction in affected patients shows a shortened odontoid, with
sist of characteristic vertebral body abnormalities that are described a soft tissue mass of variable size. The soft tissue mass, which is of
well in pediatric and musculoskeletal radiology texts. Their spines intermediate signal on the T1-weighted images and low signal on the
are usually imaged to determine the site and cause of cord compres- T2-weighted images, probably represents a combination of unossified
sion (Fig. 3-70), which occurs frequently in mucopolysaccharidoses fibrocartilage and reactive change (747). A small os odontoideum may
types IV and VI (59,736,737,750). The most common location for the be present. The combination of the anterior soft tissue mass and the
cord compression is at the atlantoaxial (C1-2) joint (Fig. 3-70D and indentation of the posterior arch of C1 may narrow the spinal canal
F). Atlantoaxial instability may occur in these patients as a result of and compress the spinal cord at that level. No abnormal enhance-
laxity of the transverse ligament or because of hypoplasia or absence ment is seen. Another cause of cord compression at the C1-2 level is
of the odontoid (Fig. 3-70A and D). Flexion/extension views are dural thickening resulting from intradural deposition of collagen and
often necessary to demonstrate the laxity of the transverse odontoid mucopolysaccharides. This is seen as a thickening of the soft tissue

Barkovich_Chap03.indd 162 5/6/2011 12:08:51 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 163

FIG. 3-68. Older child with mucopolysaccharidosis 1H, Hurler syndrome. A and B. Sagittal T1-weighted images show multiple well-defined focal
areas of hypointensity, probably mucopolysaccharide-filled perivascular spaces, in the cerebral white matter and corpus callosum. C. Axial T2-weighted
image shows well-marginated foci of hyperintensity superimposed upon some subtle, diffuse hyperintensity in the centrum semiovale.

FIG. 3-69. Mucopolysaccharidosis II, Hunter syndrome. A. Sag-


ittal T1-weighted image shows multiple small cysts (white arrows)
in the corpus callosum. B and C. Axial T2-weighted images show
dilated perivascular spaces and multiple cystic enlargements of
perivascular spaces (white arrowheads) throughout the cerebral
hemispheres.

Barkovich_Chap03.indd 163 5/6/2011 12:08:52 AM


164 Pediatric Neuroimaging

FIG. 3-70. Mucopolysaccharidosis IIIA (Sanfillipo) and IV (Maroteaux-Lamy) syndromes.


A. Sagittal T1-weighted image of a patient with Sanfillipo syndrome shows two cysts (large white
arrowheads) in the body of the corpus callosum. Note that the basiocciput (small white arrows)
is small and the odontoid process of C2 (large white arrow) is absent. The ligaments connect-
ing the odontoid to the basiocciput (small white arrowheads) are hypertrophied. B and C. Axial
FLAIR image (B) and coronal T2-weighted image (C) of the same patient show the cysts in the
corpus callosum (white arrows) and show slight hyperintensity of the cerebral white matter, result-
ing in indistinct cortical-white matter junctions. D. Sagittal T1-weighted image of a child with
Maroteaux-Lamy syndrome shows an abnormally thick clivus (large white arrows) compressing
the pons. Abnormal soft tissue (t) surrounding the dens is probably a combination of mucopoly-
saccharide and hypertrophied ligaments. The vertebral bodies (small white arrows) are small and
pointed anteriorly, separated by large intervertebral discs. The calvarium is thick, the posterior fossa
is small, and there is a dilated perivascular space (white arrowhead) in the corpus callosum. E. Axial
T2-weighted image of the same patient as in (D) shows large sylvian fissures (s) and dilated optic
sheaths (white arrows) containing atrophic optic nerves due to increased intracranial pressure. F.
Axial T2-weighted image at the foramen magnum shows anteroposterior compression of the cervi-
comedullary junction (arrows) by the soft tissues surrounding the unstable dens. G and H. Sagittal
T1 (G) and T2 (H)-weighted images show spinal stenosis secondary to intervertebral disc enlarge-
ment, likely due to the deposition of mucopolysaccharides.

Barkovich_Chap03.indd 164 5/6/2011 12:08:53 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 165

FIG. 3-70. (Continued)

posterior to the dens, resulting in narrowing the subarachnoid space tions have been identified in peroxisomes (756). Among the most
at that level. One final cause of cord compression in these patients is important are betaoxidation of a specific set of fatty acids and their
gibbous formation in the thoracic spine. This results from the vertebral derivatives, synthesis of ether-phospholipids and plasmalogens, alpha-
deformities and abnormal enlargement of intervertebral discs (Fig. oxidation of phytanic acid, and biosynthesis of cholesterol and bile
3-70G and H) and is most common in Morquio disease (mucopolysac- acids (49,50,757,758). Some authors classify peroxisomal disorders
charidosis type IV) (59,736,737,750). Meningeal thickening may result into three major groups, based upon the severity of the enzyme dys-
in cyst formation in mucopolysaccharidoses types 1H and II (59). function, whereas others divide them into two groups (Table 3-14),
Proton MR spectroscopy results have been reported in a few of the Deficiencies in Peroxisomal Biogenesis and Single Peroxisomal Protein
mucopolysaccharidoses (751). Affected patients tend to have multiple Deficiencies (756,759), which are described below in greater detail.
resonances from glycosaminoglycans deposited in the brain; these are Peroxisomal disorders show remarkable clinical phenotypic het-
seen from about 3.3 to 4.4 ppm, upfield from the normal creatine and erogeneity but dysmorphic features, hepatointestinal dysfunction
choline peaks (752). Care should be taken not to mistake these peaks and CNS involvement are common in many of them; therefore per-
for a lack of water suppression. Other reports suggest a decreased NAA/ oxisomal diseases should always be suspected in a neonate presenting
choline ratio and elevated glutamate, glutamine, and myo-Inositol. with a polymalformative syndrome associated with severe neurologi-
cal disturbances. The imaging abnormalities in peroxisomal disorders
Multiple Sulfatase Deficiency have a wide spectrum of patterns, but the most characteristic patterns
Multiple sulfatase deficiency is rare autosomal recessive disorder that include neuronal migration disorders with abnormal myelination
has clinical and imaging features of both mucopolysaccharidoses and (e.g., Zellweger syndrome, neonatal adrenoleukodystrophy) or sym-
metachromatic leukodystrophy (676); therefore, it is listed here as a metrical demyelination that most typically involves the corticospinal
subcategory of mucopolysaccharidoses. As its name indicates, multiple tracts, corpus callosum, and posterior greater than anterior white mat-
sulfatase enzymes (those involved in metachromatic leukodystrophy ter structures (e.g., pseudoneonatal adrenoleukodystrophy, X-linked
and in the various forms of mucopolysaccharidoses) are deficient; the adrenoleukodystrophy-adrenomyeloneuropathy complex). Classic
residual enzyme activities vary considerably, explaining the significant X-linked adrenoleukodystrophy has already been discussed in section
phenotypic variations. The most frequent form occurs in early child- IV.B.1.c as a white matter disorder with central white matter involve-
hood, but rare neonatal and juvenile forms also exist (676). ment. A group of peroxisomal disorders with generalized loss of perox-
Clinically, facial dysmorphia (similar to that seen in mucopoly- isomal function, present at birth. A few of these disorders will be briefly
saccharidoses), hepatosplenomegaly, microcephaly (macrocephaly in discussed, with focus primarily on imaging characteristics. Interested
the neonatal form (753) ), delayed development, progressive spasticity, readers are referred to review articles or texts in Child Neurology or
blindness and deafness are usually found. Imaging findings in multiple Metabolic Disease for further details (756,760,761).
sulfatase deficiency represent the combination of abnormalities seen
in metachromatic leukodystrophy and mucopolysaccharidoses, nota- Peroxisomal Biogenesis Disorders
bly variable patterns of white matter disease and diffuse brain atrophy; Several disorders seem to result from deficient peroxisomal biogenesis,
occasionally, these are associated with enlarged perivascular spaces and including Zellweger syndrome, neonatal adrenoleukodystrophy, and
craniocervical junction abnormalities (754,755). infantile Refsum disease. Common to all three are liver disease, vari-
able neurodevelopmental delay, retinopathy, and perceptive deafness
Peroxisomal Disorders with onset in the first months of life. All of these disorders seem to
Peroxisomes are small cellular organelles that contain multiple com- be inherited as autosomal recessive traits (762). The underlying prob-
pounds that are essential for normal growth and development of the lem appears to be mutations in peroxisomal biogenesis genes, which
organism. The biochemical functions that take place in peroxisomes code for proteins (peroxins) essential for peroxisome formation (763).
are beyond the scope of this book; more than 40 enzymatic func- Mutations that completely destroy the protein’s function seem to cause

Barkovich_Chap03.indd 165 5/6/2011 12:08:55 AM


166 Pediatric Neuroimaging

TABLE 3-14 Classification of Peroxisomal Disorders


Group A: Peroxisome Biogenesis Disorders
1. Zellweger spectrum disorders
a. Zellweger syndrome
b. Neonatal adrenoleukodystrophy (NALD)
c. Infantile Refsum disease
2. Rhizomelic chondrodysplasia punctata (RCDP)
3. Refsum disease secondary to PEX7 mutation
Group B: Loss of Single Peroxisomal Function (Peroxisomes Present)
1. Disorders of peroxisomal beta-oxidation
a. X-Linked adrenoleukodystrophy and variants
b. D-bifunctional protein deficiency
c. Acyl-CoA oxidase deficiency (pseudo-NALD)
d. 2-methylacyl-CoA racemase deficiency
e. SCPx deficiency
2. Disorders of etherphospholipid biosynthesis
a. Dihydroxyacetone phosphate (DHAP) acyltransferase deficiency
b. Alkyl dihydroxyacetone phosphate (DHAP) synthase deficiency
3. Disorders of fatty acid alpha-oxidation
a. Phytanoyl-CoA 2-hydroxylase deficiency
b. 2-Hydroxyphytanoyl-CoA lyase deficiency
c. Di-and trihydroxycholestanoic acidemia
d. Glutaryl-CoA oxidase deficiency (Glutaric aciduria type III)
e. Classic Refsum disease (Phytanoyl-CoA hydrolase deficiency)
4. Disorders of glyoxylate metabolism
a. Hyperoxaluria type 1 (AGT deficiency)
b. Acatalasemia
Modified from Raymond GV, Naidu S, Moser HW. Peroxisomal disorders. In: Swaiman KF, Ashwal S, Ferriero DM, eds.
Pediatric Neurology: Principles and Practive, 4th ed. Philadelphia, PA: Mosby Elsevier, 2006:735–758; Wanders RJ,
Waterham HR. Biochemistry of mammalian peroxisomes revisited. (Annu Rev Biochem) 2006;75:295–332, with permission.

the more severe (Zellweger) phenotype, while mutations that allow (most commonly near the foramina of Monro, Figs. 3-71B and 3-72B)
some residual peroxin function permit some peroxisomes to form and (766). The cortical malformation consists primarily of gyri that are
result in the less severe (neonatal adrenoleukodystrophy and infantile too numerous and too small (Fig. 3-71); pathology studies suggest
Refsum disease) phenotypes (762). that these do not represent true polymicrogyria, but merely too many
Zellweger syndrome (also called cerebrohepatorenal syndrome, gyri of decreased amplitude (769). On imaging, the appearance most
OMIM 214100) results from a nearly complete absence of peroxi- commonly looks like polymicrogyria in the region of the sylvian fis-
somes, resulting in a wide range of biochemical abnormalities, includ- sures (Figs. 3-71A and C and 3-72A). Multiple small gyri (clearly not
ing impaired catabolism of very long chain fatty acids, bile acid polymicrogyria) are often found in the anterior frontal and tempo-
intermediates, pristanic, phytanic, and pipecolic acids, as well as low ral lobes (Fig. 3-71A). In addition, some patients have a malforma-
levels of plasmalogens due to impaired synthesis (764). This autosomal tion resembling pachygyria or band heterotopia (see Chapter 5),
recessive syndrome can be caused by mutations of any of a number of with a thick layer of arrested neurons deep to a thin outer cortex, in
genes involved in peroxisome biosynthesis including those on chro- the perisylvian and perirolandic cortex (Fig. 3-72). Subependymal
mosomes 7q21 (PEX1), 8q (PEX2), 6q (PEX3), 12 (PEX5), 6p (PEX6), germinolytic cysts, most commonly seen in the caudothalamic notch
17 (PEX12), 1p36.2 (PEX14), and 22q11.21 (PEX26) (44); in addition, (Fig. 3-71B) are identified best on sagittal and coronal T1-weighted
a Zellweger syndrome locus on 7q11 is suspected on the basis of chro- images. (Other conditions with germinolytic cysts include congeni-
mosome aberrations (765). Affected patients have multiple congeni- tal viral infections, fetal circulatory disorders, D-2-hydrdoxyglutaric
tal anomalies (craniofacial, eyes, liver, bones, brain) and are severely aciduria, pyruvate dehydrogenase E1-alpha deficiency, and complex
hypotonic and weak from birth, may have severe psychomotor retar- mitochondrial dysfunction with pontocerebellar hypoplasia (770).
dation, dysmorphic facial features, hypotonia, seizures, periarticular Proton MRS results have also been reported in Zellweger syndrome,
calcifications, and hepatomegaly with impaired liver function; survival but are nonspecific. NAA is significantly reduced, even in the neona-
is typically less than 1 year (756). tal period (Fig. 3-71D). Lactate and lipid peaks are seen at long echo
Imaging findings in Zellweger syndrome (Fig. 3-71) (766) reflect times of 270 to 280 ms (771).
the pathologic characteristics (767,768). Patients have profound Neonatal adrenal leukodystrophy (NALD, OMIM 202370) differs
hypomyelination (Fig. 3-71A), malformations of the cerebral cortex fundamentally from X-linked adrenal leukodystrophy radiologically
(Figs. 3-71A and C and 3-72A), and subependymal germinolytic cysts as well as clinically; the choice of name for this disorder is unfortu-

Barkovich_Chap03.indd 166 5/6/2011 12:08:55 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 167

FIG. 3-71. Three-week-old neonate with Zellweger syndrome. A. Axial T2-weighted image shows an increased number of abnormally small gyri (white
arrows) in the frontal lobes and polymicrogyria (black arrows) in the sylvian fissures. Myelination is delayed. B. Axial T1-weighted image shows germinolytic
cysts (white arrows) near the caudothalamic notch of both lateral ventricles. Abnormally small gyri with shallow sulci are seen in the posterior sylvian
fissures (white arrowheads). C. Coronal T2-weighted image shows polymicrogyria (black arrows) in the walls of the posterior sylvian fissures. The cerebellum
is abnormally small and has abnormal foliation. D. Single voxel proton MRS (TE = 288 ms) from the basal ganglia shows abnormally small NAA peak.

nate and confusing (it results from the finding of adrenal atrophy and The head size is small and the corpus callosum extremely atrophic as
cytoplasmic inclusions in the first case described (772). The distinction a result of the white matter atrophy. Patients usually die in late infancy
of NALD from Zellweger syndrome is less clear; indeed, some authors (775). Imaging results vary from nearly complete absence of myelin
consider the two disorders to be parts of a continuum that includes in the cerebral white matter and severe loss of white matter volume
Zellweger (the most severe phenotype), NALD (intermediate pheno- (47) to subtle polymicrogyria (45) to inflammatory demyelination of
type) and infantile Refsum disease (OMIM 266510, least severe pheno- the corticospinal tracts, cerebellum, and posterior cerebral white mat-
type), as all result from mutations of the same gene and have significant ter (similar to X-linked adrenoleukodystrophy (section IV.B.1.c) in
clinical overlap (107,756,766,768,773,774). It is postulated that less location and enhancement) (777,778). Distinction from X-linked ALD
severe mutations allow preservation of some peroxisomal function that is easy, however, as affected children present at younger ages (infancy
results in a somewhat less severe phenotype (756,762), but attempts to instead of the second half of the first decade) and include both boys
differentiate them genetically (or biochemically) do not reliably pre- and girls.
dict the clinical course (756). Patients with milder forms of peroxi-
somal biogenesis disorders are typically born hypotonic at birth with Imaging Differential Diagnosis of Peroxisomal Biogenesis Dis-
mild cranial dysmorphism (midface hypoplasia), poor feeding, and orders Several disorders can have a similar neuroimaging appearance
hepatomegaly. Seizures and developmental delay developing during to that of the peroxisomal biogenesis disorders. These include peroxi-
infancy (775), and tremor, ataxia, hyperreflexia, sensory deficits, and somal bifunctional enzyme defect (764,780) and peroxisomal thiolase
progressive auditory and visual dysfunction develop over time (776). deficiency (pseudo-Zellweger) (781), which have hypomyelination

Barkovich_Chap03.indd 167 5/6/2011 12:08:55 AM


168 Pediatric Neuroimaging

FIG. 3-72. Zellweger syndrome with band heterotopia. A. Axial T2-weighted image shows bilateral perisylvian polymicrogyria (white arrows) as well as a
layer of gray matter several millimeters deep to the cortex (black arrowheads), representing a band heterotopia. B. Coronal T1-weighted image shows large
bilateral germinolytic cysts (large white arrows) at the caudothalamic notch of both lateral ventricles, as well as a faint band heterotopia (small white arrows)
just deep to the cortex over both posterior frontal convexities.

with perisylvian polymicrogyric-type cortical malformations, and acyl- appears to have several variants (Type 1 OMIM 215100, Type 2
CoA oxidase deficiency (see next paragraph) (782), which has a similar OMIM 222765, Type 3 OMIM 600121), characterized by deficien-
MRI appearance to infantile Refsum disease. In addition, patients with cies in several peroxisomal proteins, including phytanoyl-coenzyme
these disorders have neonatal courses and phenotypic manifestations A hydroxlyase, alkyl-DHAP synthetase, 3-ketoacyl-CoA thiolase,
that resemble those in peroxisomal biogenesis disorders (50). Other and dihydroxyacetonephosphate acyltransferase (DHAP-AT) (786).
differential considerations from a neuroimaging perspective include Patients with the DHAP-AT form of rhizomelic chondrodysplasia
the CMDs with brain malformations (see Chapter 5) and congenital punctata have mutations of the PEX7 gene, which is located on chro-
cytomegalovirus infections (see Chapter 11). All of these disorders mosome 6q22-24 (787–789). Affected patients are short in stature,
have hypomyelination associated with cortical malformations. Differ- with disproportionate shortening of the proximal parts of the extremi-
entiation is made by associated findings, which include brainstem and ties (rhizomelia). They have a characteristic facies, with frontal boss-
cerebellar anomalies in CMDs (Chapter 5), and white matter gliosis ing, flat nasal bridge, and small nares, and associated microcephaly,
with diffuse or multifocal polymicrogyria (if present) in congenital cataracts, and ichthyosis (756). Development is marked by a severe
cytomegalovirus infection (Chapter 11). motor and cognitive delay. Plain X-ray shows severe shortening of
long bones, metaphyseal cupping and splaying, and disturbed ossifi-
Acyl-CoA-oxidase Deficiency cation of the humeri and femora, with stippling of the epiphyses of
Acyl-CoA-oxidase deficiency (ACOX1, OMIM #264470) is an auto- the long bones (Fig. 3-74). MRI of the brain shows patchy T2 pro-
somal recessive disorder in which very long chain fatty acids accumulate longation, sulcation abnormalities, and subependymal heterotopia
due to mutations of the ACOX1 gene at chromosome 17q25 (782,783). (Figs. 3-74 and 3-75). We have seen compression of the cervicomedul-
Affected patients are dysmorphic and typically present during the sec- lary junction and tethering of the spinal cord (Fig. 3-74). Proton MRS
ond or third year of life with hypotonia, generalized seizures with tonic reportedly shows elevated levels of mobile lipids and the presence of
jerks, failure to thrive, poor response to visual and auditory stimuli, loss acetate and other ketone bodies, characterized by a peak at 1.9 ppm,
of motor achievements, and hepatomegaly (784,785). Death often ensues just upfield from NAA (790).
during the middle of the first decade (785). Brain MRI is character-
ized by abnormal T1 hypointensity and T2/FLAIR hyperintensity in the Nonrhizomelic Chondrodysplasia Punctata
corticospinal tracts, periventricular white matter, and corpus callosum Nonrhizomelic chondrodysplasia punctata is a disorder caused by
(Fig. 3-73) (784); the pattern is similar to that of X-linked adrenoleu- an isolated deficiency of dihydroxyacetonephosphate acyltransferase
kodystrophy and infantile Refsum disease. Characteristics of diffusion (791). Affected patients present with failure to thrive and microcephaly;
imaging and proton MR spectroscopy have not been reported. subsequent examination shows axial hypotonia with limb spasticity,
a long philtrum, and thin lips. Epilepsy almost always develops, usually
Rhizomelic Chondrodysplasia Punctata starting as febrile seizures during the first few years of life. Radiologic
Rhizomelic chondrodysplasia punctata is an autosomal recessive dis- exam by skeletal survey shows chondrodysplasia punctata without rhi-
ease of peroxisome biogenesis, in which the main biochemical prob- zomelia. MRI of the brain shows patchy T2 prolongation, predomi-
lem is a deficiency in the biosynthesis of plasmalogens. This disorder nantly in the deep white matter (792).

Barkovich_Chap03.indd 168 5/6/2011 12:08:56 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 169

FIG. 3-73. Acyl-CoA-oxidase deficiency. The MRI appearance of this disorder is similar to that to X-linked adrenoleukodystrophy and infantile Refsum
disease. A. Axial T2-weighted image shows abnormal hyperintensity in the pontine corticospinal tracts (black arrowheads) and cerebellar white matter
(white arrows). B. Coronal T2-weighted image shows abnormal hyperintensity in the cerebral white matter (white arrows), cerebellar white matter (white
arrowheads), and middle cerebellar peduncles (black arrows). C. Axial T2-weighted image shows abnormal hyperintensity in the callosal splenium and pari-
etal white matter with extension into the posterior limbs of the internal capsules (white arrows). D. Axial postcontrast T1-weighted image shows enhance-
ment in the callosal splenium (white arrowheads), peritrigonal white matter (white arrows), and posterior limbs of the internal capsules (black arrows).
(These images are courtesy of Erik Gaensler, San Francisco.)

Wilson Disease and progression is slower than in the hepatic form (57). Several clinical
Wilson disease, also known as hepatolenticular degeneration (OMIM phenotypes have been identified. The classic form is characterized by
277900), is an autosomal recessive disorder in which a deficient trans- extrapyramidal signs (dystonia and Parkinson-like features). In other
port protein prevents the elimination of copper from cells (mainly patients, cerebellar signs (ataxia and wing-beating tremor) or bulbar
hepatocytes) and incorporation into ceruloplasmin in blood. As a signs (swallowing difficulties, dysarthria) and seizures may dominate
result, large amounts of copper accumulate in mitochondria, an caus- the neurological picture (795). Deterioration in school performance
ing free radical formation and oxidative damage in multiple organs and behavior, intellectual impairment or emotional disturbances may
(liver, brain, kidney, eye) (793). The responsible gene is called ATP7B also be observed. In patients with neurologic presentations, the diag-
and is located at chromosome 13q14.3-21.1 (794). Initial symptoms in nosis is usually made from the presence of Kayser-Fleischer rings, rings
children are usually related to liver failure (jaundice or portal hyperten- of green pigmentation in the cornea. However, when Wilson disease
sion). When neurologic symptoms predominate, the appearance of the presents with hepatic symptoms, Kayser-Fleischer rings may not yet
illness is delayed until the end of the first decade or the second decade be present.

Barkovich_Chap03.indd 169 5/6/2011 12:08:57 AM


170 Pediatric Neuroimaging

FIG. 3-74. Rhizomelic chondrodysplasia punctata. A. Antero-


posterior radiograph shows irregularity and abnormal stippling
of multiple joints (white arrows), including shoulders, elbows,
hips, and costo-vertebral joints. B. Sagittal T2-weighted image
at the cranio-cervical junction shows an edematous upper cervi-
cal spinal cord (black arrows) that is compressed by the posterior
arch of C1 (white arrowhead). Foliation of the cerebellar vermis
appears abnormal and the ventral pons appears small. C. Sagittal
T2-weighted image of the thoracolumbar spine shows abnormal
appearing vertebrae resulting from the chondrodysplasia. The tip
of the conus medullaris is low and the filum terminale (white
arrowheads) is thick, suggesting a tethered spinal cord. D. Axial
T2-weighted image at the level of the lateral ventricles shows large
ventricles, abnormally hyperintense white matter, and irregular,
abnormal sulcation of the cerebral cortex.

FIG. 3-75. Rhizomelic chondrodysplasia punc-


tata. A. Axial T2-weighted image shows patches
of subcortical T2 prolongation (open white
arrows) and a periventricular nodular heteroto-
pion (open black arrow). B. Axial T2-weighted
image at a higher level shows asymmetrical
patchy white matter hyperintensities (arrows).
A venous malformation is present in the parietal
region. (This case courtesy Dr. Curtis Sutton.)

Barkovich_Chap03.indd 170 5/6/2011 12:08:57 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 171

CT findings of Wilson disease consist of low density in the basal changes may resolve after chelation therapy (795). Lesions of the claus-
ganglia and variable cerebral white matter atrophy (796). Brain MRI tra are inconsistently seen but, when present, may be suggestive of Wil-
in clinically symptomatic patients is usually, but not always, abnormal. son disease. Reduced diffusivity may be seen in actively involved regions
Indeed, in a small portion (27%) of patients with mild neurological (Fig. 3-76E). The presence of deep gray nucleus involvement seems to
manifestations, conventional MRI may be normal; conversely, in about correlate well with neurologic disability, striatal lesions with pseudo-
one third of clinically asymptomatic patients subtle abnormalities may parkinsonism, and putaminal lesions with dystonia (802). Cortical and
be found (797,798). Patients presenting with hepatic failure in child- white matter atrophy are present in up to 70% or patients as the disease
hood are usually found to have bilateral, symmetric T1 shortening (high advances (Fig. 3-76D) (801) and, occasionally, focal areas of abnormal
intensity on T1-weighted images) in the globus pallidus (Fig. 3-76A) signal intensity are seen in the cerebral hemispheric white matter, pri-
and dorsal midbrain, secondary to the hepatic failure and portosys- marily frontally and temporally (803,804); subcortical white matter is
temic shunting (799,800). The T1 changes are believed to result from typically affected more than deep white matter (801). Although cere-
abnormal accumulation of manganese; they may be conspicuous even bral involvement is most commonly bilateral and symmetrical, asym-
in clinically well-controlled patients who do not have other detectable metric or unilateral involvement can occur (805). Proton spectra are
CNS abnormalities (on T2-weighted images). MR findings in patients normal in treated patients (806–808). Untreated patients show slight
presenting with CNS signs and symptoms consist of T1 hypointensity decrease in all metabolites; the mean values of NAA/Cho and NAA/Cr
and T2/FLAIR hyperintensity in the putamina, caudate nuclei, globi were lower in patients with Wilson disease than in the control subjects
pallidi, dorsal mesencephalon, thalami, cerebral peduncles, superior (Fig. 3-76F) (808). Myo-Inositol/creatine ratios were increased in the
cerebellar peduncles, and claustra (Fig. 3-76) (801); some of the T2 basal ganglia on short echo spectroscopy (809).

FIG. 3-76. Wilson disease. A. Coronal


T1-weighted image shows abnormal hyper-
intensity of the globi pallidi (g) bilaterally. B.
Axial T2-weighted image shows abnormal
hyperintensity (white arrows) in the dorsal
midbrain. C. Axial T2-weighted image at
the level of the basal ganglia shows abnor-
mal hyperintensity (white arrows) in the
caudate heads and putamina. D and E. Axial
T2-weighted (D) and diffusion-weighted
(E) images 5 years later shows enlarged CSF
spaces and some new hyperintensity (white
arrows in D) in the thalami, compatible with
progression of disease. The reduced diffusiv-
ity (white arrows in E) in the lateral thalami
indicates active cellular injury. F. Proton MR
spectrum (TE = 288 ms) shows reduced
choline and creatine peaks.

Barkovich_Chap03.indd 171 5/6/2011 12:08:59 AM


172 Pediatric Neuroimaging

FIG. 3-76. (Continued)

Mitochondrial Disorders (Respiratory Chain Disorders) weakness, hypotonia, delayed development, and cardiopulmonary
The mitochondrial disorders (Tables 3-15 and 3-16) are a group of dis- failure leading to early death. The other major multisystemic presenta-
eases characterized by disorders of mitochondrial function that result tion is that of Leigh syndrome; indeed, complex I deficiency seems to
in (a) elevated NADH/NAD+ ratio in cells, (b) impaired adenosine
triphosphate (ATP) production in affected cells, (c) increased super-
oxide radical formation, and (d) functional impairment of numerous
metabolic pathways (810–816). Mitochondrial disorders are relatively TABLE 3-15 Disorders of the
common metabolic disorders of childhood, with recent demonstration Respiratory Chain
of a birth prevalence of inherited mitochondrial disorders to be greater
than 1 in 5,000 (817). Single or multiple organs (brain, heart, muscles,
I. Complex I Deficiency
kidney, liver, endocrine glands, bone marrow) can be involved, with
A. Myopathy
the striated muscles and brain most commonly affected (810–814).
B. Multisystemic disorder
A mitochondrial disorder should be suspected in patients presenting
1. Fatal Infantile disorder
with an unexplained combination of neuromuscular and/or non-
2. Leigh syndrome
neuromuscular symptoms, a progressive course, and involvement of
seemingly unrelated organs or tissues (815), particularly if associated II. Complex II Deficiency
with certain “red flags” such as short stature, neurosensory hearing A. Succinate dehydrogenase deficiency
loss, progressive external ophthalmoplegia, axonal neuropathy, diabe- B. Coenzyme Q10 deficiency
tes mellitus, hypertrophic cardiomyopathy, or renal tubular acidosis 1. Myopathic form
(4). It should also be suspected in severely ill newborns when there is 2. Ataxic form
little evidence suggestive of an intrapartum hypoxic-ischemic insult 3. Infant encephalomyopathic form
or when MR features are unusual for hypoxic-ischemic injury (818). III. Complex III Deficiency
Classification of these disorders is difficult. Classification neither by A. Multisystemic disorders
phenotype nor by morphology has been satisfactory, particularly in 1. GRACILE syndrome (growth retardation, aminoaciduria,
children in whom classic “syndromic” findings tend to be absent (4). cholestasis, iron overload, lactic acidosis, early death)
Classification by metabolic pathway involved has been similarly unsat- 2. Leigh syndrome
isfactory. Therefore, the term mitochondrial disorder is becoming B. Myopathy
restricted to disorders of a single biochemical pathway, the respiratory C. Infantile histiocytoid cardiomyopathy
chain (819).
IV. Complex IV Deficiency
The respiratory chain contains five functional units, called com-
A. Myopathic forms
plexes I-V, that are embedded in the inner mitochondrial membrane
B. Multisystemic forms
(Table 3-15). Its function is to transfer electrons from reduced nucle-
1. Leigh syndrome
otides and flavoproteins to molecular oxygen with resultant oxidation
2. Alpers syndrome
of NADP and FAD. Almost all of these disorders are associated with
lactic acidosis and an elevated lactate/pyruvate ratio (819). Complex I V. Complex V Deficiency
(reduced NADP-CoQ reductase) is the largest complex of the respira- A. Multisystemic forms
tory chain. Patients with complex I deficiency can be subclassified as 1. Leigh syndrome
those with myopathy or multisystemic disorder (819). The myopathy, 2. NARP (Neuropathy, Ataxia, Retinitis Pigmentosa)
manifested as exercise intolerance and limb weakness, typically develops 3. Familial bilateral striatal necrosis
in childhood or early adult life. The multisystemic involvement can be 4. Fatal infantile multisystemic disease
manifested as a fatal infantile disorder with congenital lactic acidosis,

Barkovich_Chap03.indd 172 5/6/2011 12:09:00 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 173

mitochondrial disorders and may raise the possibility of the diagnosis;


TABLE 3-16 Clinical Classification of these include seizures, hypotonia, peripheral neuropathy, ataxia, oph-
thalmoplegia, bulbar signs, short stature, mental deterioration, muscle
Mitochondrial Disorders weakness, cardiomyopathy, cardiac arrhythmia, exercise intolerance,
and neurosensory hearing loss (4,811–813,824). When the classic symp-
I. Exclusive or Predominant Muscle Involvement. tom complex of a specific mitochondrial disorder is found, a specific
A. Fatal infantile myopathy diagnosis is rather straightforward. However, such findings are rare in
B. Benign infantile myopathy children. This blurring of symptom complexes has created controversy
II. Predominant Brain Involvement concerning the classification of mitochondrial disorders. Some groups
A. Subacute necrotizing encephalomyelopathy (Leigh syndrome) (811,814,825) believe that certain clinical features are adequately clus-
1. Pyruvate dehydrogenase complex tered in some patients to allow the identification of syndromes that
2. Respiratory chain complex (cytochrome c oxidase) are useful in patient management. Other investigators believe that
3. ATPase 6 subunit of complex V the overlap of features among the “syndromes” is too great to make
B. Alpers syndrome the clinical classifications useful; they await a biochemical/molecular
C. Myoclonic epilepsy with ragged-red fibers (MERRF) genetic classification (3,4,812,813,816,826–828). Moreover, many dis-
D. Trichopoliodystrophy (Menkes disease) orders of organic acid and fatty acid metabolism lead to abnormal
E. Mitochondrial Encephalopathy with lactic acidosis and mitochondrial function (829), as do some neurodegenerative disorders
stroke-like episodes (MELAS) (830); should these be classified as mitochondrial disorders? Overall,
F. Glutaric Acidurias Types I and II the classification has not become any clearer in the past 15 years; there-
G. Familial Mitochondrial Encephalopathy with fore, it remains sufficient to understand that mitochondrial disorders
macrocephaly, cardiomyopathy, and Complex I deficiency and other metabolic disorders are not always clearly separate and distinct
H. Neonatal lactic acidosis, complex I/IV deficiency, and fetal entities, and that the disorders in this group, even when distinguishable,
cerebral disruption have significant overlap.
Considering the considerable overlap among mitochondrial disor-
III. Other
ders, it is not surprising that the imaging findings are not distinctive.
A. Progressive external ophthalmoplegia
Some patients may have only delayed myelination or a nonspecific T2
1. Isolated
prolongation in the white matter (831). Others may have white matter
2. With retinitis pigmentosa and other organ involvement
cavitation or cortical malformations (832). Recently, it has been noted
(Kearns-Sayre syndrome)
that cerebellar atrophy is common in many mitochondrial disorders
3. Encephalomyopathy in adults
(833). However, certain imaging characteristics should cause mito-
4. Mitochondrial neurogastrointestinal encephalopathy (MNGIE)
chondrial disorders to be included in the differential diagnosis. Disor-
ders of mitochondrial function should be considered in any infant or
child who has abnormalities of the deep cerebral gray matter or dorsal
be one of the major causes of Leigh syndrome (see discussion later in brainstem, in particular if white matter disease or cerebellar atrophy
this section) (819). Complex II (succinate-CoQ reductase) deficiency is is present, as well. The finding of significantly increased lactate in the
less common; it typically presents as an encephalomyelopathy. Succinate brain parenchyma or CSF by proton MR spectroscopy also supports
dehydrogenase deficiency (see discussion later in this section) belongs the possibility of mitochondrial disease (834) (please note, however,
to this group, as does CoQ10 deficiency. The latter can cause a myopa- that mild elevation of lactate is common in the neonate and nonspe-
thy characterized by recurrent myoglobinuria and CNS dysfunction cific). MRS of CSF may be more sensitive than MRS of cerebral tissue,
that includes seizures, ataxia, or mental retardation. When ataxia pre- as lactate is slowly cleared from brain tissue into the CSF before being
dominates, significant brainstem involvement or cerebellar atrophy may resorbed into the systemic circulation (835).
be present (see section V.E.3.f on cerebellar atrophy in this chapter for Certain mitochondrial disorders, such as MERRF (myoclonus, epi-
more details). Complex III (coenzyme Q-cytochrome-c oxidoreductase) lepsy, ragged red fibers) and Leber hereditary optic neuropathy are seen
defects are divided into three forms. The first is a generalized multisys- primarily in adults and, therefore, are not discussed in this book.
temic disorder manifested by limb weakness, exercise intolerance, and
various neurologic signs (819). The second is a tissue-specific syndrome Mitochondrial Encephalomyopathy with Lactic Acidosis and
manifested as a pure myopathy with childhood onset (819). The third is Strokelike Episodes (MELAS)
a pure cardiomyopathy manifested in early infancy (819). In addition, MELAS (OMIM 540000) refers to a group of disorders causing episodes
a rapidly fatal infantile disorder called GRACILE (growth retardation, of throbbing headache, nausea, vomiting, and permanent or reversible
aminoaciduria, cholestasis, iron overload, lactic acidosis, early death) has stroke-like events (hemianopsia and hemiparesis) in conjunction with
been described in Finland (820). Complex IV (cytochrome c oxidase) some of the signs and symptoms of generalized mitochondrial disease
defects can also be divided into myopathic and systemic forms (821). (811,836–841). Patients can present at any age, most commonly in
Myopathies include a fatal infantile syndrome and a benign infantile syn- the second decade (841). Serum and CSF lactate are usually elevated
drome. The main multisystemic presentation is Leigh syndrome; Alpers at the time of presentation. Affected patients may have any of several
disease has also been associated with complex IV deficiency, but these deletions of mitochondrial DNA (842), the most common mutation
cases are not well characterized. Complex V (mitochondrial ATP syn- being in the tRNALeu gene (815). The cause of the stroke-like events
thase) deficiencies are associated with clinical findings of both Leigh syn- is unknown. Currently, the disorder is believed to result from derange-
drome and NARP (neuropathy, ataxia, and retinitis pigmentosa), as well ments in cytochrome oxidase enzymatic function and consequent
as a milder condition known as familial bilateral striatal necrosis (819). effects on neuronal ATP levels (842).
Major and minor diagnostic criteria for mitochondrial disorders Imaging studies in the acute phase show swelling and T1 hypoin-
have been proposed (822,823), but no consensus has been reached. tensity and T2/FLAIR hyperintensity in the affected areas of the brain
However, certain clinical features are characteristic of many of the (Figs. 3-77 and 3-78), primarily the parietal and occipital cortex and

Barkovich_Chap03.indd 173 5/6/2011 12:09:00 AM


174 Pediatric Neuroimaging

subcortical white matter (840,843,844) (but any cortical area may atrophy then ensues (850). The lesions are not restricted to a specific
be affected) and in the basal ganglia (Fig. 3-77) (845,846). In some arterial distribution (Fig. 3-78)—indeed, single lesions often cross
patients, the dorsal brainstem and spinal cord are involved (847); vascular boundaries—helping to distinguish MELAS from embolic or
cerebellar atrophy may develop in late stages of the disease (833,848). thrombotic infarction. MR spectroscopy shows high lactate in affected
Sequential scans may show resolution and subsequent reappearance of areas of brain (Fig. 3-78C) (845,851,852). However, as infarcts of any
the abnormal areas (840,843,849) or, more commonly, development cause seem to result in local increases in lactate (853), the presence
of new lesions (Fig. 3-78). Eventually, T1 hyperintensity may appear of lactate in the region of an acute cortical lesion is certainly not spe-
in the affected cortex, indicative of permanent cortical damage, and cific for MELAS; the presence of lactate in areas of the brain that are

FIG. 3-77. Mitochondrial encephalopathy with lactic acidosis and strokelike episodes (MELAS). This teenage girl with a history of short stature and devel-
opmental delay presented with acute blindness. A. Axial noncontrast CT scan shows low attenuation (white arrows) in the occipital lobes. Other areas of low
attenuation (black arrows) are seen in the corpus striatum. B. Axial FLAIR image from the following day shows additional lesions (regions of hyperintensity)
in the left occipital pole (smaller white arrows) and left cingulate gyrus (larger white arrow). Notice that the left occipital lesion crosses vascular boundaries, a
characteristic of the cortical lesions of MELAS. C. Axial post contrast T1-weighted image shows enhancement of the left occipital and cingulate gyrus lesions,
in addition to the posterior putamina (small white arrows) and left superior frontal gyrus (large white arrow). D. Diffusion weighted image (b = 1,000) shows
reduced water motion (manifested as hyperintensity) in the left occipital pole (small white arrows) and superior frontal gyrus (large white arrow).

Barkovich_Chap03.indd 174 5/6/2011 12:09:00 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 175

FIG. 3-78. MELAS in a young child: sequential infarc-


tions. A and B. Axial T2-weighted image (A) and diffusion
weighted image (B) show hyperintensity (white arrows) in
the right parietal cortex, indicating subacute infarction.
C. Single voxel spectrum (TE = 288 ms) shows slightly
reduced NAA and a markedly enlarged lactate doublet
(Lac). D and E. Axial FLAIR (D) and diffusion weighted
image (E) 2 weeks later show new infarctions in the left
temporal lobe (white arrows) and pulvinar (white arrow-
head), regions with different vascular distributions.

Barkovich_Chap03.indd 175 5/6/2011 12:09:01 AM


176 Pediatric Neuroimaging

not visibly abnormal on T2 or diffusion images is more suggestive of The disorder is associated with mutations of the nuclear-encoded
mitochondrial disease. Short echo time MRS shows elevated glucose DNA polymerase-gamma gene (POLG) located at chromosome 15q25,
and severely reduced NAA, glutamate, and creatine (854). Reports although many reported patients have had mutations of mitochondrial
describing the findings of diffusion imaging in MELAS are conflict- DNA. There appears to be some overlaps of progressive external oph-
ing. Some authors report reduced diffusion in some affected areas thalmoplegia with MNGIE (see following section).
(855,856), while others report increased diffusion (857). This dif- The neuroimaging findings in Kearns-Sayre syndrome and oph-
ference may reflect the acuity of the lesion or its severity. In either thalmoplegia plus appear very similar. CT scans show cortical and
event, findings on diffusion images should not be used to differentiate white matter atrophy, hypodensity of cerebral and cerebellar white
MELAS from other causes of cortical injury at this time. Of interest, matter, and variable hypodensity or calcification of the deep cerebral
the size of the diffusion abnormality may increase over several weeks and cerebellar nuclei (59,868). It is not clear whether the calcification
on sequential studies (850). This progression and the crossing of vas- is the result of the primary disorder or the associated hypoparathyroid-
cular boundaries may be useful in separating MELAS from embolic or ism that often accompanies the syndrome (869). MR scans (Figs. 3-79
thrombotic infarcts. Perfusion studies show increased local blood flow and 3-80) show patchy T2 hyperintensity in the white matter, predomi-
(858,859) but decreased local glucose uptake and oxygen extraction nantly subcortical, with early involvement of the subcortical U fibers
fraction (860,861) in affected brain tissue in the acute phase. (Fig. 3-79) and sparing of periventricular areas (845,867). Later the
deep cerebral white matter and the deep gray matter nuclei, particu-
Kearns-Sayre Syndrome/Progressive External larly the dorsal midbrain, medial and posterior thalami, and the globi
Ophthalmoplegia pallidi, show T2 hyperintensity (Fig. 3-80) (831,845,867,870,871). The
Kearns-Sayre syndrome (OMIM 530000) and progressive external characteristic MR findings may not be present early in the course of
ophthalmoplegia (OMIM 258450, also called ophthalmoplegia plus) the disease (831). The affected white matter shows reduced diffusion
are discussed together because both are mitochondrial disorders char- (Fig. 3-79) (11), which may persist over several years (Fig. 3-80) (872).
acterized by progressive external ophthalmoplegia with cardiac and DTI in a single patient with Kearns-Sayre syndrome appeared to show
retinal manifestations. To establish the diagnosis of Kearns-Sayre syn- low fractional anisotropy and increased diffusivity compared to age-
drome, patients must have, as a minimum, external ophthalmoplegia, matched normals (873). Proton MR spectroscopy of affected regions
retinitis pigmentosa, and onset of neurologic or muscular dysfunction shows increased lactate and reduced NAA.
before the age of 20 years (811,814,838,839,862). Some authors require
elevated CSF protein (863), heart block (59,862), or cerebellar ataxia Mitochondrial Neurogastrointestinal Encephalomyopathy
(815) for clinical diagnosis. Patients may also have dementia, short Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE,
stature, sensorineural hearing loss, endocrine dysfunction, elevated OMIM 603041) is a multisystemic disorder clinically characterized
serum and CSF lactate, and muscle weakness (811,814,863,864). Many by severe gastrointestinal dysmotility (often pseudoobstruction),
different mitochondrial DNA deletions have been found in affected cachexia, progressive external ophthalmoplegia, peripheral neuropa-
patients (44,865). Progressive external ophthalmoplegia patients have thy, and leukoencephalopathy (874); the most common neurologic fea-
exercise-induced or permanent pareses, progressive external ophthal- tures are peripheral neuropathy, ptosis, ophthalmoparesis, and hearing
moplegia, peripheral neuropathy, hypoacusis, pyramidal and extrapy- loss (875). Average age at onset is 19 years, but varies from infancy to
ramidal symptoms, cerebellar syndromes, and dementia (866,867). the fifth decade (875). The disorder is caused by mutations in the gene

FIG. 3-79. Kearns-Sayre syndrome, early disease phase. A. Axial T2-weighted sequence shows T2 prolongation (black arrows) in the dorsal mesencephalon.
B. Axial T2-weighted image at a higher level shows mild subcortical T2 prolongation (white arrows). Note that the subcortical U fibers are involved even at
this early stage. C. Diffusion weighted image (b = 1,000) shows reduced water motion (manifested as hyperintensity, arrows) in the subcortical white matter.
D. Coronal FLAIR image again shows involvement of subcortical white matter with sparing of periventricular white matter (arrows). Note that the globi
pallidi are not involved at this stage of the disease.

Barkovich_Chap03.indd 176 5/6/2011 12:09:02 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 177

FIG. 3-79. (Continued)

FIG. 3-80. Kearns-Sayre syndrome, later disease phase. A and B. Axial T2-weighted images show abnormal hyperintensity in the midbrain (black arrows, A),
globi pallidi (white arrowheads, B) and subcortical white matter (white arrows, B). C. Axial diffusion weighted image shows abnormal hyperintensity in the sub-
cortical white matter (white arrows), indicated reduced diffusivity. D. Single voxel proton MR spectrum from the basal ganglia shows elevated lactate (Lac).

Barkovich_Chap03.indd 177 5/6/2011 12:09:03 AM


178 Pediatric Neuroimaging

encoding thymidine phosphorylase (TYMP, located on chromosome a number of causes that are likely to impair energy production (885).
22q13.32-qter) (876). A form without leukoencephalopathy can be Many disorders can result in Leigh syndrome; indeed, mutations of
caused by mutations of the DNA polymerase gamma gene (POLG) both mitochondrial and nuclear DNA, involving genes coding for pro-
at chromosome 15q25; this has led to the speculation that there is an teins in respiratory chain complexes I, II, III, IV, and IV, mitochondrial
overlap of mitochondrial-induced progressive external ophthalmople- tRNA, pyruvate dehydrogenase complex, and Coenzyme Q10 have
gia (see previous section, V.D.6.b) and MNGIE (877). Mitochondrial been shown to cause Leigh syndrome (3,44,886). Four groups seem to
deletions may be associated with MNGIE, as well (878). This discussion account for the large majority of cases; these and a few others of inter-
of Kearns-Sayre, PEO, and MNGIE nicely shows the difficulties associ- est will be discussed in some detail.
ated with organizing mitochondrial disorders into distinct clinical/genetic The first group is that with defects of the pyruvate dehydrogenase
syndromes. complex; biochemical defects can involve any of the three catalytic
Neuroimaging shows the presence of diffuse leukoencephalopa- subunits of the enzyme complex or the activation of the complex by
thy in patients with mutations of TYMP (875,879), but not in patients pyruvate dehydrogenase phosphatase (887,888). Pyruvate dehydroge-
with POLG mutations (877). CT of patients with leukoencephalopa- nase deficiency typically has the classic imaging findings described in
thy reveals diffuse hypodensity of cerebral and cerebellar white matter. Leigh syndrome with T1 hypointensity and T2/FLAIR hyperintensity
MRI shows diffuse high signal intensity of the periventricular and deep of the corpus striatum (caudate and putamen) and cortex in addition
cerebral white matter on T2-weighted and FLAIR images (Fig. 3-81); to delayed myelination (Fig. 3-82). Long echo time proton MRS shows
subcortical U fibers and corpus callosum are typically spared. Thalami large lactate peaks and small NAA peaks in affected regions. Diffusion-
and basal ganglia may show patchy T2 and FLAIR hyperintensity or weighted images show reduced water diffusivity in acutely affected
may be spared; in some patients, the hyperintensity extends caudally regions and increased diffusivity in chronically affected regions, which
into the internal and external capsules and brainstem (11,880). may show frank cavitation. However, cerebral dysgenesis can also be
seen, particularly in E1 alpha subunit (PDHA) deficiency, which is the
Disorders Causing Leigh Syndrome most common enzyme defect in PDH deficiency. This X-linked disor-
Leigh syndrome (OMIM 256000) refers to a symptom complex with der (PDHA1 is located at Xp22.2-p22.1) is associated with dysgenesis
characteristic, but variable, clinical and pathologic manifestations of the corpus callosum, absence of the medullary pyramids, ectopic
(338,881). Affected infants and children typically present toward the olivary nuclei, dysplasia of the cerebellar dentate nuclei, subcortical
end of the first year of life with hypotonia and psychomotor dete- heterotopia, and polymicrogyria (889,890).
rioration. Ataxia, ophthalmoplegia, ptosis, dystonia, and swallowing The second group has cytochrome oxidase deficiency (COX, respi-
difficulties almost inevitably ensue. Classic pathologic abnormalities ratory chain complex IV); this is currently believed to be the most com-
include microcystic cavitation, vascular proliferation, neuronal loss, mon cause of Leigh syndrome (891,892). The COX enzyme complex
and demyelination in the midbrain, basal ganglia, cerebellar dentate is composed of 13 structural subunits, some of which are encoded by
nuclei, and, occasionally, cerebral white matter (338,882–884). mitochondrial DNA, some by mitochondrial tRNA genes, and some by
Although it has been known for some time that several biochemi- nuclear DNA (891). Leigh syndrome is caused by a defect that is inher-
cal and genetic abnormalities cause Leigh syndrome, the different ited as an autosomal recessive trait. Mutations of SURF1, a gene located
genetic causes are only now being elucidated. The disorder seems to on nuclear chromosome 9q34 and important in the assembly and main-
be the result of defective terminal oxidative metabolism from any of tenance of cytochrome oxidase activity, are probably the most common

FIG. 3-81. Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) due to TYMP mutation.


Axial T2-weighted images (A and B) show diffuse leukoencephalopathy with relative sparing of corpus
callosum, internal capsules (arrows) and basal ganglia. Minimal thalamic involvement (white arrowheads)
is identified.

Barkovich_Chap03.indd 178 5/6/2011 12:09:04 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 179

FIG. 3-82. Leigh syndrome due to pyruvate dehydrogenase deficiency. Axial FLAIR images show abnor-
mal in the corpus stratum (large white arrows in A), posteromedial thalami (white arrowheads in A), and
several regions of cerebral cortex (small white arrows in B).

cause of this form of the disease, being responsible for as many as one hyperintensity in the subthalamic nuclei, medulla, inferior cerebel-
third of the COX deficiency patients (891,893–895). Affected patients lar peduncles, inferior olives and nucleus of the solitary tract in the
present near the end of the first year of life with progressive enceph- medulla, central tegmental tract and reticular formation in the dorsal
alopathy, hypotonia, ataxia, abnormalities of eye movement, swallow- pons, and periaqueductal gray matter of the mesencephalon in nearly
ing difficulties, and central respiratory problems (891,896). Mutations all cases (Fig. 3-83). Involvement of the substantia nigra, red nuclei, and
of other genes needed for COX assembly are associated with clinical cerebellar dentate nuclei are seen less commonly, while involvement of
variations of Leigh syndrome: SCO2 and COX15 genes with cardioen- the medial thalami is seen in some cases but is uncommon (896–899).
cephalopathy, SCO1 gene with hepatoencephalopathy, and COX10 with The acutely affected areas show reduced diffusion (Fig. 3-83D). In our
nephroencephalopathy (819). Imaging features of a number of patients experience, lactate is not always seen on proton MRS in these patients.
with SURF1 mutation associated with COX deficiency have been Of note, SURF1 mutation with COX deficiency has also been described
described (896–898). Early scans may be normal (898). As the disease with exclusive white matter involvement (900); this observation empha-
progresses, they develop characteristic T1 hypointensity and T2/FLAIR sizes the remarkable phenotypic variations of mitochondrial disorders!

FIG. 3-83. Leigh syndrome secondary to SURF1 mutation in a 15-month old child. A–C. Axial T2-weighted images through the brain stem show involve-
ment of the inferior olives (large black arrows in A), central tegmental tracts (black arrows in B), dorsal midbrain (large white arrows in C), red nuclei (small
white arrows in C), and subthalamic nuclei (white arrowheads in C). D. Axial diffusion weighted image (b = 1000) shows reduced diffusion (hyperintensity)
in most of the affected area of the midbrain.

Barkovich_Chap03.indd 179 5/6/2011 12:09:05 AM


180 Pediatric Neuroimaging

FIG. 3-83. (Continued)

A third group of mutations, which show maternal inheritance, causes of Leigh syndrome, however, is the nearly constant involvement
appear to arise from mutations to the mitochondrial ATPase 6 gene of the mamillary bodies and the frequent involvement of the frontal
(Complex V) (901–906). Mutations in this family may also be respon- cortex (917,919); the latter may be associated with poor prognosis
sible for clinically milder Leigh-like syndromes, such as bilateral stri- (919). Calculated Dav images will show reduced diffusivity in affected
atal necrosis (907,908) and NARP (neuropathy, ataxia, and retinitis areas in the acute phase. Proton MR spectroscopy reveals elevated lac-
pigmentosa) (909). These patients seem less homogeneous than the tate and decreased NAA in affected regions of the brain (917).
SURF1 group, but in limited experience have involvement of the ante- Complex II deficiencies account for about 2% of mitochondrial
rior putamina, dorsal mesencephalon, and dorsal pons (Fig. 3-84). encephalomyopathies (920). Leukodystrophy and Leigh syndrome have
A fourth group, which may be responsible for a large group of been among the wide range of presentations. Succinate dehydroge-
patients, is complex I deficiency (910–914). Few reports are available nase deficiency can present with either of these phenotypes (920,921).
regarding neuroimaging findings in this disorder, although extensive Patients typically present with acute neuromotor deterioration late
white matter cavitation has been reported in a few cases (Fig. 3-85) in the first year or early in the second year of life. MRI shows either
(832,914) and typical basal ganglia involvement in others (915). We basal ganglia disease typical of Leigh syndrome or T2/FLAIR hyper-
have noted callosal cavitation to be common (Fig. 3-85A), along with intensity of periventricular and deep cerebral white matter together
cerebellar atrophy (Fig. 3-85A) and high lactate levels on proton MRS with cerebellar white matter (Fig. 3-86A–C). Diffusivity is reduced in
(Fig. 3-85D). With the accumulation of more clinical and laboratory actively involved areas (Fig. 3-86D). Diagnosis can be confirmed by
data, more precise patterns of radiologic and clinical features will proton MR spectroscopy, which shows a large singlet (succinate) at
emerge, along with refinements in classification. 2.4 ppm in white matter (Fig. 3-86E) but not in gray matter. We have
A less common group is that of infants with thiamine deficiency. seen other disorders due to Complex II deficiencies with abnormalities
Although very rare in developed countries, infantile thiamine deficiency largely confined to the posterior fossa and sparing of supratentorial
is an important cause of Leigh syndrome because it can be treated; structures.
therefore, it should always be considered in the proper clinical setting, In summary, the patterns of injury in patients with Leigh syn-
particularly if the mamillary bodies are affected. Infants develop this drome vary with the mutation that is the cause of the syndrome; these
disorder when their diet is deficient in thiamine (vitamin B1) and they patterns are described in the previous paragraphs and summarized in
have a mutation of the gene coding for one of the enzymes that use thi- Table 3-17. Diffusion characteristics and proton spectroscopic char-
amine as a cofactor (611,916). The lack of thiamine is typically either acteristics vary depending upon the acuity of the lesion. In the acute
the result of being breastfed by mothers who were themselves thiamine phase of injury, when mitochondrial function is acutely impaired and
deficient or being fed infant formula lacking thiamine Affected infants ATP production acutely reduced, water diffusivity will be reduced. If no
typically present in the first year of life with gastrointestinal symptoms permanent damage ensues, diffusivity may return to normal. However,
(vomiting or diarrhea) accompanied by apathy, apnea, or seizures. if significant astrogliosis or necrosis ensues in the injured area, diffusiv-
Vertical nystagmus may be present of neurologic examination (917). ity will be increased. MR spectroscopy has shown decreased NAA and
Analysis of the CSF and serum at the time of admission will typically elevated lactate, with lactate elevation most pronounced in areas with
show elevated lactate. CT shows low attenuation of the frontal lobes or acute mitochondrial dysfunction (and consequent anaerobic glycoly-
basal ganglia (918). MRI shows the characteristic bilateral symmetrical sis) and in those areas most severely affected on the imaging studies
putaminal T2 hyperintensity of Leigh syndrome with frequent involve- (845,851,852). As other disorders that involve the basal ganglia may
ment of the medial thalami, periaqueductal gray matter, and dorsal not have increased basal ganglia lactate (922), the presence of lactate in
brainstem, as well. What distinguishes thiamine deficiency from other a patient with a characteristic imaging pattern supports the diagnosis

Barkovich_Chap03.indd 180 5/6/2011 12:09:06 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 181

FIG. 3-84. Neuropathy, ataxia, and retinitis pigmentosa (NARP).


A. Sagittal T2-weighted image shows abnormal hyperintensity (white arrows)
in the dorsal brainstem. B and C. Axial T2-weighted (B) and coronal FLAIR
(C) images shows abnormal diffuse and focal hyperintensity in the lentiform
nuclei (large white arrows), caudates (white arrowheads), and hypothalamus
(small white arrows). The extent of abnormality is better appreciated on the
FLAIR image. D and E. Axial diffusion weighted image (D) shows reduced
diffusivity in the cerebral peduncles (white arrowheads) and midbrain
tegmentum (white arrows), while axial postcontrast T1-weighted image
(E) shows enhancement (white arrows) in the areas that had reduced
diffusivity.

Barkovich_Chap03.indd 181 5/6/2011 12:09:06 AM


182 Pediatric Neuroimaging

FIG. 3-85. Leigh syndrome due to complex 1 deficiency. A. Sagittal T1-weighted image shows cavitation of the posterior body and splenium of the cor-
pus callosum (white arrows) and cerebellar vermian atrophy (white arrowheads). B. Axial T1-weighted image shows low signal intensity of periventricular
white matter (arrows and arrowheads), with more marked hypointensity, suggesting cavitation posteriorly (white arrows). C. Coronal FLAIR image shows
heterogeneous hyperintensity of periventricular and deep white matter, with some central areas of hypointensity (white arrows) indicating complete cavita-
tion. Cerebellar white matter (white arrowhead) is involved. D. Single voxel proton MRS from frontal white matter showing a low NAA peak and markedly
elevated lactate (Lac).

of Leigh syndrome. However, lactate is absent in up to 50% of patients blindness, and liver disease with micronodular cirrhosis is often diag-
with Leigh syndrome at the time of MRI (3), and the presence of lactate nostic (927). Onset is usually in the first few years of life, and may
is not specific for mitochondrial disorders (ischemia, infection, inflam- be as early as the first few weeks. Overt evidence of hepatic disease is
mation, and neoplasm can also cause elevated lactate). variable, but abnormalities of hepatic chemistry may be detected early
(925,929). Pathologically, the brain shows spongiform cortical atrophy,
Alpers Disease which is most pronounced in the occipital region, and atrophy of the
Alpers disease (OMIM 203700) is a rare autosomal recessive, progres- basal nuclei, particularly the thalami and globi pallidi (59,884,925).
sive multisystemic disorder characterized by predominant involvement Reports of imaging of patients with Alpers disease are few. CT find-
of the cerebral gray matter and the liver (824,884,923–925). The disease ings include focal hypodensities of both gray and white matter, followed
seems to be caused by mutations of the nuclear gene encoding mito- by diffuse atrophy (59,824). On MRI, authors note T2 hyperintensity
chondrial DNA polymerase gamma, POLG1, located on chromosome in cortex, subcortical white matter and basal ganglia, diminished white
15q25 (926,927). Patients typically present with intractable seizures, matter, delayed myelination, and cortical thinning that is most severe
particularly myoclonic jerks, following some early developmental delay in the frontal, posterior temporal and occipital lobes (845,925,930);
or failure to thrive (824,884,925,928). Identification of the clinical tet- progression of the disorder is often rapid (931). No reports of MRS in
rad of refractory seizures, episodic psychomotor regression, cortical Alpers disease have been published.

Barkovich_Chap03.indd 182 5/6/2011 12:09:08 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 183

FIG. 3-86. Leigh syndrome due to succinate dehydrogenase deficiency (complex


II). A–C. Axial T2-weighted images show extensive T2 hyperintensity in the middle
cerebellar peduncles and cerebellar white matter (white arrowheads, A), corpus cal-
losum (small white arrows, B), periventricular and deep cerebral white matter (large
white arrows, B and C). D. Axial average diffusivity (Dav) image shows reduced Dav
in the corpus callosum (small black arrows) and corticospinal tracts (black arrow-
heads) but increased diffusivity (hyperintensity) in the deep cerebral white matter.
E. Single voxel proton MRS from the frontal white matter shows reduced NAA and
a large singlet at 2.4 ppm representing succinate (Su). Succinate was not seen in gray
matter voxels. (This case courtesy Dr. Vernon Byrd, Charlotte.)

Barkovich_Chap03.indd 183 5/6/2011 12:09:09 AM


184 Pediatric Neuroimaging

TABLE 3-17 Causes and Imaging Characteristics in Leigh Syndrome


Cause Imaging Characteristics
Pyruvate dehydrogenase complex deficiency Involvement of corpus striatum and, in infants, thalami. White matter cavities.
Sometimes malformations.
Cytochrome Oxidase deficiency with SURF1 mutation Subthalamic nuclei, periaqueductal gray matter, central tegmental tract,
cerebellar nuclei, cerebellar peduncles, inferior olivary nuclei
Complex V deficiency with mtATPase6 mutation Anterior putamina, globi pallidi, dorsal mesencephalon and pons
Infantile thiamine deficiency Frontal cortex, mamillary bodies, putamina, periaqueductal gray matter, dorsal
brainstem
Succinate dehydrogenase deficiency Periventricular and deep white matter in all cerebral lobes and cerebellum.
Corpus callosum may cavitate. MRS shows large singlet at 2.4 ppm (succinate)
Complex I deficiency with MTND1 mutation Subthalamic nuclei, corpus striatum, dorsal brain stem (especially medulla) (913)
Other Complex I deficiencies Callosal and white matter involvement with cavitation. May have cerebellar atrophy.

Trichopoliodystrophy (Menkes Disease) life (938). Pathologic examination shows diffuse atrophy of cerebral
Trichopoliodystrophy (932) (OMIM 309400) is an X-linked recessive and cerebellar hemispheres with thin-walled, tortuous cerebral arter-
mitochondrial disorder that results from impaired intestinal absorption ies (884). Microscopic examination shows widespread spongiform
of copper with consequent impairment of cytochrome oxidase activity degeneration of the gray matter, sometimes exhibiting frank cavita-
in the mitochondria (cytochrome c contains two copper atoms (884) ). tion. The volume of white matter is reduced and the white matter is
The responsible ATP7A gene, which codes for a copper transporting hypomyelinated (884).
ATPase called Menkes protein (MNK), has been localized to Xq13.3 Imaging findings in Menkes disease are nonspecific, but as a con-
(933,934). Generically, the disease is related to a defect of “transmem- stellation of findings are rather characteristic (939–942). Skeletal sur-
brane copper transport mechanism,” which has several consequences veys show bones that are osteoporotic with flared metaphyses of long
at different critical levels of copper trafficking within the organism. bones, rib fractures and Wormian bones in the skull (Fig. 3-87A). Rapid
Impairment of normal intestinal absorption of copper causes cop- progression of atrophy, with resultant subdural hematoma formation
per deficiency in blood (ceruloplasmin level is also low) and copper and T1 hyperintensity/T2 hypointensity in the cerebral cortex, has
accumulation within the organelle-free cytosol of epithelial cells of the been observed by neuroimaging, (Fig. 3-87C) (845,942–944). Cerebral
small intestine. Transport of copper is impaired at the cellular level, as arteries are elongated and tortuous (Fig. 3-87B). An important point
well (MNK is localized to the trans-Golgi network and is required for in this respect is that rapid brain atrophy in the presence of large bilat-
normal transport of copper within the cell); hence, copper cannot be eral subdural hematomas and apparent cortical blood is not necessarily a
delivered to those enzymes requiring copper as an essential cofactor. manifestation of asphyxic or physical trauma and that trichothiodystro-
Therefore, even if copper is present within the cytosol, intraorganelle phy should be a consideration in such cases.
(in particular intramitochondrial) copper concentration is severely
depleted. Specifically, from a neurologic perspective, copper in the Glutaric Aciduria Type I (Glutaryl-CoA Dehydrogenase
brain is trapped within endothelial cells of vessels and astrocytes, while Deficiency)
neurons are in a state of copper deficiency. Thus, oral or even intrave- Glutaric aciduria Type I (GA1, OMIM 231670) is an autosomal reces-
nous copper supplementation is ineffective in the disease. sive disorder caused by deficiency of glutaryl-CoA dehydrogenase, an
Early diagnosis is difficult, but the decreased activity of dopamine- enzyme located in mitochondria and involved in the metabolism of
b−hydroxylase causes elevation of the ratio of dihydroxyphenylacetic L-lysine, L-hydroxylysine, and L-tryptophan. The responsible GCDH
acid to dihydroxyphenylglycol in plasma, which may aid early diagno- gene is located at chromosome 19p13.2 and contains 11 exons. Muta-
sis and, hopefully, treatment (935). Affected patients are often born tions of different exons can result in different phenotypes. It is proposed
prematurely and typically present in infancy with truncal hypotonia, that glutaric acid and 3-hydroxyglutaric acid, which accumulate in the
hypothermia, failure to thrive, and seizures (884,932). In milder cases, brain in this disorder, affect cell function and eventually lead to the
prominent cerebellar ataxia, dysarthria, and moderate cognitive defi- acute striatal damage that is commonly seen in this disorder (945), pos-
cit are noted, but seizures are absent (936). Head circumference at sibly due to excitotoxicity by overstimulation of N-methyl-D-aspartate
birth may be normal or decreased; a reduction with respect to the receptors (946). Patients may initially present with an acute encephal-
normal growth curve soon becomes evident. Patients have coarse, opathy, with macrocephaly, or with gradual neurological deterioration,
stiff, sparse hair with broken, nodular, frayed ends (884,932); hence including hypotonia, progressive dystonia or choreoathetosis, and
the disease is referred to as “kinky hair disease.” Their skin is hypop- tetraplegia (947). Regardless of initial presentation, most patients will
igmented, hyperextensible, and their joints are hypermobile. Some have an acute encephalopathic crisis by age 18 months (range: neonate
patients develop the “occipital horn syndrome,” in which a wedge- to 37 months), usually triggered by a febrile illness with some degree
shaped calcification forms within the tendinous insertions of the of dehydration (946). Following the acute illness, most motor skills
trapezius and sternocleidomastoid muscles at their attachment to the have been lost and a severe dystonic dyskinetic movement disorder is
occipital bone (936,937). Most patients die before the third year of present (948). Intellectual function can be relatively unaffected (948).

Barkovich_Chap03.indd 184 5/6/2011 12:09:10 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 185

FIG. 3-87. Trichopoliodystrophy (Menkes kinky hair


disease). A. Axial CT image (bone algorithm) at age
11 weeks shows multiple Wormian bones (white arrows)
in the posterior calvarium. B. Axial T2-weighted MR
image shows prominent CSF spaces and multiple tor-
tuous vessels (black arrows) in and around the circle of
Willis. C. Follow-up T1-weighted image at age 5 months
shows profound atrophy with enormous bilateral sub-
dural hematomas. (These images courtesy Dr. Susan
Blaser, Toronto.)

Rarely, patients remain asymptomatic into adulthood (949). Pathology hyperintensity (954). Central tegmental tracts along the floor of the
shows neuronal loss and astrogliosis of the basal ganglia in addition to fourth ventricle often show bilateral symmetrical T2 hyperintensity, as
spongiform changes in the cerebral white matter (838,947). well (954). Chronic subdural hematomas are seen in 20% to 30% of
Neuroimaging findings are important to suggest the diagnosis and affected patients, typically after relatively minor trauma, and are often
direct the biochemical analysis. Imaging findings include large bilateral accompanied by retinal hemorrhage (947,948). Although it is impor-
frontotemporal CSF spaces (open Sylvian fissures) that resemble bilat- tant to rule out child abuse in these children (especially if retinal hem-
eral anterior sylvian/temporal arachnoid cysts (Figs. 3-88 and 3-89) but orrhages are also present), it is also important to recall that patients
are a result of hypoplasia of the frontal and temporal opercula (946). with large subarachnoid spaces or large arachnoid cysts are more likely
These may be detected prenatally or in the early prenatal period by to develop subdural hematomas from relatively minor trauma (see
ultrasound (950). T2/FLAIR hyperintensity is seen in the basal ganglia Chapter 8). Ultimately, diffuse cerebral atrophy with frontotemporal
(most obviously in the putamen and caudate, and less conspicuously in predominance develops.
the globus pallidus) and white matter (Figs. 3-88 and 3-89); the white Although none of the above conventional imaging findings is
matter changes may not be seen early in course of the disease, and are specific for glutaric aciduria type 1, the constellation of “open Sylvian
usually seen only after at least one acute event of clinical decompensa- fissures” in conjunction with bilateral basal ganglia disease in a mac-
tion (Fig. 3-89) (946). Myelination is delayed (947,951–953). As the rocephalic child presenting with dystonia, is highly suggestive of the
disease progresses, the basal ganglia become atrophic and the corti- disease. It is noteworthy that macrocephaly and Sylvian fissure abnor-
cal sulci may enlarge. Abnormalities are often seen within the upper malities are present in both benign and malignant clinical phenotypes
midbrain with a pattern reminiscent of the so-called giant panda face (and are not affected by medical treatment). Therefore, the finding of
traditionally described in Wilson disease. During metabolic crisis, the Sylvian fissure abnormalities in an infant with macrocephaly (even with-
cerebellar nuclei may also be abnormal and present with T2/FLAIR out basal ganglia lesions), especially in ethnic groups with known high

Barkovich_Chap03.indd 185 5/6/2011 12:09:10 AM


186 Pediatric Neuroimaging

FIG. 3-88. Glutaric aciduria Type 1 in a 5-day old infant. A–C. Axial T1 (A), T2 (B) and average diffusivity (C) images show abnormal signal intensity in the
posterior putamina (white arrows), compatible with acute/subacute injury. Note the enlarged sylvian fissures (f) due to hypoplasia of the frontal and tempo-
ral opercula. D. Coronal FLAIR image shows germinolytic cysts (black arrows) in addition to the abnormally hyperintense putamina (black arrowheads).

disease prevalence, should prompt a targeted laboratory work-up for GA1 NAA (955). In the chronic phase, low NAA is found without lactate
in order to prevent the devastating consequences of a possible meta- (946).
bolic crisis and the resultant neurological crippling from basal gan- 18F-fluorodeoxyglucose PET scanning in glutaric aciduria type
glia damage. Intervention and modificaiton of the course of disease 1 shows diminished glucose uptake in the basal ganglia, thalami, the
is usually in the form of special diets which limit intake of lysine and insula, and the temporal opercular cortex (956). It is not clear what
tryptophan. relationship the diminished metabolic activity of the opercular region
Advanced imaging techniques may have value in the diagnos- has to the enlargement of the sylvian fissures and anterior middle
tic imaging workup of GA1. Diffusion-weighted imaging may show cranial fossa.
reduced diffusion in the white matter and reduced or increased dif-
fusion in the deep gray nuclei (955), dependent upon the stage of the Glutaric Aciduria Type II (Multiple Acyl CoA Dehydrogenase
disease; reduced diffusion is seen after acute clinical decompensation, Deficiency)
whereas increased diffusion is seen in the more chronic phases of the Glutaric aciduria Type II, also known as multiple acyl CoA dehydro-
disease. Proton MR spectroscopy findings probably also vary with the genase deficiency or MADD (OMIM 231680), results from a defect of
stage of the disease. Lactate is slightly elevated in the white matter the mitochondrial electron transport chain at coenzyme Q. It can be
during and in the early subacute phase after decompensation (955). caused by mutations to at least three different genes: ETFA at 19q13.3,
In the late subacute phase, MRS shows increased choline and reduced ETFB at 15q23-25, and ETFDH at 4q32-qter. It differs from glutaric

Barkovich_Chap03.indd 186 5/6/2011 12:09:10 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 187

FIG. 3-89. Glutaric aciduria Type


1 in an infant with developmental
delay and macrocephaly. A. Axial
T2-weighted image shows enlarged
sylvian fissures (black arrows) due to
hypoplasia of the temporal opercula
and abnormal hyperintensity of the
central tegmental tracts (black arrow-
heads). B. Axial T2-weighted image
shows abnormal hyperintensity of
the basal ganglia (black arrows) and
the periventricular and deep white
matter (white arrows), classic find-
ings when seen in conjunction with
the enlarged sylvian fissures.

aciduria type I in that multiple acyl-CoA dehydrogenase deficiencies finding suggest that abnormal mitochondrial function began during
result in large excretion not only of glutaric acid but also of lactic, eth- the period of organogenesis.
ylmalonic, butyric, isobutyric, 2-methyl-butyric, and isovaleric acids.
This disorder can result from deficiency of any one of three molecules: Friedreich Ataxia
the alpha and beta subunits of electron transfer flavoprotein and elec- Friedrich ataxia is discussed in the section on Metabolic Disorders Pri-
tron transfer flavoprotein dehydrogenase. It has been proposed that marily involving the Cerebellum.
these be called glutaric aciduria types IIa, IIb, and IIc, respectively
(957). Affected patients may present as neonates or infants, usually Ethylmalonic Encephalopathy
with hypoglycemia, hypotonia, and acidosis; facial dysmorphism and Ethylmalonic encephalopathy (OMIM 602473) is an extremely rare
visceral anomalies may be present. Those presenting in infancy rarely autosomal recessive disorder. Only about forty cases have been identi-
survive beyond a few weeks. Those presenting later in infancy are fied worldwide, mostly in Mediterranean and Arab populations. It is
managed by dietary means in an attempt to prevent the progression of characterized by increases in ethylmalonic, methylsuccinic, and lactic
the disease (838,958). Some reports of adolescent or adult presentation acids, which are found in the serum. The cause is mutations of a gene
are published (959); these older patients are reported to have move- called ETHE1, located at chromosome 19q13.32 (963). Most identified
ment disorders (960). mutations show loss of function of the protein product, ETHE1, which
Imaging studies show basal ganglia involvement (838,960). is targeted to mitochondria and, after cleavage of a short leader peptide,
Patients we have seen with glutaric aciduria type II showed T2 pro- is internalized into the mitochondrial matrix (963). Although the exact
longation in the caudate head, posterior putamen, and cerebral function of the ETHE1 protein is not known, the severe consequences of
hemispheric white matter. We have also seen T2 prolongation in the its malfunction suggest that its function is critical for energy production.
periatrial white matter, middle cerebral peduncles, callosal splenium, Known clinical phenotypes include neonatal onset with acute metabolic
and corpus striatum; in one patient, reduced diffusion was present crisis, and early or late infantile onset with slowly progressive enceph-
only in the white matter sites, not in the corpus striatum (Fig. 3-90). alopathy with or without metabolic decompensation. Patients with the
Other reports describe hypoplasia of the temporal lobes and, in one slowly progressive phenotype present with neuromotor delay, pyrami-
instance, absence of the cerebellar vermis (961,962). Proton MRS dal and extrapyramidal signs, ataxia and dysarthria (964,965). The most
shows elevated lactate and high Ch/Cr (indicating dysmyelination) prominent systemic manifestation of the disease is vasculopathy (tortu-
(958,962). ous retinal veins, usually not present at birth), cutaneous petechiae and
ecchymoses, orthostatic acrocyanosis, persistent microhematuria and
Neonatal Lactic Acidosis, Complex I/IV Deficiency, chronic diarrhea. The disease leads to death by early childhood in most
and Fetal Cerebral Disruption cases, the major cause of death is complications related to vasculopathy.
Patients with neonatal lactic acidosis, complex I/IV deficiency, and fetal Few imaging reports of patients with ethylmalonic encephalopathy
cerebral disruption present in the neonatal period with seizures and have been published. Some affected patients appear to have patchy T2
encephalopathy. They are usually small in stature and have small heads and FLAIR hyperintensity in the basal ganglia, cerebral white matter,
(<3rd percentile) and dysmorphic features (770). Antenatal polyhy- and sometimes brainstem, findings typical of mitochondrial disorders
dramnios may be detected. TORCH infection may be suspected. Anal- (963,966). Progressive atrophy is seen on sequential studies (967).
ysis of mitochondria shows severe combined deficiency of complexes Another report found CNS malformations: tethered cord and Chiari
I and IV in muscle and liver. Imaging shows multiple anomalies includ- I malformation (968).
ing subcortical heterotopia with callosal hypogenesis (see section on
subcortical heterotopia in Chapter 5), subependymal (germinolytic) Nonspecific Mitochondrial Disorders
cysts, subcortical calcifications, and abnormally hyperintense (on As discussed in the opening paragraph of this section, some brain disor-
T2-weighted images) in the brainstem and cerebellum (770) ). These ders that are associated with mitochondrial dysfunction do not fit into any

Barkovich_Chap03.indd 187 5/6/2011 12:09:11 AM


188 Pediatric Neuroimaging

FIG. 3-90. Glutaric aciduria type II. A–D. Clockwise from upper
left, echo-planar T2 (EP T2), diffusion-weighted (DWI), diffusiv-
ity image (ADC), and diffusion weighted images (DI) show T2
prolongation and reduced diffusion in the lateral pons bilater-
ally. E–H. Echo-planar T2, diffusion-weighted, apparent diffusion
image, and diffusion images at the level of the lateral ventricles
show T2 prolongation in the basal ganglia, periventricular white
matter, and splenium of the corpus callosum. However, reduced
diffusion (dark on ADC, bright on DI) is only seen he at the
periphery (arrows) of the white matter lesions, suggesting that
most of the T2 prolongation is chronic, not acute.

of the syndromes that have been described or have overlapping features Imaging findings are nonspecific with increased water (low attenu-
of multiple mitochondrial “syndromes.” These nonspecific mitochon- ation on CT, T1 hypointensity and T2/FLAIR hyperintensity on MRI)
drial disorders can present in patients of any age, from neonates to senior in the brainstem, deep gray matter nuclei, cerebral/cerebellar white
citizens (3,4,816,969,970). Presenting signs and symptoms are variable, matter, cerebral or cerebellar cortex (831), indistinguishable from the
as in all mitochondrial disorders (3,4,811,816,845), with seizures, short images of the other disorders discussed in this section. Remember that
stature, mental deterioration, muscle weakness, exercise intolerance, and mitochondrial disorders can have a broad range of appearances, from
neurosensory hearing loss being the most common (811–813,824). isolated brainstem involvement (Fig. 3-91) (899) to isolated cerebellar

Barkovich_Chap03.indd 188 5/6/2011 12:09:12 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 189

FIG. 3-91. Nonspecific mito-


chondrial disorder with iso-
lated brain stem involvement.
Axial T2-weighted images show
abnormal hyperintensity (black
arrows) in the pons (A) and mid-
brain (B) with normal appearing
cerebellum.

atrophy (Fig. 3-92) (971,972) to isolated white matter involvement that mutations of other complexes in the respiratory chain, is a common
may include small to moderate sized white matter cysts (832,914,973). cause of Leigh syndrome (978). Clinical phenotypes can be divided
As illustrated many times in this section, diffusion imaging and spec- into three major presentations (neonatal, infantile, or benign) (979)
troscopy may be useful to differentiate acute metabolic injury from that, as stated earlier, depend on the residual enzyme activity (974).
subacute or chronic injury. Neonatal presentation is characterized by hypotonia, episodic apnea,
convulsions, weak suck, dysmorphic features (broad nasal bridge,
Pyruvate Dehydrogenase Deficiency upturned nose micrognathia, posteriorly rotated ears, short fingers
The pyruvate dehydrogenase complex is a large multienzyme com- and arms, hypospadias), lethargy, low birth weight, failure to thrive,
plex that catalyzes the oxidative decarboxylation of pyruvate to acetyl and coma (819). Some patients may have life threatening congenital
CoA in the mitochondrial matrix. It is composed of five enzymes, lactic acidosis or acute flaccid paralysis (980). The infantile pheno-
containing three major subunits (E1, E2, and E3), all of which have type usually has onset between ages 3 and 6 months with psycho-
smaller subunits within them. It is becoming clear that mutations of motor retardation, hypotonia, convulsions, episodic apnea, ataxia,
the alpha subunit of E1 (located on chromosome Xp22.1) are, by far, pyramidal tract signs, decelerating head growth, ophthalmoplegia,
the most common cause of human metabolic disease (974). However, optic atrophy, peripheral neuropathy, dysphagia, and cranial nerve
the clinical manifestations of the disease vary widely depending on palsies (819). Other infants and children have benign syndromes such
the precise mutation (and its effect on enzyme function) and, when as intermittent weakness or ataxia (often associated with intermittent
present in women, the degree of inactivation of the affected X chro- lactic acidosis) or episodic dystonia (975), and still others may have
mosome. Overall, symptoms seem to correlate pretty well with the nonspecific signs or symptoms, such as failure to thrive, hypotonia,
residual activity of the enzyme complex (974). Uncommonly, muta- or developmental delay (978). Most commonly, patients make little
tions of the E2 (975) or E3 (976,977) binding protein component of or no subsequent developmental progress. Lactic acidosis is typically
the pyruvate dehydrogenase complex can cause disease. As stated in found at the time of clinical presentation, with a normal lactate/
the previous section, pyruvate dehydrogenase deficiency, along with pyruvate ratio.

FIG. 3-92. Mitochondrial disor-


der resulting in isolated cerebellar
atrophy. Sagittal (A) and coronal (B)
T1-weighted images show shrunken
cerebellar cortex with widening of
cerebellar sulci.

Barkovich_Chap03.indd 189 5/6/2011 12:09:13 AM


190 Pediatric Neuroimaging

Neuroimaging by MRI shows findings typical of Leigh syn- the degree to which the patient sustains some capacity for elimination
drome (see examples in previous section on mitochondrial disorders, of nitrogen waste products (987). With severe disturbance of molecular
particularly Fig. 3-82) in about half of affected patients (41,981). In function, patients become symptomatic as neonates, within the first few
other patients, MRI shows atrophy of varying degrees with injury to days of life, with progressive irritability, lethargy, poor feeding, hypo-
the cerebrum and cerebellum or, rarely, frank cerebral malformation, thermia, and seizures; neurodevelopmental outcome is related to the
the most common being callosal agenesis. (Pyruvate dehydrogenase duration of neonatal hyperammonemic coma (988). Those patients
deficiency is one of the few inborn errors of metabolism that can result with better molecular function present later (in childhood, adolescence,
in brain malformations). Correlations between the genetic defect and or adulthood) and typically have intermittent neurologic dysfunction
the imaging appearance have not yet been fully established, although that may present as movement disorders, seizures, ataxia, lethargy, or
patients with mutations limited to the E1a or E2 component of the confusion; in the mildest cases, the symptoms may be psychiatric ones
pyruvate dehydrogenase complex may have abnormalities limited to the such as hyperactive behavior, mood disturbances, or psychosis (986).
globi pallidi (975,980). Delayed myelination can be seen, as can frank Episodes can be triggered by anesthesia for minor surgery (988). In
leukodystrophy (981). Studies in some patients show primarily white ornithine carbamyl transferase deficiency, which is X-linked dominant
matter disease involving cerebellum, posterior limb of the internal (Table 3-18), girls are variably affected and boys always severely affected
capsule, and occipital white matter (41). Other patients have multiple (989). Improved therapy has lead to better long term outcome, but most
cysts in the cerebral white matter that may appear prenatally or post- survivors have developmental disabilities (986).
natally and seem to be the result of necrosis (982); ventriculomegaly In our experience, similar patterns of injury are seen with OCTD,
often ensues (983). Because damage often begins prenatally, diffusivity CPSD, and citrullinemia (990), and possibly with argininosuccinic
of affected white matter is often increased at the time of imaging (983). aciduria and hyperargininemia. The CT pattern in affected patients is
Proton MRS almost always shows elevated lactate (981,983) (and may nonspecific, with a pattern of diffuse edema (Fig. 3-94A), particularly
show a small pyruvate peak at 2.36 ppm (984) ); therefore, a finding of in infancy, as a result of hyperammonemia. Images later in the course
callosal agenesis in a patient with severe lactic acidosis strongly sug- of the disease may show both focal and diffuse areas of hypodensity
gests a diagnosis of LDH deficiency. on CT. On MRI, the appearance is somewhat more specific, as certain
regions of the cerebral cortex and deep gray matter show T1 hypoin-
Disorders of the Urea Cycle and Ammonia tensity and T2/FLAIR hyperintensity; in particular, the insular cortex
Disorders of the urea cycle result from disruption of the major pathway (posterior more than anterior), perirolandic cortex, and basal ganglia
for the disposal of nitrogen in the body; they include ornithine carbamyl (particularly the globi pallidi) show early T2 hyperintensity and swell-
transferase deficiency (ornithine transcarbamylase deficiency), carbamyl ing (Figs. 3-94 and 3-95). In affected neonates, T1 hyperintensity of
phosphate synthetase deficiency, argininosuccinic aciduria (arginino- affected deep gray and cortical structures will appear within the first
succinate lyase deficiency), citrullinemia (argininosuccinate synthetase few days of life (Fig. 3-96). Care should be taken not to misdiagnose
deficiency), and hyperargininemia (arginase deficiency) (985,986). All hypoxic-ischemic injury in these children; the differentiation can be
result in hyperammonemia and elevation of glutamine levels that may made by the predominant globus pallidus and putaminal injury in
be worsened by high protein intake or illness. Differentiation is made urea cycle disorders, in contrast to the predominant thalamic injury
by biochemical and genetic testing (Table 3-18). The timing of onset of in neonatal hypoxia-ischemia. In older patients, the appearance of the
these disorders depends upon the nature of the molecular defect and cerebral cortex in the acute phase shows T2 hyperintensity and swelling

TABLE 3-18 Differentiation of Urea Cycle Disorders


Disease/OMIM Number Gene/Locus Enzyme Defect Biochemical Features
CPTD/237300 CPS1 2q35 Carnitine palmityl transferase No citrulline in plasma
No orotic aciduria
OTCD/311250 OTC Xp21.1 Ornithine transcarbamylase Orotic aciduria
No citrulline in plasma
Citrullinemia types I ASS 9q34.1 Argininosuccinic acid synthetase High plasma citrulline, ammonia,
(neonatal) and III alanine. High urinary orotic acid
(infantile)/215700
Citrullinemia SLC25A13 7q21.3 Argininosuccinic acid synthetase High plasma citrulline, ammonia
type II/605814 in liver
Argininosuccinic ASL 7cen-q11.2 Arginosuccinase High plasma argininosuccinate.
Aciduria/207900 Moderate citrulline in plasma
Hyperargininemia/207800 ARG1 6q23 Arginase Elevated plasma arginine
OMIM, Online Mendelian Inheritance in Man; OCTD, ornithine carbamyl transferase deficiency; CPTD, carbamyl phosphate synthetase deficiency.

Barkovich_Chap03.indd 190 5/6/2011 12:09:14 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 191

FIG. 3-93. Mitochondrial dis-


order with white matter cavita-
tion. Sagittal T1-weighted image
(A) shows hypointensity (black
arrows) indicating cavitation in
the posterior body and splenium
of the corpus callosum. Cavita-
tion in the periventricular and
deep white matter is verified by
the hypointensity of the affected
white matter (black arrows) on the
coronal FLAIR image (B). Cer-
ebellar atrophy is present, as well.

FIG. 3-94. Ornithine transcarbamylase


deficiency in a 7-year-old child. A. Axial
noncontrast CT scan shows diffuse edema
manifested as sulcal effacement and low
attenuation of the cerebral cortex resulting
in loss of cortical-white matter contrast.
B. Axial T2 weighted image shows edema
manifested as hyperintensity and thicken-
ing of the insula and frontal lobe cortex.
C. Coronal FLAIR image also shows hyper-
intensity of the insular cortex (white arrow-
heads), as well as temporal cortex (large
white arrows) and cingulate sulci (small
white arrows). D. Follow up study shows
development of diffuse cerebral atrophy.

Barkovich_Chap03.indd 191 5/6/2011 12:09:14 AM


192 Pediatric Neuroimaging

FIG. 3-95. Citrullinemia in a 5-year-old child. A and B. Axial T2-weighted images show edema in the globi pallidi (white arrows), insular cortex and
underlying external and extreme capsules (white arrowheads), left cingulate cortex, and medial left frontal cortex. C. Axial diffusion weighted image shows
reduced diffusivity in the insulae (white arrowheads) and left cingulate cortex (white arrow). D. Single voxel proton MRS (TE = 30 ms) shows reduced NAA
and increased glutamate/glutamine (Glx, at 2.1–2.5 and 3.8 ppm) in the basal ganglia.

of the cortex (Figs. 3-94 and 3-95) (990); the insulae and cingulate gyri Methylmalonic and Propionic Acidemias
are most commonly affected and, in general, the frontal lobes seems
Both methylmalonic and propionic acidemia are autosomal recessive
more affected than the parietal, occipital, or temporal lobes. The peri-
disorders that cause ketoacidosis and excretion of the respective acids in
rolandic and occipital cortex seem to be specifically spared (990). As
the urine. Propionic acidemia (OMIM 606054) is caused by mutation in
the disease progresses, atrophy develops; it is often severe and may be
the genes encoding propionyl-CoA carboxylase, PCCA (at chromosome
multicystic. Two characteristics of brain involvement are noteworthy:
13q32) or PCCB (at chromosome 13q21-23) (994). Methylmalonic aci-
(a) the subcortical U fibers are not spared–the subcortical white mat-
demia (OMIM 251000) is caused by mutation of the gene (MUT) encod-
ter is involved severely and early (991) and (b) cerebral involvement is
ing methylmalonic CoA mutase, located at chromosome 6p21 (995),
often slightly asymmetric. Proton MRS in both of these disorders in
which functions with its coenzyme cobalamin (vitamin B12) to convert
the acute or subacute phase shows lactate (a doublet centered at 1.33
methylmalonyl-CoA to succinyl-CoA. Both diseases often present early
ppm) and of glutamine/glutamate resonances (at 2.1–2.5 ppm, just
in life with episodes of metabolic acidosis, vomiting, tachypnea, leth-
downfield from NAA, and at 3.75 ppm, also attributable to glutamine
argy, and seizures, often leading to coma and death. Survivors are typi-
(992,993) ). Because of the short T2 relaxation times, the glutamine/
cally small with small heads; they suffer from quadriparesis, movement
glutamate peaks are best seen with short echo time (20–30 ms) STEAM
disorders, and psychomotor retardation with episodic vomiting, keto-
sequences (Fig. 3-95) (992).

Barkovich_Chap03.indd 192 5/6/2011 12:09:16 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 193

FIG. 3-96. Ornithine transcarbamylase deficiency in a neonate. A and B. Axial T1-weighted images show hyperintensity in the lentiform nuclei, particu-
larly the globi pallidi (small white arrows), insular cortex, and perirolandic cortex, in addition to hypointensity of the caudate heads (large white arrows).
C. Axial T2-weighted image shows diffusely abnormal hyperintensity of the white matter (which is isointense to CSF) and the basal ganglia, indicating dif-
fuse edema. D. Proton MR spectrum (TE = 288 ms) shows an abnormal lactate peak (Lac) and an abnormal glutamate peak (Glx).

sis, and coma (40,284,838,996). Other patients present with progressive T2 hyperintensity (MRI) is sometimes observed in the periventricu-
encephalopathy with central hypotonia of varying severity, sometimes lar white matter (40,59,1000,1001); myelination delay is present early
associated with movement disorders or dystonia (40,997,998). Neuro- and both cortical and white matter volume loss is often present in later
logic examination reveals central hypotonia with pyramidal tract signs stages. In the acute phase of a clinical decompensation reduced diffu-
and symptoms at time of crises; dystonia and choreoathetosis are com- sivity may be seen in the affected regions, likely a result of mitochon-
mon sequelae of basal ganglia involvement (999). drial dysfunction (1002).
CT and MRI reveal increased water (low attenuation and T1 Proton MR spectroscopy in propionic aciduria reportedly shows
hypointensity, T2/FLAIR hyperintensity), most commonly in the decreased NAA and myo-I and increased glutamate/glutamine in the
white matter and globi pallidi in methylmalonic acidemia (Fig. 3-97) basal ganglia (999); reduced NAA is also reported in MMC (1002).
and in the putamina and caudate nuclei in propionic acidemia (Fig. Lactate elevation was present in one reported case (1002). PET studies
3-98) (40,996,997,1000,1001). CT often shows calcification of the show increased uptake of 18-fluoro-2-deoxyglucose early in the course
basal ganglia in MMC (1001), and we have seen cystic changes of the of the disease (first year of life). Later, in the second and third years,
globi pallidi on MRI (Fig. 3-97B). Abnormal hypodensity (CT) and decreased uptake is noted in the basal ganglia (998).

Barkovich_Chap03.indd 193 5/6/2011 12:09:16 AM


194 Pediatric Neuroimaging

FIG. 3-97. Methylmalonic aciduria;


13-month-old child with developmen-
tal delay. A. Axial FLAIR image shows
abnormal hyperintensity of the deep
cerebellar nuclei (black arrows). B. Axial
T2-weighted image shows abnormal
hyperintensity in the globi pallidi (white
arrows). A small cyst (white arrowhead) is
present in the left globus pallidus.

GM1 and GM2 (Tay-Sachs and Sandhoff Diseases) mental retardation, and seizures. Those with the late infantile/juvenile
Gangliosidoses form have progressive psychomotor retardation beginning in early
GM1 gangliosidosis (OMIM 230500) is a rare lysosomal storage dis- childhood (typically between ages 1 and 5 years) with development of
ease characterized by a deficiency in the activity of lysosomal beta- seizures, spasticity, and a movement disorder; death ensues within a
galactosidase. The result is an accumulation of GM1 ganglioside few years. Children or adults with the chronic form have slowly pro-
and asialo-GA1 in the brain and of oligosaccharide in the abdomi- gressive dystonia, dysarthria, ataxia, myoclonus, and extrapyramidal
nal viscera (1003). Three forms of the disease have been described signs (1003,1004). Patients with the late infantile and chronic forms
(1003,1004); all are the result of mutations of the GLB1 gene located lack facial dysmorphism, hepatosplenomegaly and dysostosis.
on chromosome 3p21.33 (1005). Of interest, mutations of this same GM2 gangliosidoses are autosomal recessive disorders of sphin-
gene can cause Morquio type B disease (see section V.D.3); not surpris- golipid storage caused by a deficiency of hexosaminidase. The degra-
ingly, clinical overlap can be seen between the diseases (1006). Those dation of GM2 ganglioside to GM3 ganglioside is dependent on the
patients with the (most common) infantile form have dysmorphic enzyme lysosomal N-acetylhexosaminidase, which consists of two
facial features, osseous dysplasias, hepatosplenomegaly, hypotonia, major isoenzymes, A and B. A deficiency of isoenzyme A, coded by
the HEXA gene at chromosome 15q23-24, causes Tay-Sachs disease
(OMIM 272800) while deficiencies of the beta subunit of both isoen-
zymes A and B (coded for by the HEXB gene at chromosome 5q13)
cause Sandhoff disease (OMIM 268800) (1007). A third type of the
disease, called the AB variant of Tay-Sachs disease or the AB variant
of GM2 gangliosidosis (OMIM 272750) is caused by deficiency of the
GM2 activator protein, which mediates the interaction between the
water-soluble beta hexosaminidase A and its membrane-embedded
substrate, GM2 ganglioside (1008). All three of these disorders result
in an abnormal accumulation of GM2 ganglioside in the cytoplasm of
neurons; the resulting extensive neuronal loss and white matter degen-
eration lead to brain atrophy. Clinical and imaging findings are similar
in the diseases. In the most common infantile forms, patients present
with psychomotor retardation and hypotonia followed by neurologic
deterioration during the second half of the first year of life. Progressive
motor weakness, spasticity, dystonia, choreiform movements, ataxia,
blindness, macrocephaly, and seizures ensue (838,1009). After a period
of 3 to 10 years, the child becomes bedridden and demented (1010). In
the juvenile form of the disease, presentation is during the middle or
second half of the first decade, typically with gait disturbances, inco-
ordination, and developmental delay (1011), whereas patients with the
adult form typically present in middle age with progressively impaired
cognition (1012). In both of these disorders, the neuroimaging find-
FIG. 3-98. Propionic academia in a 22-month old child. Axial T2-weighted ings are typically limited to cerebellar>cerebral atrophy, so they are
image shows abnormal hyperintensity of the head of the caudate nucleus more fully discussed in the section on cerebellar disorders at the end
and putamen, as well as reduced myelination of the cerebral white matter. of this chapter.

Barkovich_Chap03.indd 194 5/6/2011 12:09:17 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 195

FIG. 3-99. GM2 gangliosidosis (Sandhoff disease). A. Axial CT


image shows abnormal high attenuation of the thalami (white
arrows). Note that the normal gray matter attenuation of the
basal ganglia is not present. B and C. Axial T2-weighted images
show slight hypointensity of the thalami (small white arrows)
compared to the cerebral cortex, patchy hyperintensity (large
white arrows) of the basal ganglia, and hyperintensity (white
arrowheads) of the periventricular white matter.

Neuroimaging findings in GM1 gangliosidosis, Tay-Sachs guishing feature (Fig. 3-100). Diffuse, progressive T2 hyperintensity
and Sandhoff disease are nearly identical (1004,1009,1013,1014), in the white matter (Figs. 3-99B and C and 3-100) is seen in both Tay-
although there are some subtle differences. CT studies show high Sachs and Sandhoff diseases, occasionally presenting a leukodystro-
attenuation in the thalami (Fig. 3-99A) and low attenuation in the phy-like appearance; however, the corpus callosum usually remains
white matter in early stages, with cerebral and cerebellar atrophy in well myelinated. Atrophy and T2 shortening of the thalami is seen in
later stages (1015). MRI shows some T1 hyperintensity in the thal- older patients with Sandhoff disease (59,1009). Eventually, cerebral
ami (1009,1014). T2 hyperintensity is seen in corpus striatum (Fig. and cerebellar atrophy ensues.
3-100) and in posteromedial thalami in Tay-Sachs disease (1014). Reports of short echo (30 ms) proton spectroscopy of the basal
One report of MRI in the late infantile form of GM1 gangliosido- ganglia in children with Tay-Sachs disease showed elevated myo-
sis reported T1 hyperintensity, with hypointensity on both T2- and Inositol and choline associated with reduced NAA compared with
T2*-weighed images in the acute phase, and generalized atrophy age-matched controls (1017,1018). Short echo MRS of a patient with
with atrophic, T2 hyperintense putamina, and shrunken, profoundly Sandhoff disease showed reduced NAA with elevated choline and myo-
hypointense globi pallidi on T2-weighted and FLAIR images (1016). Inositol in addition to a peak at 2.07 ppm that has been attributed to
The T2 hyperintensity in the basal ganglia of GM2 gangliosidoses N-acetylhexosamine; the highest concentrations of N-acetylhexo-
may render the basal ganglia isointense to surrounding white mat- samine are in white matter and thalami (1019).
ter (Fig. 3-100), in contrast to the dramatic high signal of the basal
ganglia seen in organic acidemias and mitochondrial disorders. T2 Fucosidosis
hypointensity and relatively reduced diffusion are seen specifically Fucosidosis (OMIM #230000) is a lysosomal disorder caused by defi-
in the ventral thalamic nucleus in Tay-Sachs, which may be a distin- ciency of alpha-L-fucosidase, the enzyme that hydrolyzes fucose from

Barkovich_Chap03.indd 195 5/6/2011 12:09:18 AM


196 Pediatric Neuroimaging

FIG. 3-100. GM2 gangliosidosis (Tay-


Sachs disease) in a 14-month-old. A and
B. Axial T2-weighted images show hyper-
intensity of the basal ganglia and thalami,
with the exception of a rounded area (white
arrows) in the ventral thalamus, which is
hypointense. C. Axial T2-weighted image
at the level of the centrum semiovale
shows absence of normal hypointensity
from myelination at this age. D. Diffusion
image at the level of (A) shows abnormally
increased diffusion (black arrows) through
most of the deep gray nuclei, but reduced
diffusion (white arrows) in the anterior
thalami, where the T2 hypointensity was
seen in (A).

glycolipids and glycoprotein; it is classified as a glycoproteinosis. The and FLAIR images of patients in late infancy and early childhood
enzyme deficiency results in the accumulation of a variety of alpha-L- (Fig. 3-101); this may be the only finding in late infancy (402). Asso-
fucose-rich storage products in many organs, such as the liver, spleen, ciated T2 shortening (manifested as hypointensity on T2-weighted
skin, heart, pancreas, thymus, thyroid, kidneys, and brain (1020). The images and most easily seen on FLAIR images) develops in the medial
genetic locus for alpha-L-fucosidase has been assigned to 1p34.1-36.1, and lateral segments of the globi pallidi (Fig. 3-101). Hyperintensity
and has been designated FUCA1 (1020). Clinically, this disease is divided of the hypothalami may be seen (1020). These features are somewhat
into two forms, infantile (type I) and juvenile (type II), although a con- reminiscent of findings in the GM2 gangliosidoses. In patients with
tinuous spectrum of the disease exists, dependent upon the severity of more longstanding disease, MRI shows diffuse atrophy with nonspe-
the enzyme deficit (401). Affected patients have progressive neurologic cific T2 prolongation in periventricular white matter and T1 and T2
deterioration along with angiokeratoma corporis diffusum, tiny purple shortening (T2 hypointensity) in the globi pallidi (1021). The appear-
or red raised cutaneous lesions that are initially found on the trunk, ance is nearly identical to that in the neuronal ceroid lipofuscinoses.
nonprogressive hepatomegaly (40%) and splenomegaly (25%) (401). Proton MR spectroscopy of the affected white matter shows the pres-
Skeletal findings are of dysostosis multiplex, which shows characteris- ence of a doublet centered at about 1.2 ppm (referred to, in error, as
tic abnormalities of spine, pelvis, and hips (401,1021). Pathology shows lactate in the literature) and a broad “hump” corresponding to multiple
prominent neuronal loss in gray matter, especially the thalamus, hypo- peaks from about 3.4-3.8 ppm (1023). The combination of imaging
thalamus, cerebral cortex, and the Purkinje cells and dentate nuclei of and MRS findings is probably specific.
the cerebellum (1022).
Imaging studies show findings characteristic of diffuse gray mat- l-2-hydroxyglutaric Aciduria
ter loss. CT shows atrophy and hypodensity in white matter and globi L-2-hydroxyglutaric aciduria (OMIM 236792) is an autosomal recessive
pallidi. MRI reports indicate variable findings (1020,1021). Abnor- disorder caused by mutations of the L2HGDH gene at chromosome
mal hyperintensity is present in the white matter on T2-weighted 14q22 (1024). The gene encodes a putative mitochondrial protein,

Barkovich_Chap03.indd 196 5/6/2011 12:09:19 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 197

which the authors dubbed “duranin,” with homology to FAD-


dependent oxidoreductases (1024). Affected patients present with a
slowly evolving moderate motor delay (mainly presenting as ataxia),
macrocephaly, and, sometimes, febrile seizures during the first year of
life (1025). Mental deficiency of variable severity typically becomes
manifest during the second year (1026–1029), sometimes followed by
dystonia and pyramidal signs. Rarely, the disease may have a fulmi-
nant course resulting in infantile death (1030). Pathologic analysis of
affected brains shows spongiform degeneration of cerebral and cer-
ebellar white matter with intense gliosis and vacuolation of the neu-
ropil. The subcortical white matter contains numerous hyperplastic
astrocytes and is severely demyelinated with cystic cavities. The basal
ganglia and cerebellum are less affected, with spongiosis but only mild
neuronal loss and no cavitations (1026–1028,1030–1032).
The MRI findings are virtually pathognomonic for the disease.
White matter abnormalities exhibit a very characteristic centripetal
and slightly anteroposterior gradient; subcortical U-fibers are most
severely affected with virtually total loss of normal signal intensity.
FIG. 3-101. Infantile fucosidosis. Axial T2-weighted image in a 13-month
Conversely, periventricular white matter, and in particular central
old child shows abnormal hypointensity of the white matter, most notable cortical-spinal tracts and corpus callosum are spared (Fig. 3-102).
in the peritrigonal regions (black arrowheads). Note the marked absence The extreme and external capsules, as well as the anterior limb and
of myelin in the posterior limbs of the internal capsules (white arrows) genu of the internal capsules are abnormal. Cerebellar white matter
and the hypointensity of the globi pallidi (white arrowheads). (Courtesy of is usually spared. Although at first glance the disease seems to have a
Dr. Susan Blaser, Toronto.) leukodystrophy-like appearance, the basal ganglia are always at least

FIG. 3-102. L-2-hydroxyglutaric aciduria in an 11-year-old, early phase. A. Axial T2-weighted image
shows abnormal hyperintensity in the cerebellar nuclei (white arrows). B. Axial T2-weighted image
shows abnormal hyperintensity of the globi pallidi (white arrows) more than putamina and caudates.
Subcortical white matter hyperintensity (white arrowheads) is present. C and D. Axial FLAIR (C) and
coronal T2 (D) images better show subcortical white matter hyperintensity (white arrowheads). Note
hyperintensity of cerebellar nuclei (white arrows) in (D).

Barkovich_Chap03.indd 197 5/6/2011 12:09:20 AM


198 Pediatric Neuroimaging

FIG. 3-103. L-2-hydroxyglutaric aci-


duria, later phase. Axial T2-weighted
images (A and B) show involvement of
globi pallidi (black arrows) more than
putamina and more extensive involve-
ment of white matter.

subtly abnormal (714,1026,1029,1033). The thalami are normal, but form of the disease has been identified; the causative gene (RANBP2)
the cerebellar nuclei are always abnormal and somewhat swollen encodes nuclear pore protein Ran Binding Protein 2 (1047) and maps
(Fig. 3-102) (714,1025,1026,1029,1033). Over time, more extensive to 2q12.1-2q13 (1048,1049). When the disease affects multiple family
involvement of white matter is noted (Fig. 3-103) and affected struc- members or has recurrent episodes in an individual, mutations of this
tures tend to atrophy (714,1025,1029,1033). When patients present as gene should be suspected (1047).
neonates, CT may show cerebellar hypodensity in the first few days Pathologic analysis of affected brains shows edema, causing necro-
of life (1030). Eventually, this may evolve to mild to severe cerebellar sis of the neurons and glia, in the thalamus, brainstem tegmentum,
atrophy, particularly involving the vermis. Water diffusivity is reported and cerebellar dentate nucleus. There is florid petechial hemorrhage
to be normal in most of the brain; it is reduced in acutely affected around small caliber intraparenchymal vessels. There may also be non-
white matter (11) and slightly increased in chronically affected sub- hemorrhagic white matter lesions in the cerebral and cerebellar paren-
cortical white matter (1034). Short echo time proton spectroscopy chyma. An absence of inflammatory cells, except a few extravasated
is reportedly normal other than slightly reduced NAA and increased leukocytes, differentiates this disorder from ADEM and acute hemor-
myo-Inositol (1034). rhagic encephalomyelitis (1037).
As described above, when the disease course is relatively benign Neuroimaging studies in acute necrotizing encephalitis show bilat-
and protracted, affected patients survive into adulthood. Lately, a eral thalamic lesions, which are continuous with lesions in the lateral
growing body of data suggests the increased incidence (at least 10%) of putamina and external/extreme capsules (Fig. 3-104); these often cavi-
brain tumors in patients with L-2-hydroxyglutaric aciduria (1035). The tate and become hemorrhagic, particularly in their central portions
explanation of this is yet unclear, but it is noteworthy that recently the (Fig. 3-105) (1050). Ring enhancement around the hemorrhagic areas
potential oncogenic role of 2-hydroxyglutarate (a product of tumor- develops after a few days. The anterior putamina tend to be spared.
associated IDH mutants) has been advocated (1036). Most studies also show low attenuation and T2/FLAIR hyperintensity
in the tegmentum of the brainstem and, in some, the nuclei and deep
Acute Necrotizing Encephalitis white matter of the cerebellum. In about half of affected patients the
The term acute necrotizing encephalitis (ANE) refers to an acute cerebral hemispheres are involved, usually by patchy hypodensity and
encephalopathy with bilateral thalamotegmental involvement that T2 hyperintensity (Fig. 3-104); some cavitate, but no hemorrhage is
occurs in infants and children; infants between ages 6 and 18 months seen in these cerebral hemispheric lesions (1037). Reduced diffusivity
are most commonly affected (1037–1040). A history of a mild anteced- is reported in the affected regions of the brainstem, cerebellum, and
ent illness (fever and upper respiratory or gastrointestinal infection) is thalami during the first few days after onset of symptoms (1045,1051).
elicited in more than 90% of affected patients. Then, after 0.5 to 3 days, However, our experience has been that diffusivity is increased (Fig.
the patient has an acute onset of severe neurological symptoms. In more 3-104), possibly because of the rapid tissue necrosis, unless the injured
severe cases, the patient develops convulsions (40%) or impaired con- area is hemorrhagic.
sciousness (28%) or vomiting (20%), and coma usually ensues within
24 hours. Recent reports suggest that the disorder may sometimes be Hypomyelination with Atrophy of the Basal Ganglia
rather mild, with alterations of consciousness or motor signs as the and Cerebellum
only symptoms (1041). Biochemical studies show elevated aspartate Hypomyelination with atrophy of the basal ganglia and cerebellum
aminotransferase in 82%, elevated alanine aminotransferase in 70%, (OMIM 612438, also called HABC and hypomyelinating leukodystro-
and elevated lactate dehydrogenase in 77%. CSF opening pressure is phy 6) is a newly described disorder of unknown cause. Clinical presen-
elevated and analysis reveals elevated protein but no cells (1037–1040). tation varies with the severity of the disease. Most affected children are
Although some reports have implicated influenza A and B viruses initially seen because of some difficulty learning to walk; subsequently,
(1042,1043) and human herpes virus 6 (1044,1045) in the pathogenesis, they have frank motor deterioration with an extrapyramidal movement
it is now believed that the most likely cause is immune-mediated after disorder consisting of dystonia, choreoathetosis, and rigidity, increas-
infection (1037,1042,1046). In support of this, an autosomal dominant ing spasticity, and ataxia (34,1052). Severely affected children are noted

Barkovich_Chap03.indd 198 5/6/2011 12:09:21 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 199

FIG. 3-104. Acute necrotizing encephalopathy (ANE). 3-year-


old with seizures after 1 day of fever, vomiting, diarrhea. A. Axial
noncontrast CT shows low attenuation in bilateral thalami (T)
and cerebral white matter. B–D. Axial T2-weighted images
show abnormal hyperintensity in the dorsal pons, cerebellar
white matter (B), thalami, posterior putamina and external/
extreme capsules (C), and cerebral white matter (D). E. Axial
average diffusivity image shows increased diffusivity (arrows)
in the thalami and external/extreme capsules. F. Single voxel
proton MRS (TE = 288 ms) from the thalamus shows low NAA
and markedly elevated lactate (Lac).

Barkovich_Chap03.indd 199 5/6/2011 12:09:22 AM


200 Pediatric Neuroimaging

FIG. 3-105. Acute necrotizing encephalopathy with thalamic necrosis and hemorrhage. Axial SE T2-weighted images show edema extending into the
posterior putamina, external and extreme capsules (white arrows in A). Note the hypointensity (black arrows in A), indicating hemorrhagic necrosis, in the
thalami. More superior image (B) shows hyperintensity(white arrows in B) extending into the perirolandic white matter.

to have poor vision or oculogyric eye movements and absent motor MRI shows diffuse hypomyelination, with hyperintense cere-
development when they are only a few months old (1053). Variable bral white matter compared to gray matter on T2-weighted images
nystagmus, decreased hearing, short stature, and microcephaly have (Fig. 3-106); progressive loss of myelin is seen on sequential scans (1052).
been reported (1052). Cognitive deficits are variable, but all affected Sometimes, the anterior portion of the posterior limb of the internal
children appear to have learning difficulties and require special edu- capsule may show slight T2 hypointensity. Of interest, the white matter
cation (34,1052). EEG reveals slowing of background activity in all does become hyperintense compared to gray matter on T1-weighted
patients. images (1052). These findings of hypomyelination, probably due to

FIG. 3-106. Hypomyelination with atrophy of the basal ganglia and cerebellum (HABC) in a 3-year-old. A. Sagittal T1-weighted image shows a thin cor-
pus callosum and cerebellar atrophy (white arrows). B. Axial T2-weighted image shows marked hypomyelination and nearly complete absence of caudate
heads and putamina. Globi pallidi (white arrows) can be seen.

Barkovich_Chap03.indd 200 5/6/2011 12:09:23 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 201

both lack of deposition and further myelin loss, have been confirmed
by histopathology (1052). The corticospinal tracts are most severely
affected and appear hyperintense in the pons and midbrain, highlighted
by the hypointensity of the surrounding structures (34). Depletion of
corticospinal tract axons in the brainstem was confirmed by histo-
pathology (1052). Volume of cerebral white matter is mildly reduced
(Fig. 3-106) and seems to diminish over time. In addition, the putamen
and, to a lesser degree the caudate nucleus, are small and show pro-
gressive atrophy on sequential scans; globi pallidi and thalami appear
to be spared. The cerebellum also undergoes progressive atrophy
(Fig. 3-106), with the vermis being more severely involved than the
hemispheres (34); histopathology shows the most severe involve-
ment to be in the internal granular layer of the cerebellar cortex
(1052). Proton MRS shows normal cortical spectra, but reveals
increased myo-Inositol and creatine peaks with normal NAA in the
white matter, representing white matter gliosis with preservation of
axons. Spectra of the basal ganglia are similar to those of white mat-
ter (34). The effects of this disorder on water diffusion, as measured
by diffusion-weighted imaging and diffusion tensor imaging, has not
been reported.

3-Methylglutaconic Aciduria
3-methylglutaconic aciduria (MGA) is a fairly common biochemical
disorder that is seen in several syndromes with diverse clinical features
(1054). It is usually divided into five categories based upon clinical and FIG. 3-107. 3-methylglutaconic aciduria, type I. Axial T2-weighted image
laboratory findings (1055). Patients with the autosomal recessive type shows T2 prolongation in the caudate heads and the anterior halves of the
I (OMIM 250950) typically present with neurologic signs and symp- putamina.
toms. These patients have mutations in the AUH gene (on chromo-
some 9), which encodes 3-methylglutaconyl-CoA hydratase (1056).
The phenotypic presentation varies, some patients manifesting delayed
development of language and hyperchloremic acidosis associated with and type IV 3MGA (and in the uncategorized phenotype with hyper-
gastroesophageal reflux, while others have much more severe pheno- methioninemia) the most characteristic imaging presentation is sym-
type, including seizures, cerebellar findings, and atrophy of the basal metrical bilateral basal ganglia disease (T2/FLAIR hyperintensity in
ganglia (582). In other types of 3MGA the deficient enzyme, hence the caudate heads and anterior halves of the putamina) with cerebel-
the source of excreted 3-methylglutaconic acid is yet unknown. As the lar atrophy (Fig. 3-107). In addition, myelination of the cerebral white
most commonly and severely affected organs in 3MGA are the brain, matter is delayed and, especially in advanced stages of the disease, dif-
heart, and sometimes the liver and skeletal muscles, it is possible that fuse cerebral atrophy as well as focal white matter alterations may also
increased excretion of 3-methylglutaconic acid is only an epiphe- be present (41,998,1061).
nomenon, or a biochemical marker for a group of unspecified energy
metabolism disorders. The primary defect is likely in the mitochondrial Molybdenum Cofactor Deficiency
respiratory chain. Molybdenum cofactor deficiency (OMIM 252150) is an autosomal
Barth syndrome or type II 3-methylglutaconic aciduria (OMIM recessive disorder in which molybdenum cofactor, a component that
302060) is caused by mutations of the TAZ gene at chromosome Xq28 is essential to the function of three enzymes, is deficient. Molybdenum
(1057) and characterized by short stature, congenital cardiomyopa- cofactor deficiency can be caused by mutation of genes (MOCS1 at
thy, neutropenia, and recurrent infections (1058,1059). In type III chromosome 14q24 and MOCS2 at chromosome 6p21.3) that encode
3-methylglutaconic aciduria also called Costeff syndrome, Behr syn- enzymes essential for the two separate steps in the formation of molyb-
drome, or optic atrophy plus (OMIM 258501, caused by mutation denum cofactor (1064). The clinical phenotype is identical in these two
of the OPA3 gene at chromosome 19q13.2-q13.3) (1060), early onset groups (1064). A third type of molybdenum cofactor deficiency, called
optic atrophy is accompanied by extrapyramidal signs and ataxia; type C, is caused by mutation in the gephyrin gene (GEPH at chromo-
the condition is well known among Jews of Iraqi descent. Type IV, or some 5q11) (1065).
nonspecific, 3-methylglutaconic aciduria is a clinically heterogeneous Patients with molybdenum cofactor deficiency present with
autosomal-recessive condition characterized neurologically by delayed severe neurologic signs and symptoms in first few days of life;
psychomotor development and slowly developing, worsening spastic- this neonatal encephalopathy can mimic that of hypoxic-ischemic
ity. Neonatal hypoglycemia and lactic acidosis have been noted in some injury. The leading symptom is drug-resistant epilepsy with a
cases (998,1061), as has presentation as Leigh syndrome (1062). Type V burst suppression pattern on EEG (1066,1067). Other features can
3-methlylglutaconic aciduria (OMIM 610198, caused by muta- include feeding difficulties, mental retardation, lens dislocations,
tions in the DNAJC19 gene at 3q26.3) is characterized by early-onset and myoclonic spasms (1068). Biochemical abnormalities include
dilated cardiomyopathy with conduction defects, nonprogressive cer- elevated urinary sulfite, low plasma urate, and very low urinary uric
ebellar ataxia, testicular dysgenesis, and growth failure in addition to acid. Pathologic exam of affected brains shows severe diffuse brain
3-methylglutaconic aciduria (1063). atrophy with diffuse myelin loss, cortical and central neuronal loss,
Imaging findings in type II and type II 3MGA are usually unre- gliosis, and cystic changes that are most severe in the basal ganglia
markable, although occasionally atrophy may be found. In both type I (1069–1071).

Barkovich_Chap03.indd 201 5/6/2011 12:09:24 AM


202 Pediatric Neuroimaging

FIG. 3-108. Molybdenum co-factor defi-


ciency. A. Subacute phase. Axial T2-weighted
image shows T2 prolongation in the caudate
heads (solid black arrows) and white matter,
T2 shortening in the shrunken lentiform
nuclei (open black arrows) and thalami. B.
Chronic phase. Axial T2-weighted image
shows volume loss in the basal ganglia.

Neuroimaging studies show profound injury to the brain. In the Diagnosis is made by detection of increased urinary excretion of
neonatal period, neuroimaging shows decreased attenuation (CT), and sulfite and S-sulphocysteine associated with low plasma cystine lev-
T1 and T2 prolongation (hypointensity on T1-weighted images, hyper- els. Decreased sulfite oxidase activity is found in cultured fibroblasts.
intensity on T2-weighted and FLAIR MR images) of the white matter Molecular analysis shows deletions in the mitochondrial genome
and caudate nuclei, suggestive of edema (1070,1072,1073). During the (1079). If the children survive, they manifest progressive microceph-
first weeks after birth, ultrasound shows rapid cystic degeneration of aly with long tract signs, poor head control, truncal hypotonia and
the white matter with enlargement of the ventricles and subarachnoid appendicular spasticity (830).
spaces (1074). During the same period, reduced diffusivity in affected Neuropathologic exam shows cystic encephalomalacia of the white
areas can be detected by diffusion-weighted imaging (1074). In the matter with marked astrogliosis, cortical atrophy, and shrunken, gliotic
subacute phase, the basal ganglia may show T1 hypointensity and T2 basal ganglia (830,1080).
hyperintensity, similar to that in profound neonatal hypotensive injury Imaging studies early in the course of the disease show edema in
(see Chapter 4) and urea cycle disorders (see section V.D.8). As the the cerebral cortex, white matter, and basal ganglia (Fig. 3-109A–D)
changes evolve into the more chronic phase, the caudate and lentiform (1081,1082). The hippocampi are relatively spared. T1 shortening is
nuclei show marked diminution in size (Fig. 3-108) and the white mat- seen at the cortical-white matter junction and in the basal ganglia
ter shows injury that may evolve into frank cystic changes (1072,1073). (Fig. 3-109A and B), followed by atrophy of the basal ganglia and
Graf et al. have reported a patient in whom globus pallidus involvement cystic degeneration of the white matter within the first month (Fig.
predominated (1075), whereas Teksam et al. have reported intracranial 3-109H and I) (1082). After a few weeks, the brain degenerates into
hemorrhage (1076). Differentiation from severe neonatal hypoxic isch- cystic encephalomalacia (1081). Diffusion-weighted imaging shows
emic injury (1077) is made by the predominant thalamic involvement markedly reduced diffusion in the basal ganglia, cerebral cortex, and
in hypoxic-ischemic injury (see Chapter 4) and its relative absence in subcortical white matter in the subacute phase (Fig. 3-109E and F)
this disorder. (1080). Long echo (TE = 288 ms) proton MR spectroscopy shows
very low NAA and markedly elevated lactate in the subacute phase
Isolated Sulfite Oxidase Deficiency (Fig. 3-109G) (1080).
Isolated sulfite oxidase deficiency (OMIM 272300) is a rare auto-
somal recessive disorder that leads to early neurologic deterioration Toxin Ingestions
and, in many cases, early death. The causative gene, called SUOX, It is important to remember that inborn errors of metabolism are
has been mapped to chromosome 12q13.2-13.3 (1078). Sulfite oxi- toxic exposures in which the toxins are intrinsic. It stands to reason,
dase is a soluble mitochondrial enzyme, located within the inter- therefore, that similar patterns of injury will result from exposure to
membranous space, which catalyzes the oxidation of endogenous or external toxins, such as drugs. Figure 3-110 is an example of brain
exogenous sulfite to sulfate. Sulfite oxidation is the final step in the injury from ingestion of substances that had toxic effects. Notice
metabolism of sulfur derived from sulfur-containing amino acids that both gray matter and white matter are involved; the imaging
(830). The presentation is similar to that of molybdenum cofactor features could be seen in an inborn error of metabolism. Toxic expo-
deficiency (section V.D.17). Affected neonates typically feed poorly, sure should always be considered if disease manifestations or disease
with frequent vomiting, are lethargic, and develop seizures in the first onset are in any way atypical, particularly the acute onset of neu-
few days of life. EEG shows diffuse slowing and, sometimes, bilat- rological signs and symptoms in a previously healthy adolescent or
eral diffuse epileptiform activity. Lens ectopia is common (1079). teen.

Barkovich_Chap03.indd 202 5/6/2011 12:09:25 AM


FIG. 3-109. Isolated sulfite oxidase deficiency. Newborn with seizures and vomiting. A and B. Axial T1-weighted images show blurring of the cortical-
white matter junction and abnormal hyperintensity in the lentiform nuclei (white arrows) as well as heterogeneous white matter with multiple foci of hyper-
intensity (white arrowheads). C and D. Axial T2-weighted images show abnormal heterogeneous hyperintensity of white matter (isointense to CSF) with
hyperintense basal ganglia (black arrows) and multiple areas of cortical edema (white arrows). E and F. Axial diffusivity (Dav) maps show reduced diffusivity
in the basal ganglia (black arrows) and multiple cortical regions (black arrowheads). G. Single voxel proton MRS (TE = 135 ms) shows reduced NAA and high
lactate (Lac) in the frontal white matter. H and I. Follow-up axial FLAIR images at age 6 months show severe atrophy with multicystic encephalomalacia.
Note the multiple layers of subdural hematomas (s) due to the atrophy.

Barkovich_Chap03.indd 203 5/6/2011 12:09:25 AM


204 Pediatric Neuroimaging

FIG. 3-110. Ten-year-old girl after


ingestion of multiple drugs. Axial
noncontrast CT images (A–D) show
marked hypoattenuation and swell-
ing of the cerebellum, globi pallidi,
and cerebral white matter to involve
the subcortical U fibers. Early hydro-
cephalus is present.

METABOLIC AND NEURODEGENERATIVE term cerebellar atrophy will refer to a cerebellum that is small and
has enlarged cerebellar fissures (Fig. 3-111) or if it has been shown to
DISORDERS PRIMARILY INVOLVING THE undergo progressive loss of volume. The pons is usually normal in cer-
CEREBELLUM ebellar atrophy, as the connections with the cerebellum are typically
well established before the atrophy begins; if atrophy begins in utero,
Differentiation of Cerebellar Atrophy from the pons will be proportionately small. The term cerebellar hypoplasia
Hypoplasia will refer to a small cerebellum in which the fissures are of normal
A large number of metabolic diseases affect the cerebellum; how- size compared with the folia (Fig. 3-112); cerebellar hypoplasia is usu-
ever, relatively few of these affect the cerebellum predominantly ally associated with pontine hypoplasia. For a reference, the normal
(Table 3-19). This section will discuss some disorders that affect cerebellar vermis extends from the inferior colliculi to the obex on
the cerebellum in childhood. Before discussing specific disorders, the midline sagittal MRI of a neonate and from the intercollicular sul-
however, it is important to discuss the difference between the imag- cus in an older infant or child. Note that these imaging appearances
ing appearance of cerebellar atrophy and cerebellar hypoplasia. The are fairly consistent, regardless of the specific cause of the hypoplasia

Barkovich_Chap03.indd 204 5/6/2011 12:09:27 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 205

TABLE 3-19 Childhood Disorders with Significant Cerebellar Involvement


A. Cerebellar Atrophy p. Ataxia with selective vitamin E deficiency
1. With Ataxia as an Early Clinical Sign q. Abetalipoproteinemia
a. Friedrich ataxia r. Cayman ataxia
b. Late-onset Tay-Sachs disease s. Dentatorubropallidoluysian atrophy
c. Pontocerebellar hypoplasia 2. Without Ataxia as an early sign
d. Mitochondrial disorders (particularly disorders of a. Congenital disorder of glycosylation 1a
respiratory chain complex I and coenzyme Q) b. Infantile neuroaxonal dystrophy
e. Cerebrotendinous xanthomatosis c. Infantile olivopontocerebellar atrophy
f. DNA polymerase gamma disorders (mitochondrial d. Progressive encephalopathy with edema, hypsarrhythmia,
recessive ataxia syndrome and optic atrophy (PEHO) syndrome
g. Neuronal ceroid lipofuscinoses (all types, cerebellar e. Langerhans cell histiocytosis
findings most pronounced in late infantile form and f. Wolfram syndrome
progressive epilepsy/mental retardation form) g. Hypomyelination with atrophy of the basal ganglia and
h. Ataxia-telangiectasia (see Chapter 6) cerebellum (discussed in previous section)
i. Ataxia telangiectasia-like disorder h. 3-methylglutaconic aciduria type I and IV
j. Ataxia with oculomotor apraxia, types 1 and 2 B. Cerebellar Hypoplasia
k. Autosomal recessive ataxia of Charlevoix-Saguenay 1. Marinesco-Sjögren syndrome
l. Infantile onset spinocerebellar ataxia 2. X-linked nonprogressive congenital cerebellar hypoplasia
m. Mevalonic kinase Deficiency 3. Höyeraal-Hreidarsson syndrome
n. Marinesco-Sjögren syndrome 4. Revesz syndrome
o. Spinocerebellar ataxias

or atrophy. As it seems pointless to show multiple cases with indis- those presenting in before the age of 20 years are traditionally clas-
tinguishable features, this section will illustrate only disorders with sified as “autosomal recessive” ataxias, while those presenting at later
somewhat distinctive features. ages are grouped as “autosomal dominant” ataxias. Although the age
of onset in all of these disorders can be variable, this traditional group-
ing is quite useful for the purposes of this book. Table 3-20 lists the
Cerebellar Ataxias
autosomal recessive ataxias with their genetic characteristics, main
Many genetic disorders cause cerebellar ataxia as an early sign of neu- clinical features, and imaging findings. More details are included in the
rologic disease. These can roughly be divided into two major groups; descriptions of the disorders that follow.

FIG. 3-111. Cerebellar atrophy. Axial (A) and coronal (B) T1-weighted images show a small cerebellum with shrunken cortex, diminished white matter,
and very enlarged fissures. The pons is of normal size, indicating that atrophy occurred after early infancy.

Barkovich_Chap03.indd 205 5/6/2011 12:09:28 AM


206 Pediatric Neuroimaging

FIG. 3-112. Cerebellar hyp-


oplasia. A. Moderate hypopla-
sia. The vermis (white arrows)
is small, not extending superi-
orly to the intercollicular sulcus
or inferiorly to the obex. Fis-
sures are not enlarged, how-
ever, indicating that no atrophy
has occurred. The pons (white
arrowheads) is small. B. Severe
hypoplasia. The vermis (white
arrow) is tiny, but has normal
lobulation and fissures are not
enlarged. Not the pons (white
arrowheads) is extremely small.

Cerebellar Atrophy mapped to chromosome 9q13, with no evidence of genetic heteroge-


neity (1083). The function of the encoded protein (called frataxin) is
Friedreich Ataxia unknown, but it is known that frataxin is located at the inner mito-
Friedreich ataxia (OMIM 229300) is the commonest form of hered- chondrial membrane and its mutation seems to trigger deficiency
itary ataxia, with a prevalence of 2.1 × 10−5. This is an autosomal of iron-sulphur cluster-containing enzymes, such as mitochondrial
recessive disorder; the genetic locus of the FXN gene has been respiratory chain complexes I-III (815). Deficiency of frataxin is gen-

TABLE 3-20 Cerebellar Ataxia Presenting in Childhood and Adolescence


Disorder Gene Locus Protein Function Clinical S/Sx Imaging
Friedreich Ataxia FXN 9q13 Mitochondrial iron Ataxia, nystagmus, weakness, amyotrophy, Normal Cb Spinal cord
metabolism unstable gaze fixation, Babinski, deaf- atrophy
ness, cardiomyopathy, diabetes, scoliosis
Late-Onset HEXA 15q23-24 Glycosphingolipid Ataxia, nystagmus, weakness, amyotro- Cb atrophy
Tay-Sachs metabolism phy, saccadic ocular pursuit, spasticity,
hypotonia, tremor, myoclonus, dystonia,
impaired cognition, epilepsy
Cerebrotendinous CYP27 2q33-ter Bile-acid synthesis Ataxia, weakness, amyotrophy, spasticity, Cb atrophy, cerebral atrophy,
xanthomatosis myoclonus, dystonia, Parkinsonism, diffusely increased FLAIR
impaired cognition, epilepsy, cataracts, and T2 white matter
tendon xanthomas signal intensity
Mitochondrial COQ2 9p13.3 Not known Hypotonia, motor delay, ataxia, or seizures Cb atrophy (vermis and
CoQ10 deficiency APTX 6q21 hemispheres)
PDSS1 10p12.1
PDSS2 4q21-q22
CABC1 1q42.2
Mitochon- POLG 15q22-26 Mitochondrial Ataxia, nystagmus, weakness, amyotro- Mild vermian atrophy,
drial DNA DNA repair and phy, ophthalmoplegia, saccadic ocular increased FLAIR and T2
polymerase replication pursuit, Babinski, tremor, myoclonus, white matter signal inten-
gamma choreoathetosis, impaired cognition, sity in Cb white matter,
disorders epilepsy, impaired hearing, hepatic thalami. In some variants,
failure T2 hyperintensity in
occipital lobes, brain stem.
Spinocerebellar Many Many Many Ataxia, weakness, amyotrophy Cb atrophy, spinal cord
ataxias 1–27 atrophy in some, pontine
atrophy in some

Barkovich_Chap03.indd 206 5/6/2011 12:09:29 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 207

TABLE 3-20 Cerebellar Ataxia Presenting in Childhood and Adolescence (Continued)


Disorder Gene Locus Protein Function Clinical S/Sx Imaging
Dentatorubropal- ATN1 12p13.31 Unstable CAG Myoclonus, cerebellar ataxia, and mental Cb and midbrain atrophy,
lidoluysian repeat retardation cerebral atrophy, white
atrophy matter volume loss.
Ataxia- ATM 11q22-23 DNA damage Ataxia, nystagmus, weakness, amyotrophy, Cb atrophy
Telangiectasia response oculomotor apraxia, Babinski, tremor,
myoclonus, dystonia, choreoathetosis,
diabetes, oculocutaneous telangiectasias,
radiosensitivity, immunodeficiency
Ataxia- MRE11 11q21 DNA damage Ataxia, nystagmus, weakness, amyotrophy, Cb atrophy
Telangiectasia- response oculomotor apraxia, dystonia, chore-
like disorder oathetosis, radiosensitivity, immuno-
deficiency
Ataxia with oculo- APTX 9p13 DNA repair, Ataxia, nystagmus, weakness, amyotrophy, Cb atrophy, mostly in
motor apraxia possibly RNA oculomotor apraxia, ophthalmoplegia, vermis.
type 1 processing gaze fixation instability, hypotonia,
tremor, dystonia, choreoathetosis,
impaired cognition, scoliosis, optic
atrophy
Ataxia with oculo- SETX 9q34 unknown Ataxia, nystagmus, weakness, amyotrophy, Cb atrophy, mostly in
motor apraxia oculomotor apraxia, saccadic ocular vermis.
type 2 pursuit, strabismus, Babinski, tremor,
myoclonus, dystonia, choreoathetosis,
impaired cognition, scoliosis
Ataxia of SACS 13q11 Protein folding Ataxia, nystagmus, weakness, amyotro- Cb vermian atrophy, spinal
Charlevoix- phy, saccadic ocular pursuit, spasticity, cord atrophy.
Saguenay Babinski, tremor, myoclonus, dystonia,
choreoathetosis, impaired cognition
Infantile-onset C10orf2 10q24 DNA replication Ataxia, weakness, amyotrophy, ophthal- Cb atrophy, spinal cord atro-
spinocerebellar moplegia, hypotonia, choreoathetosis, phy, brain stem atrophy.
ataxia impaired cognition, epilepsy, impaired
hearing, optic atrophy, hypogonadism
Cayman ataxia ATCAY 19p13.3 Neurotransmitter Ataxia, nystagmus, hypotonia, tremor, Cb atrophy
metabolism psychomotor delay
Marinesco- SIL1 5q31 ? Protein folding Ataxia, nystagmus, weakness, amyotro- Mild Cb atrophy
Sjögren phy, strabismus, hypotonia, tremor,
syndrome psychomotor delay, impaired cognition,
scoliosis, skeletal deformities, cataracts,
hypogonadism
Ataxia with Vit E TTPA 8q13.1- Vit E homeostasis Ataxia, amyotrophy, Babinski, tremor, RP, Normal or mild Cb atrophy
Deficiency 13.3 cardiomyopathy
Abetalipopro- MTP 4q22-24 Lipoprotein Ataxia, nystagmus, amyotrophy, RP, car- Normal
teinemia metabolism diomyopathy, acanthosis
Refsum Disease PHYH 10 pter- Fatty acid oxidation Ataxia, amyotrophy, RP, cardiomyopathy, Normal Cb
PEX7 p11.2 Peroxisomal skeletal deformities, renal failure,
6q22-q24 protein elevated CSF protein
importation
Autosomal SYNE1 6q Not known Dysarthria, ataxia, dysmetria, Cb vermian and hemispheric
recessive Cb hyperreflexia, nystagmus atrophy
ataxia Type 1

S/Sx, signs and symptoms; Cb, cerebellum; RP, retinitis pigmentosa; Vit, Vitamin.

Barkovich_Chap03.indd 207 5/6/2011 12:09:29 AM


208 Pediatric Neuroimaging

erally caused by expansion of the GAA triplet within the first intron build-up of ganglioside within neurons and glia is slower, the onset of
of the FXN gene; normally there are 33 or fewer GAA triplets, while symptoms later, and the progression of the disease more protracted
in pathological expansions, there are from 67 to more than 1,000 (1011). Patients with the juvenile form of the disease typically present
triplets (1084). The disease shows considerable variability in age of in the middle or end of the first decade with gait disturbances, incoor-
onset, rate of progression, severity, and extent of disease involvement dination, speech problems, and developmental delay; muscle wasting,
(1085). Although the disease can present at nearly any age, onset is proximal weakness, and bladder/bowel incontinence ensue during the
before the age of 10 years in 35% to 40%. Gait and stance ataxia and following decade (1011). Patients with the adult onset of the disease
lower limb areflexia are constant findings. Dysmetria, dysarthria, present in middle age with progressive cognitive disability (1012);
lower extremity areflexia, Babinski signs, scoliosis, and decreased most patients also suffer from psychotic episodes and bipolar disorder
vibratory sensation are present in the majority of affected patients; (1089,1090).
hypertrophic cardiomyopathy is present in nearly half (1085). A Unlike the infantile form, patients with late onset GM2 gan-
high incidence of diabetes mellitus (25%) is present in childhood gliosidosis typically have little or no white matter or basal ganglia
onset cases (1083). The main pathologic features are early loss of abnormalities on neuroimaging. Instead, their studies show pro-
large sensory neurons in the dorsal root ganglia, followed by degen- gressive cerebellar greater than cerebral atrophy (1011,1012,1091).
eration of the posterior columns of the spinal cord, spinocerebellar Proton MR spectroscopy (TE = 144 ms) shows reduced NAA in the
tracts, and pyramidal tracts (1086). MRI is most often normal but thalami and cerebral white matter compared with age-matched con-
may show atrophy of the superior vermis (Fig. 3-113), inferior cer- trols (1092).
ebellar hemispheres, spinal cord, and, occasionally, brainstem (Fig.
3-113) (1083,1087). Recently, DTI using tract based spatial statistics Ataxia with Oculomotor Apraxia, Types 1 and 2
has been used to study this disorder, finding increased diffusivity in
the cerebellum and decreased fractional anisotropy in the superior Ataxia with oculomotor apraxia type 1 (OMIM 208920) is an early
cerebellar peduncles (1088). onset autosomal recessive cerebellar ataxia with peripheral axonal
neuropathy, oculomotor apraxia, and hypoalbuminemia. It is caused
Ataxia-Telangiectasia by mutation of the APTX gene at 9p13 (1093), the protein product
of which is thought to play a role in DNA, and possibly RNA, repair
Ataxia telangiectasia is a fairly common cause of ataxia and cerebel- (1094). Affected patients typically present with cerebellar ataxia, senso-
lar atrophy in children, with prevalence as high at one per 40,000 in rimotor neuropathy (with dorsal column involvement and areflexia),
the United States. Some classify it as a phakomatosis. This disorder is ocular movement disorders (nystagmus, fixation instability, oculomo-
discussed in Chapter 6. tor apraxia), extrapyramidal signs, and mild cognitive impairment
(1095–1097). MRI shows cerebellar atrophy with predominantly verm-
Late Onset GM2 Gangliosidosis ian involvement (1096).
The “classic” infantile forms of GM2 gangliosidosis (Tay-Sachs dis- Formerly known autosomal recessive spinocerebellar ataxia, ataxia
ease and Sandhoff disease) were described in section IV.D.10 on dis- with oculomotor apraxia type 2 (OMIM 606002) is an autosomal reces-
eases involving gray and white matter. Some patients have less severe sive disorder caused by mutations (missense, in frame deletion or splice
involvement, probably because they are compound heterozygotes, mutations, or truncations) of the SETX gene at chromosome 9q34-qter
with one severe and one mild mutation (1089); as a consequence, the (1098) and characterized by the onset of gait ataxia during the second

FIG. 3-113. Friedreich ataxia. A. Sagittal T1-weighted image shows a small cerebellar vermis with think folia and prominent fissures (white arrows) in the
superior vermis. The cisterna magna is abnormally large. The pons is normal. B. Axial T2-weighted image though the pons shows a large fourth ventricle
and abnormal hyperintensity (white arrows) in the dorsal pons.

Barkovich_Chap03.indd 208 5/6/2011 12:09:29 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 209

decade, associated with oculomotor apraxia (∼50%), peripheral neu- in synthesis of CoQ10 can cause the syndrome (1106). In this section,
ropathy (>95%), impaired smooth pursuit (100%), and gaze-evoked only the ataxia/cerebellar atrophy form (971,972) will be discussed; in
nystagmus (90%). Pyramidal signs are present in approximately 20%, one report, this deficiency was present in 13% of patients with undiag-
tremor and extrapyramidal signs in 10% to 15%, and cognitive changes nosed cerebellar ataxia and atrophy (971). It has been postulated that,
are found in some populations (1099,1100). MRI is similar to that in because CoQ10 has lower concentration in the cerebellum than other
type 1, with cerebellar atrophy that is largely confined to the vermis regions of the brain that the cerebellum is particularly susceptible to
(1099–1101). injury in mitochondrial disorders (1107). However, a recent study
indicates that cerebellar volume loss can be seen in many mitochon-
Autosomal Recessive Spastic Ataxia drial disorders, including complex I deficiency (Fig. 3-114), complex
(of Charlevoix-Saguenay) II deficiency, complex IV deficiency, mutations of the catalytic subunit
of mitochondrial DNA polymerase g, MELAS syndrome, MGNIE syn-
Autosomal recessive spastic ataxia (OMIM 270550) is a disorder that
drome, and those with multiple respiratory chain deficiencies (833). In
was initially described in Quebec (1102), but has since been found
addition, some patients with coenzyme Q10 deficiency have a different
to be fairly widespread (1097,1103). Molecular genetic analyses have
clinical syndrome, with infantile encephalomyopathy and nephropathy
mapped the responsible gene, named SACS, to chromosome 13q12
(1108), while others have a predominantly myopathic form with recur-
(1104). The protein product of the gene, called sacsin, is thought to
rent myoglobinuria and CNS involvement (1109). Therefore, although
be involved in protein folding, and is expressed in a variety of regions
the presence of cerebellar atrophy in children should raise suspicion for
within the central nervous system, including the cerebral cortex, the
a mitochondrial disorder, the evaluation of mitochondria should not
granular layer of the cerebellar cortex, and the hippocampus (1103).
be restricted to coenzyme Q10.
Affected patients present in the third or fourth year of life with gait
Initial symptoms of coenzyme Q10 deficiency are quite variable.
unsteadiness after a delay in walking. Later, examination shows spastic
Encephalopmyopathic forms, infantile encephalopathy, Leigh syn-
ataxia, slurred speech, increased deep tendon reflexes, abnormal plantar
drome, and infantile multiorgan disease have been reported as well as
responses, nystagmus, retinal striations (due to hypermyelinated reti-
the ataxic encephalopathy with cerebellar atrophy (971,1110–1112).
nal axons), defects in ocular pursuit movements, and often mitral valve
The ataxic encephalopathy form typically is initially manifested as
prolapse (1102). EMG shows axonal neuropathy, with absent sensory
hypotonia and motor delay, ataxia, or seizures; all seem to have their
action potentials and mildly reduced motor conduction. Intelligence is
onset in the first decade. Ataxia becomes apparent by age 10 years and
usually normal. Imaging typically shows cerebellar vermian atrophy; in
affects the trunk, limbs, and speech. Long tract signs (hyperreflexia,
addition, some cases show diminished cerebral white matter and a thin
Babinski signs, spasticity) are found in about half of affected patients.
corpus callosum (1103,1105).
Other findings include myoclonus and ophthalmoparesis (971). Imag-
ing studies show progressive atrophy of the cerebellar vermis and
Mitochondrial Disorders Causing Cerebellar Atrophy hemispheres (971).
Coenzyme Q10 Deficiency
Deficiency of coenzyme Q10 (ubiquinone, a component of the mito- SANDO Syndrome
chondrial respiratory chain) can result in phenotypes of encephalomy- The so-called SANDO (sensory ataxic neuropathy, dysarthria, and
opathy, multisystemic organ failure, isolated myopathy, Leigh syndrome ophthalmoparesis) syndrome (OMIM 607459) is caused by muta-
with growth retardation, and progressive ataxia with cerebellar atrophy tions of the catalytic subunit of mitochondrial DNA polymerase g,
(OMIM 607426). Mutations of several genes that code proteins involved which is believed to function in both the replication and repair of

FIG. 3-114. Mitochondrial complex I deficiency with cerebellar atrophy. A. Sagittal T1-weighted image shows cerebellar atrophy with prominent fissures
(white arrows) in the markedly hypointense superior vermis. Note cavitation (white arrowheads) in the callosal genu, suggesting possible complex I or com-
plex II deficiency. B and C. Axial FLAIR images show hyperintensity of the superior cerebellum, indicating active disease, and hypointensity (supporting
cavitation) in the callosal genu and deep frontal white matter (white arrows).

Barkovich_Chap03.indd 209 5/6/2011 12:09:30 AM


210 Pediatric Neuroimaging

the mitochondrial genome (1113). Affected patients present in child- portion of the pons. As a result, abnormal development of the pon-
hood or adolescence, usually with sensorimotor ataxia, headaches, or tine nuclei, cerebellar nuclei, or the cerebellar cortex, or injury to these
seizures. Dysarthria, external ophthalmoplegia, or myoclonus may structures during the developmental period (with subsequent degen-
ensue (1113,1114). MRI shows T2 and FLAIR hyperintensity of the eration of axons coursing through the middle cerebellar peduncles)
cerebellar white matter with progressive cerebellar atrophy. Some cases result in hypoplasia of the cerebellar hemispheres and the pons. Thus,
have also been reported with hyperintensity in the thalami, occipital pontocerebellar hypoplasia is a heterogeneous disorder and most of
white matter, and brainstem (1114). the entities that make up the disorder have genetic causes (1121). Five
specific clinical syndromes have been defined in which the pons and
Infantile Onset Spinocerebellar Ataxia cerebellum are hypoplastic in infancy; these are referred to as the pon-
Still another mitochondrial disorder causing cerebellar ataxia is infan- tocerebellar hypoplasias types 1 to 5 (1122). These disorders are also
tile onset spinocerebellar ataxia (OMIM 271245), a disorder that has included in Chapter 5, but are principally discussed here, as their clini-
been described mainly in Finland (1115). The disorder is caused by cal courses are progressive.
mutation of the C10orf2 gene on chromosome 10q24; the protein Patients with type 1 pontocerebellar hypoplasia (PCH1, OMIM
product is involved in DNA replication (1115,1116). Affected patients 607596, caused by mutations of the VRK1 gene at chromosome 14q32)
present during the first 2 years of life with a progressive course that typically present with progressive microcephaly, central hypotonia,
includes cerebellar ataxia, hypotonia, sensory neuropathy with areflexia, visual impairment, abnormal eye movements, and delayed psychomo-
ophthalmoplegia, optic atrophy, epilepsy, and (in females) primary tor development (1123,1124). They are often myopathic from involve-
hypogonadism (1115,1116). MRI is normal at the time of clinical pre- ment of the anterior horn cells of the spinal cord, and may be born
sentation but shows progressive atrophy of the cerebellum, brainstem, with contractures of the extremities (1125). Both the cerebellum and
and spinal cord, starting at approximately 6 years of age (1116,1117). pons are small at birth and show further atrophy on subsequent stud-
ies; therefore, PCH1 is best considered a form of cerebellar atrophy that
Infantile Olivopontocerebellar Atrophy begins prenatally. A characteristic feature is that there is relative sparing
Autosomal dominant inheritance, onset during adulthood, and a of the vermis (Fig. 3-115), which is different from most cerebellar atro-
slowly progressive course characterize most cases of olivopontocer- phies (in most, the vermis is equally or more affected compared to the
ebellar atrophy. Infantile olivopontocerebellar atrophy is extremely cerebellar hemispheres). Associated anomalies of the cerebral cortex
rare and is characterized by rapid degeneration and death within a few and corpus callosum are commonly seen.
months or years of the onset of disease (1118). Neuroimaging by MRI Type 2 pontocerebellar hypoplasia, as described by Barth et al.
or CT shows classic findings of a small cerebellum and small pons, (1126,1127) is inherited as an autosomal recessive trait. It is genetically
with marked diminution in the size of the middle cerebellar peduncles heterogeneous, with PCH2A (OMIM 277470), caused by mutations of
(1119). This is a not a well-defined entity. Most cases are better clas- the TSEN54 gene at chromosome 17q25.1, PCH2B (OMIM 612389)
sified as congenital disorders of glycosylation (section IV.E.3.i). Oth- caused by mutations of TSEN2 gene at chromosome 3p25.1, and
ers may represent a recently described X-linked congenital cerebellar PCH2C (OMIM 612390) caused by mutations of the TSEN 34 gene at
ataxia (1120). chromosome 19q13.4. These genes encode noncatalytic (TSEN54) and
catalytic (TSEN2 and TSEN34) subunits of the tRNA splicing endonu-
Pontocerebellar Hypoplasia clease (1128). All have similar clinical characteristics. Affected patients
Cerebellar hypoplasia is frequently associated with pontine hypoplasia, are characterized by progressive microcephaly, epilepsy, and severe
probably for two reasons: (a) many cerebellar nuclei establish axonal neurological compromise, including early onset chorea and extrapyra-
connections with ventral pontine nuclei and vice versa (these axons midal dyskinesias that are progressive (1129,1130); there is essentially
form a significant amount of the bulk of the ventral pons) and (b) the no developmental progress (1126). Pathologically, it is characterized
decussation of the middle cerebellar peduncles forms the most ventral by a reduced number of folia in the cerebellum and an undersized

FIG. 3-115. Pontocerebellar hyp-


oplasia type 1. Sagittal T1-weighted
image (A) shows a small pons and
slightly hypoplastic cerebellar ver-
mis. The callosal splenium is too
small. Axial T2-weighted image
(B) shows that the vermis (black
arrows) is relatively spared com-
pared with cerebellar hemispheres.

Barkovich_Chap03.indd 210 5/6/2011 12:09:31 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 211

ventral pons. Histologic examination shows loss of both afferent and (1136), in which enzymes catalyze the synthesis and processing of
efferent cerebellar axons, marked reduction in the density of Purkinje asparagine (N)-linked glycans or oligosaccharides on glycoproteins.
and granule cells, and marked reduction in the number of neurons in These glycoconjugates play critical roles in metabolism, cell recogni-
the cerebellar nuclei. The pons shows reduced density of ventral pon- tion and adhesion, cell migration, protease resistance, host defense, and
tine neurons and diminished number of myelinated axons within the antigenicity, among others (1137). These disorders are divided into two
basis pontis (1131). In addition, segmental focal lesions are seen in the main groups and many (the number keeps increasing) subgroups that
cerebellar cortex, dentate nuclei, and inferior olives (1132). On imag- are likely to change as the disorders become better understood. Most of
ing, the pons is quite small, but may have a slight ventral bulge; the cer- them do not involve the cerebellum and will not be further discussed.
ebellum is variable in size, ranging from mildly to severely hypoplastic The most common type of congenital disorder of glycosylation is
(Fig. 3-116) (1129–1131,1133,1134). As with type 1 PCH, the vermis is Type 1a (OMIM 212065), formerly known as carbohydrate deficient
relatively spared compared with the cerebellar hemispheres. Supraten- glycoprotein syndrome. This autosomal recessive disorder is caused by
torial structures are reported to be essentially normal, but the suba- mutation in the PMM2 gene, located at chromosome 16p13.3-p13.2
rachnoid spaces may be enlarged and white matter may have abnormal (1138,1139). The underlying biochemical deficit is a reduction of
T1 hypointensity/T2 hyperintensity (Fig. 3-116). phosphomannomutase activity, impeding the conversion of mannose-
This well-defined entity should be differentiated from other 6-phosphate to mannose-1-phosphate, which is necessary for synthe-
reports of “PCH2,” which clearly describe a different disorder: onset is sis of dolichol-pyrophosphate-oligosaccharides (1138,1140). Due to
in utero (polyhydramnios, profound microcephaly), the neonates are increased understanding and more readily available testing, this disor-
profoundly affected, and imaging shows severe cerebral, as well and der is being discovered more frequently and is now understood to have
brainstem and cerebellar, atrophy (1134). Both the pons and cerebel- a variable clinical expression, ranging from severe to very mild. The
lum are much more severely affected in these cases. most severe form referred to as the neurologic multivisceral form, is
A third form of PCH (PCH3, OMIM 608027) was proposed by characterized by failure to thrive, inverted nipples, abnormal distribu-
Rajab et al. (1121), whose clinical, imaging and genetic studies demon- tion of subcutaneous fat, hepatomegaly, episodes of ascites, and peri-
strated a similar disorder to PCH2, except for the absence of dyskinesias cardial effusion. Infants generally present with severe encephalopathy
and the presence of optic atrophy, with linkage to chromosome 7q11-21. consisting of hypotonia, abnormal eye movement, and peripheral neu-
Unfortunately, no pathological information is available for this entity. ropathy; examinations reveal cerebellar hypoplasia, retinitis pigmen-
Additional PCH types were proposed by Patel et al. (1135), who sug- tosa, and pronounced psychomotor retardation (1141). Older children
gested that PCH cases with severe olivary hypoplasia (“C-shaped” infe- have ataxia, peripheral neuropathy, and moderate mental retardation
rior olives), also known as congenital olivopontocerebellar hypoplasia, (1142). About 20% die in the first year of life from severe infection,
should be distinguished as PCH4 if the vermis is relatively preserved, or liver failure, cardiac insufficiency, or status epilepticus (1141). Those
PCH5 if the vermis is not spared. However, Barth et al. performed patho- that survive show considerable motor and cognitive delay (1141,1143),
logic examinations on two patients with PCH4, finding features nearly usually noted at the age of 4 to 5 months. Motor impairment is wors-
identical to those in PCH2, only of greater severity (1132). In review- ened by the presence of cerebellar, extrapyramidal, and peripheral
ing this data, Hevner suggested that PCH2 and PCH4 are likely related nerve dysfunction with resultant axial hypotonia, muscle weakness
lesions, with their differences likely reflecting different mutations in the (especially in lower extremities), dyskinesia, ataxia, dysmetria, and
same gene or mutations of genes involving the same developmental path- tremor (1144). Progressive microcephaly and retinopathy are present
way (1122). Clearly, the understanding of these disorders is evolving. in about 80% of cases (1144). A second group, with findings restricted
to the nervous system, has a more benign prognosis. Patients in this
Congenital Disorders of Glycosylation group present with psychomotor retardation, strabismus, and retinitis
Congenital disorders of glycosylation are a group of disorders result- pigmentosa; they make developmental progress until adolescence, after
ing from defects in nine genes of the N-linked glycosylation pathway which the neurologic condition becomes static (1145). Diagnosis is

FIG. 3-116. Pontocerebellar hyp-


oplasia type 4. Sagittal T1-weighted
image (A) shows microcephaly with
disproportionately small cerebel-
lum and brain stem. The posterior
corpus callosum is too thin. Axial
T2-weighted image (B) shows
prominent CSF spaces and abnor-
mally hyperintense cerebral white
matter.

Barkovich_Chap03.indd 211 5/6/2011 12:09:31 AM


212 Pediatric Neuroimaging

that was originally described in Finland (1156), but has since been
described in populations all over the world (1157). Affected patients
present in infancy with hypotonia, hyperreflexia, poor feeding, micro-
cephaly, and delayed motor milestones and speech. Infantile spasms
develop during the first year of life. Deep tendon reflexes are initially
reduced, but become brisk as spastic quadriparesis ensues. No appar-
ent visual contact is appreciated, and ophthalmologic exam reveals
optic atrophy. Facial dysmorphisms include large earlobes, gingival
hyperplasia, and swelling of the face and the dorsum of hands and feet.
Electroencephalographic evaluation shows hypsarrhythmia, a pattern
of abnormal interictal high amplitude waves, and a background of
irregular spikes.
On pathology, cerebral and pronounced cerebellar atrophy is seen,
the essential histopathological lesions being confined to the cerebellar
cortex and the optic nerve. Severe neuronal loss is present in the inner
granular layer of the cerebellum. The Purkinje cells are relatively pre-
served in number although reduced in size, deformed and slightly mis-
aligned (dendrites were horizontally oriented). The molecular layer is
reduced in volume. The optic nerves are atrophic. Myelination appears
normal (1157,1158).
FIG. 3-117. Congenital disorder of glycosylation, type 1a. Sagittal Neuroimaging studies during the first year of life are often nor-
T1-weighted image shows a small cerebellar vermis with atrophy of the mal, other than thinning of the corpus callosum (1159). Subsequent
anterior vermis (white arrows). This is thought to represent atrophy that studies show reduced volume of cerebral white matter along with pro-
begins before birth. gressive cerebellar atrophy, involving the entire cerebellum (Fig. 3-118)
(1159). The reduced cerebral white matter volume results in progres-
sive thinning of the corpus callosum (Fig. 3-118D) (1157,1160). Serial
made by isoelectrofocusing of serum transferrin, which reveals marked neuroimaging studies showed that the disease process is operative dur-
increase of asialo- and disialotransferrin, with marked decrease of ing the postnatal period, although a prenatal onset cannot be excluded
tetra- and pentasialotransferrin (1146). (1158). Short echo proton MRS, not surprisingly, shows progressive
Neuroimaging studies show marked global cerebellar hypopla- reduction of NAA throughout the brain, along with the presence of
sia and superimposed atrophy (Fig. 3-117) that likely begins in utero small amounts of lactate (1159).
(1147) and progresses in infancy (1148,1149) but seems to stabi-
lize (1144). The anterior vermis is reportedly most severely affected Dentatorubral and Pallidoluysian Atrophy
(1150,1151). Associated imaging features include brainstem hypopla- Dentatorubral and pallidoluysian atrophy (OMIM 125370) is an auto-
sia, pontine hypoplasia, and supratentorial white matter hypoplasia or somal dominant disorder resulting from an unstable expansion of a
atrophy (1144,1152). CAG trinucleotide on the ATN1 gene on chromosome 12p13.31. The
greater the number of CAG repeats, the earlier the onset and the greater
Mevalonic Kinase Deficiency (Mevalonic Aciduria) the severity of the patient’s condition (1161,1162). The incidence of
Mevalonic kinase deficiency (OMIM 610377) is a disorder of choles- this disorder varies with race, with the highest incidence, by far, found
terol biosynthesis, resulting from a deficiency of mevalonate kinase, in Japanese; this incidence seems to parallel the frequency of CAG
the first enzyme after HMG CoA reductase in the biosynthesis of cho- repeats (1163). Age of onset ranges from the first to seventh decades;
lesterol and nonsterol isoprenes. The MVK gene encoding mevalonate as most patients present in adulthood, this disorder is discussed only
kinase is located on chromosome 12q24; the mutations thus far iden- briefly. Clinical symptoms are variable, even within a single affected
tified seem to cluster in the C-terminal end of the protein (1153). family. Most childhood-onset patients have a progressive myoclonic
Affected children present with varying degrees of severity of clinical epilepsy syndrome consisting of myoclonus, cerebellar ataxia, and
illness, which does not seem to be related to the level of activity of the mental retardation (1164). Adult-onset patients usually have cerebellar
enzyme. All patients suffer from recurrent crises consisting of fever, ataxia, choreoathetosis, and dementia; thus, they can be misdiagnosed
lymphadenopathy, hepatosplenomegaly, vomiting, diarrhea, arth- as having Huntington disease (1164,1165).
ralgia, and a morbilliform rash; these may be triggered by infection. Neuroimaging studies of patients with the juvenile form show T2/
Severely affected children have dysmorphic features (wide irregular FLAIR hyperintensity in globi pallidi and mild atrophy of cerebrum,
fontanelles, low set and posteriorly rotated ears), cataracts, hepa- cerebellum, and mesencephalon (1166). According to Miyazaki et al.
tosplenomegaly, lymphadenopathy, and anemia. They are severely (1167), however, the characteristic MR findings in the childhood-onset
developmentally delayed and may be stillborn or die in infancy. Less form of the disease are atrophy of the cerebellum and brainstem teg-
severely affected patients have psychomotor retardation, hypotonia, mentum (especially midbrain and superior pons), and periventricu-
myopathy, and ataxia. Neuroimaging studies show selective and pro- lar and/or deep white matter hyperintensity on T2-weighted images
gressive cerebellar atrophy involving both hemispheres and vermis (Fig. 3-119). T2/FLAIR hyperintensity of the white matter (correspond-
(1153–1155). ing to diffuse myelin pallor on histology (1168) ) is common, with the
number of lesions correlating with increasing age (1168). Noteworthy
Progressive Encephalopathy with Edema, in this report is the absence of definitive abnormalities involving the
Hypsarrhythmia, and Optic Atrophy (PEHO) globus pallidus, subthalamic nucleus (also known as the nucleus of
PEHO syndrome (progressive encephalopathy with edema, hypsar- Luys, from which the name of this disorder derives), and red nucleus in
rhythmia, and optic atrophy, OMIM 260565) is a recessive disorder the childhood form of the disease.

Barkovich_Chap03.indd 212 5/6/2011 12:09:32 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 213

FIG. 3-118. Progressive enceph-


alopathy with edema, hypsarrhyth-
mia, and optic atrophy (PEHO).
A. Sagittal T1-weighted image at
age 15 months shows thinning of
the corpus callosum (small white
arrows) with small cerebellum
(large white arrow) and brain stem.
B and C. Coronal T1-weighted
image (B) and axial T2-weighted
image (C) show that both cerebel-
lar hemispheres (white arrows in B)
are small and the volume of cerebral
white matter is reduced. D. Follow-
up sagittal T1-weighted image at
age 10 years shows further thinning
of the brain stem (white arrow) and
corpus callosum, and thickening of
the calvarium (white arrowheads, a
sign of further cerebral atrophy).
(These images courtesy Dr. Junichi
Takanashi, Chiba, Japan.)

Neuronal Ceroid Lipofuscinoses in childhood or it may be detected incidentally on brain MRI obtained
These disorders have already been discussed in a previous section of for other causes; neurologic symptoms from neurodegeneration are
this chapter on Gray Matter Disorders. It is important to remember that more common in the third or fourth decade (1173). The posterior
cerebellar atrophy is an important component of these disorders. More fossa is involved in more than 90% of affected patients, showing cer-
importantly, in some forms (in particular the late infantile (CLN2) and ebellar atrophy associated with symmetrical T2 hyperintensity of the
progressive epilepsy with mental retardation (CLN8) forms), the pre- cerebellar white matter and, often, a rim of T1 hyperintensity in the
senting signs (both clinical and imaging) are those of cerebellar atrophy. periphery of the cerebellar dentate nuclei (Fig. 3-120) (1169,1173). T2
hyperintensity is also frequently seen in the white matter of the pontine
Langerhans Cell Histiocytosis tegmentum, often associated with involvement of the pontine corti-
Langerhans cell histiocytosis (LCH, OMIM 604856) is a rare systemic cospinal tracts (Fig. 3-120). The cerebrum is typically involved, as well,
granulomatous disease of the dendritic system with a variable clini- showing foci of T2 hyperintensity in the cerebral white matter along
cal course that may be encountered at any age. The annual incidence with discrete symmetrical T1 hyperintensity in the globus pallidus.
in children aged younger than 10 years has been reported as 0.2 to 2 Diffuse cerebral cortical atrophy with thinning of the corpus callosum
per 100,000 children (1169). Typically, children with LCH present with may be present, but is uncommon (1169,1173).
either systemic disease (involving skin, bone, liver, spleen, lung, and
hematopoietic system) or focal brain or calvarial lesions (1170–1172) as Hypomyelination with Atrophy of the Basal
described in Chapter 7. Uncommonly, patients with LCH may develop a Ganglia and Cerebellum (HABC)
neurodegenerative pattern in the cerebellum with loss of Purkinje cells Hypomyelination with atrophy of the basal ganglia and cerebellum
and gliosis (1169). The patients may have signs of neurodegeneration (OMIM 612438) is a hypomyelinating disorder that has involvement

Barkovich_Chap03.indd 213 5/6/2011 12:09:32 AM


214 Pediatric Neuroimaging

FIG. 3-119. Dentatorubropallidoluysian atrophy (DRPLA), childhood form. A and B. Sagittal T1-weighted image (A) and axial T2-weighted images (B)
at age 2 years are essentially normal. C. Sagittal T1-weighted image at age 6 years show thinning of the midbrain (white arrowheads) and atrophy of the
cerebellum (shrunken folia and enlarged fissures, white arrows). D. Axial T2-weighted images show enlarged ventricles due to white matter volume loss and
atrophy of caudates, abnormal peritrigonal hyperintensity (white arrowheads), absence of the low signal intensity of the red nuclei (white arrows, compare
to B), and enlarged cerebral cortical sulci. Note that atrophy of the globus pallidus and subthalamic nucleus is not common in the childhood form of the
disease. (These images are courtesy of Dr. Hiroshi Oba, Tokyo, Japan.)

of the basal ganglia and white matter as well as cerebellar atrophy. It As some patients may initially present in childhood, they are briefly
is discussed in the section of this chapter on Disorders Involving Gray discussed here.
and White Matter.
Spinocerebellar Ataxias
Cerebellar Atrophy with Adolescent/Young The spinocerebellar atrophies are a group of phenotypically and geno-
typically heterogeneous group of dominantly inherited disorders. At the
Adult Onset
time of the writing of this book, at least 27 have been described (1174),
The following disorders may be detected in children, but more com- including dentatorubropallidoluysian atrophy (DRPLA) (1175). Most
monly present toward the end of the second decade or in adulthood. of these disorders have their onset in adults and differentiation is

Barkovich_Chap03.indd 214 5/6/2011 12:09:33 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 215

FIG. 3-120. Langerhans cell histocy-


tosis involving the cerebellum. Axial
T2-weighted image (A) shows abnor-
mal hyperintensity (white arrows)
pons in the region of the corticospinal
and corticopontine tracts. Coronal
T2-weighted image (B) shows abnor-
mal hyperintensity in the cerebellar
white matter (white arrows) and in
the hila of the cerebellar nuclei (black
arrowheads). Note the prominent sulci
in the cerebrum, suggesting mild cere-
bral cortical atrophy.

difficult, depending on a combination of clinical features, radiologic the pontocerebellar tracts, the optic radiations, the hippocampal for-
features, and genetic testing (1175). Some (SCA1-3, 7, 13, and DRPLA) nices and the deep cerebral white matter (1183). Two reports of MRI
have radiologic features of olivopontocerebellar atrophy, some (SCA in Wolfram syndrome noted striking brainstem and cerebellar atro-
12, 17, and 19) have both cerebral and cerebellar atrophy, and others phy in addition to atrophy of the optic system and hypothalamus
(SCA 14 and 15) have atrophy limited to the cerebellar vermis (1175). (1184,1185); both were adults at the time of the scan. However, another
A full discussion is well beyond the scope of this chapter. Interested report described normal cerebellum and brainstem other than T2 pro-
readers are referred to an increasing number of excellent works on the longation in the substantia nigra (1186). Thus, the disease is apparently
subject (1174–1177). heterogeneous; whether the imaging differences reflect differences in
the disease or just different stages of the disease (related to age, per-
Cerebrotendinous Xanthomatosis haps) remains to be clarified.
Cerebrotendinous xanthomatosis is a disorder of the hepatic bile acid
synthesis pathway caused by mutations of the CYP27 gene on chro- Cerebellar Hypoplasia
mosome 2q33-qter (1097,1178). Mutation of the protein product,
the mitochondrial sterol 27-hydroxylase, results in increased serum Marinesco-Sjögren Syndrome
cholestanol and bile alcohols; the deposition of these metabolites in Marinesco-Sjögren syndrome (OMIM 248800) is a rare autosomal reces-
the central nervous system probably causes the clinical phenotype. sive disorder characterized clinically by congenital cataracts, cerebel-
Neurologic symptoms typically begin during the late second decade or lar ataxia, hypogonadotropic hypogonadism, and mental retardation
early third decade and include ataxia with pyramidal or extrapyrami- (1187,1188). The causative gene (called SIL1) has been localized to chro-
dal signs, sensorimotor peripheral neuropathy, and seizures (11,1178). mosome 5q31 (1189) (another, very similar disorder of congenital cata-
Psychiatric problems and early dementia may ensue. Associated find- racts, facial dysmorphism, and neuropathy, or CCFDN, has been mapped
ings include early juvenile cataracts, atherosclerosis, tendon xanthomas, to 18q23 (1190) ). Cerebellar ataxia (with Purkinje and granule cell loss)
and chronic diarrhea. MRI shows generalized cerebral and cerebellar and childhood-onset cataracts are the most prominent signs of the
atrophy in addition to diffuse T2/FLAIR hyperintensity of the white Marinesco-Sjögren syndrome, but muscular lesions are also commonly
matter (11,1097,1178). Calcifications are often present in the affected present (1191,1192). The most characteristic MR finding is a very small
cerebellar white matter. cerebellum in a small posterior fossa (1193,1194), but this is not, appar-
ently, an invariable finding (1195). In addition, the anterior pituitary lobe
Wolfram Syndrome may be small and the posterior lobe may be small or absent (1195).
Wolfram syndrome was originally described as a combination of dia-
betes mellitus and bilateral optic atrophy (1179). It was later called X-Linked Nonprogressive Congenital
DIDMOAD for prominence of diabetes insipidus, diabetes mellitus, Cerebellar Hypoplasia
optic atrophy, and deafness (1180). In more than 90% of affected X-linked nonprogressive congenital cerebellar hypoplasia (OMIM
patients, the disorder (OMIM 222300) is caused by mutations of the 302500, also called X-linked spinocerebellar ataxia type 1) is an
WFS1 gene located at chromosome 4p16.1 (1181); a second form of X-linked disorder localized to an interval on chromosome Xp11.21-
the disease (WFS2, OMIM 604928) is associated with mutations of q21.3. Affected boys first present with a marked delay of early develop-
CISD2 at chromosome 4q (1182). Affected patients can also have anos- mental motor milestones. A neurologic syndrome becomes evident by
mia, ataxia, and central apnea. Pathologic analysis of the brain showed age 5 to 7 years and includes cerebellar ataxia, dysarthria, and external
severe degeneration of the optic nerves, chiasm and tracts as well as ophthalmoplegia. There are no symptoms of mental retardation, spas-
severe loss of neurons from the lateral geniculate nuclei, ventral pons, tic paraparesis, or sensory loss. Neuroimaging studies reveal hypoplasia
and the hypothalamic paraventricular and supraoptic nuclei. In addi- of the cerebellar hemispheres and vermis. The disease shows no pro-
tion, there was widespread axonal dystrophy with axonal swellings in gression beyond early childhood (1120,1196).

Barkovich_Chap03.indd 215 5/6/2011 12:09:34 AM


216 Pediatric Neuroimaging

Höyeraal-Hreidarsson Syndrome 14. Leijser LM, de Vries LS, Rutherford MA, et al. Cranial ultrasound in meta-
bolic disorders presenting in the neonatal period: characteristic features and
Höyeraal-Hreidarsson syndrome (OMIM 300240) is a multisystemic comparison with MR imaging. Am J Neuroradiol 2007;28(7):1223–1231.
disorder affecting males and characterized by prenatal onset of growth 15. Bastianello S, Bozzao A, Paolillo A, et al. Fast Spin-Echo and fast fluid-attenu-
failure, psychomotor retardation from an early age, microcephaly, ated inversion-recovery versus conventional spin-echo sequences for MR quan-
ataxia, spasticity, immunodeficiency, and thrombocytopenia and then tification of multiple sclerosis lesions. Am J Neuroradiol 1997;18:699–704.
progresses to pancytopenia (1197–1199). It is caused by mutation of 16. Moore GR, Leung E, MacKay AL, et al. A pathology-MRI study of the
the DKC1 gene at chromosome Xq28 (1200); this is the same gene that short T2 component in formalin-fixed multiple sclerosis brain. Neurology
causes X-linked dyskeratosis congenita. Neuropathological findings are 2000;55:1506–1510.
of delayed myelination and hypoplasia of the cerebellar granule cell 17. Sirrs SM, Laule C, Madler B, et al. Normal-appearing white matter in
layer without loss of Purkinje cells (1197,1198,1201). Fetal imaging Patients with phenylketonuria: water content, myelin water fraction, and
metabolite concentrations. Radiology. 2007;242(1):236–243.
may show cerebellar hypoplasia. Postnatal imaging shows a small cer-
18. Barkovich AJ. A magnetic resonance approach to metabolic disorder in
ebellum (with slightly enlarged fissures but no progression on succes- childhood. Rev Neurol 2006;43(Suppl 1):S5–S16.
sive scans) and small pons, usually with reduced cerebral white matter, 19. Florance NR, Davis RL, Lam CW, et al. Anti-N-methyl-D-aspartate recep-
a thin corpus callosum, and delayed myelination (1202,1203). Radio- tor (NMDAR) encephalities in children and adolescents. Ann Neurol
graphs may show distal metaphyseal flaring of the long bones (1203). 2009;66:11–18.
20. Spiegel R, Shaag A, Edvardson S, et al. SLC25A19 mutation as a cause of
Revesz Syndrome neuropathy and bilateral striatal necrosis. Ann Neurol 2009;66:419–424.
Revesz syndrome (OMIM 268130) is a variant of dyskeratosis congen- 21. Rahbeeni Z, Ozand P, Rashed M, et al. 4-hydroxybutyric aciduria. Brain
ita, a rare disorder characterized by bone marrow failure and integu- Dev 1994;16(Suppl):64–71.
ment abnormalities, such as abnormal skin pigmentation, dystrophic 22. Lossos A, Barash V, Soffer D, et al. Hereditary branching enzyme dysfunc-
tion in adult polyglucosan body disease: a possible metabolic cause in two
nails, and leukoplakia (1204). It is caused by mutations of the TINF2
patients. Ann Neurol 1991;30:655–662.
gene at chromosome 14q12 (1205). In Revesz syndrome, patients pres- 23. Berkhoff M, Weis J, Schroth G, et al. Extensive white-matter changes in case
ent with leukocoria due to a Coats disease–like retinopathy (1206). In of adult polyglucosan body disease. Neuroradiology 2001;43:234–236.
addition, patients can have aplastic anemia, ataxia with cerebellar hyp- 24. Benseler SM. Central nervous system vasculitis in children. Curr Rheumatol
oplasia, and multiple cerebral parenchymal abnormalities (1206,1207). Rep 2006;8:442–449.
Mental retardation is found in up to 25% of patients (1204). Imag- 25. Duzova A, Bakkaloglu A. Central nervous system involvement in pedi-
ing studies are characterized by the presence of leukocoria, cerebellar atric rheumatic diseases: current concepts in treatment. Curr Pharm Des
hypoplasia, multiple cerebral calcifications, and the presence on MRI 2008;14:1295–1301.
of multiple foci of T2 prolongation, sometimes enhancing, that are 26. van der Knaap MS, Naidu S, Kleinschmidt-DeMasters BK, et al. Autosomal
reported to represent gliosis (1207). dominant diffuse leukoencephalopathy with neuroaxonal spheroids. Neu-
rology 2000;54:463–468.
27. van der Knaap MS, Valk J. The reflection of histology in MR imaging of
REFERENCES Pelizaeus-Merzbacher disease. Am J Neuroradiol 1989;10:99–103.
28. Vaurs-Barrière C, Deville M, Sarrer C, et al. Pelizaeus-Merzbacher-
1. Kristjánsdóttir R, Uvebrant P, Hagberg B, et al. Disorders of the cere- like disease presentation of MCT8 mutated male subjects. Ann Neurol
bral white matter in children. The spectrum of lesions. Neuropediatrics 2009;65:114–118.
1996;27:295–298. 29. Uhlenberg B, Schuelke M, Rüschendorf F, et al. Mutations in the gene
2. van der Knaap M, Breiter S, Naidu S, et al. Defining and categorizing leu- encoding gap junction protein alpha-12 (Connexin 46.6) cause Pelizaeus-
koencephalopathies of unknown origin: MR imaging approach. Radiology Merzbacher-like disease. Am J Hum Genet 2004;75(2):251–260.
1999;213:121–133. 30. Loevner LA, Shapiro RM, Grossman RI, et al. White matter changes associ-
3. Finsterer J. Leigh and Leigh-like syndrome in children and adults. Pediatr ated with deletions of the long arm of chromosome 18 (18q- syndrome): a
Neurol 2008;39:223–235. dysmyelinating disorder? Am J Neuroradiol 1996;17:1843–1848.
4. Koenig M. Presentation and diagnosis of mitochondrial disorders in chil- 31. Linnankivi T, Tienari P, Somer M, et al. 18q deletions: clinical, molecu-
dren. Pediatr Neurol 2008;38:305–313. lar, and brain MRI findings of 14 individuals. Am J Med Genet A
5. Cañellas A, Gols A, Izquierdo J, et al. Idiopathic inflammatory-demyelinating 2006;140A(4):331–339.
diseases of the central nervous system. Neuroradiology 2007;49(5):393–409. 32. Haataja L, Parkkola R, Oonninen P, et al. Phenotypic variation and MRI
6. Schmitz T, Chew LJ. Cytokines and myelination in the central nervous sys- in Salla disease, a free sialic acid storage disorder. Neuropediatrics 1994;25:
tem. ScientificWorldJournal 2008;8:1119–1147. 238–244.
7. Perry A, Schmidt RE. Cancer therapy-associated CNS neuropathology: an 33. Sonninen P, Autti T, Varho T, et al. Brain involvement in Salla disease. Am J
update and review of the literature. Acta Neuropathol (Berl) 2006;111:197–212. Neuroradiol 1999;20:433–443.
8. Leake JA, Albani S, Kao AS, et al. Acute disseminated encephalomyelitis in 34. van der Knaap M, Naidu S, Pouwels P, et al. New syndrome characterized
childhood: epidemiologic, clinical and laboratory features. Pediatr Infect by hypomyelination with atrophy of the basal ganglia and cerebellum. Am
Dis J 2004;23(8):756–764. J Neuroradiol 2002;23:1466–1474.
9. Brass SD, Caramanos Z, Santos C, et al. Multiple sclerosis vs acute dissemi- 35. Biancheri R, Zara F, Bruno C, et al. Phenotypic characterization of hypo-
nated encephalomyelitis in childhood. Pediatr Neurol 2003;29(3):227–231. myelination and congenital cataract. Ann Neurol 2007;62:121–127.
10. van der Voorn JP, Pouwels PJW, Hart AAM, et al. Childhood white 36. Zara F, Biancheri R, Bruno C, et al. Deficiency of hyccin, a newly identified
matter disorders: quantitative MR imaging and spectroscopy. Radiology membrane protein, causes hypomyelination and congenital cataract. Nat
2006;241(2):510–517. Genet 2006;38(10):1111–1113.
11. van der Knaap MS, Valk J. Magnetic Resonance of Myelination and Myelin 37. Gordon K, Riding M, Camfield P, et al. CT and MR of 3-hydroxy-3-methyl-
Disorders, 3rd ed. Berlin: Springer, 2005. glutaryl-coenzyme A lyase deficiency. Am J Neuroradiol 1994;15:1474–1476.
12. Scriver CR, Beardet AL, Sly WS, et al. The metabolic and molecular bases of 38. Mahmoud AAH, Yousef GM, Al-Hifzi I, et al. Cockayne syndrome in three
inherited disease, 8th ed. New York: McGraw-Hill, 2001. sisters with varying clinical presentation. Am J Med Genet 2002;111:81–85.
13. Valle D, Beaudet AL, Vogelstein B, et al. The online metabolic and molecular 39. Ozand P, RAshed M, Millington D, et al. Ethylmalonic aciduria: an
bases of inherited diseases. In: Valle D, Beaudet AL, Vogelstein B, et al., eds. organic acidemia with CNS involvement and vasculopathy. Brain Dev
McGraw-Hill; 2009: http://www.ommbid.com/. Accessed September 29, 2009. 1994;16(Suppl):12–22.

Barkovich_Chap03.indd 216 5/6/2011 12:09:35 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 217

40. Brismar J, Ozand P. CT and MR of the brain in disorders of the propionate 67. Hanefeld F, Brockmann K, Dechent P. MR spectroscopy in pediatric white
and methylmalonate metabolism. Am J Neuroradiol 1994;15:1459–1473. matter disease. In: Gillard J, Waldman A, Barker P, eds. Clinical MR Neu-
41. Brismar J, Ozand PT. CT and MR of the brain in the diagnosis of organic roimaging. Cambridge: Cambridge University Press, 2005:755–778.
acidemias. Experiences from 107 patients. Brain Dev 1994;16(Suppl): 68. Krabbe K. A new familial infantile form of diffuse brain-sclerosis. Brain
104–124. 1916;39:74–114.
42. Al-Aqeel A, Rashed M, Ozand PT, et al. 3-methylglutaconic aciduria: ten 69. Kolodny EH. The mucolipidoses. In: Berg BO, ed. Principles of Child Neu-
new cases with a possible new phenotype. Brain Dev 1994;16:23–32. rology. New York: McGraw-Hill, 1996:1132–1136.
43. Ozand P, Gascon G, Al Essa M, et al. Biotin-responsive basal ganglia disease: 70. Wegner DA, Rafi MA, Luzi P. Molecular genetics of Krabbe disease (globoid
a novel entity. Brain 1998;121(7):1267–1279. cell leukodystrophy): diagnostic and clinical implications. Hum Mutat
44. OMIM. Online Mendelian Inheritance in Man. http://www.ncbi.nlm.nih. 1997;10:268–279.
gov/entrez/query.fcgi?db=OMIM&cmd=Limits 71. Satoh J-I, tokumoto H, Kurohara K, et al. Adult-onset Krabbe disease
45. Becker LE. Lysosomes, peroxisomes and mitochondria: function and disor- with homozygous T1853C mutation in the galactocerebrosidase gene:
der. Am J Neuroradiol 1992;13:609–620. unusual MRI findings of corticospinal tract demyelination. Neurology
46. van der Knaap MS. Myelination and myelin disorders: a magnetic reso- 1997;49:1392–1399.
nance study in infants, children and young adults. [PhD Thesis]. Free 72. Loes DJ, Peters C, Krivit W. Globoid cell leukodystrophy: distinguish-
University of Amsterdam and University of Utrecht, Netherlands, 1991. ing early onset from late onset disease using a brain MR imaging scoring
47. van der Knaap MS, Valk J. The MR spectrum of peroxisomal disorders. method. Am J Neuroradiol 1999;20:316–323.
Neuroradiology 1991;33:30–37. 73. Farina L, Bizzi A, finocchiaro G, et al. MR imaging and proton MR spec-
48. Becker LE, Yates A. Inherited metabolic disease. In: Davis R, Robertson D, troscopy in adult Krabbe disease. Am J Neuroradiol 2000;21:1478–1482.
eds. Textbook of Neuropathology, 2nd ed. Baltimore: Williams & Wilkins, 74. Itoh M, Hayashi M, Fujioka Y, et al. Immunohistological study of globoid
1990:331–427. cell leukodystrophy. Brain Dev 2002;24:284–290.
49. Lazarow PB. Peroxisome structure, function and biogenesis–human 75. Kwan E, Drace J, Enzmann D. Specific CT findings in Krabbe disease. Am J
patients and yeast mutants show strikingly similar defects in peroxisome Roentgenol 1984;143:665–670.
biogenesis. J Neuropath Exp Neurol 1995;54:720–725. 76. Ieshima A, Eda I, Matsui A, et al. CT in Krabbe’s disease: comparison with
50. Wanders RJA, Schutgens RBH, Barth PG. Peroxisomal disorders: a review. neuropathology. Neuroradiology 1983;25:323–327.
J Neuropath Exp Neurol 1995;54:726–739. 77. Farley TJ, Ketonen LM, Bodensteiner JB, et al. Serial MRI and CT findings
51. van der Knaap MS, Valk J. Myelin and white matter. In: van der Knaap MS, in infantile Krabbe disease. Pediatr Neurol 1992;8:455–458.
Valk J, eds. Magnetic Resonance of Myelin, Myelination, and Myelin Disor- 78. Lyon G, Hagberg B, Evrard P, et al. Symptomology of late onset Krabbe’s
ders, 2nd ed. Berlin: Springer, 1995:1–17. leukodystrophy: the European experience. Dev Neurosci 1992;13:240–244.
52. Lee J, Barry JA. Influence of membrane lipid packing on T2-weighted mag- 79. Demaerel P, Wilms G, Verdru P, et al. MR findings in globoid leukodystro-
netic resonance images: study of relaxation parameters in model mem- phy. Neuroradiology 1990;32:520–522.
brane systems. Magn Reson Med 1996;36:420–426. 80. Vanhanen S-L, Raininko R, Santavuori P. Early differential diagnosis of
53. Gieselmann V, Polten A, Kreysing J, et al. Molecular genetics of metachro- infantile neuronal ceroid lipofuscinosis, Rett syndrome, and Krabbe disease
matic leukodystrophy. J Inherited Metabol Dis 1994;17:500–509. by CT and MR. Am J Neuroradiol 1994;15:1443–1453.
54. Deconinck N, Messaaoui A, Ziereisen F, et al. Metachromatic leukodystro- 81. Provenzale J, Peddi S, Kurtzberg J, et al. Correlation of neurodevelopmental
phy without arylsulfatase A deficiency: a new case of saposin-B deficiency. features and MRI findings in infantile Krabbe’s disease. Am J Roentgenol
Eur J Paediatr Neurol 2008;12(1):46–50. 2009;192:59–65.
55. Eckhardt M, Hedayati KK, Pitsch J, et al. Sulfatide storage in neurons causes 82. Sasaki M, Sakuragawa N, Takashima S, et al. MRI and CT findings in
hyperexcitability and axonal degeneration in a mouse model of metachro- Krabbe disease. Pediatr Neurol 1991;7:283–288.
matic leukodystrophy. J Neurosci 2007;27(34):9009–9021. 83. Bernal O, Lenn N. Multiple cranial nerve enhancement in early infantile
56. Brady RO. Disorders of lipid metabolism. In: Berg BO, ed. Neurologic Krabbe’s disease. Neurology 2000;54:2348–2349.
Aspects of Pediatrics. Boston: Butterworth-Heinemann, 1992:145–165. 84. Beslow LA, Schwartz ES, Bönnemann CG. Thickening and enhancement of
57. Menkes JH. Textbook of Child Neurology. Philadelphia: Lea & Febiger, multiple cranial nerves in conjunction with cystic white matter lesions in
1985. early infantile Krabbe disease. Pediatr Radiol 2008;38:694–696.
58. Alves D, Pires MM, Guimaraes A, et al. Four cases of late onset metachro- 85. Given CAI, Santos CC, Durden DD. Intracranial and spinal MR imaging
matic leukodystrophy in a family: clinical, biochemical, and neuropatho- findings associated with Krabbe’s disease: case report. Am J Neuroradiol
logical studies. J Neurol Neurosurg Psychiatr 1986;49:1417–1422. 2001;22:1782–1785.
59. Kendall BE. Disorders of lysosomes, peroxisomes, and mitochondria. Am J 86. Escolar M, Poe M, Smith J, et al. Diffusion tensor imaging detects abnor-
Neuroradiol 1992;13:621–653. malities in the corticospinal tracts of neonates with infantile Krabbe dis-
60. van der Voorn JP, Pouwels PJW, Kamphorst W, et al. Histopathologic cor- ease. Am J Neuroradiol 2009;30:1017–1021.
relates of radial stripes on MR images in lysosomal storage disorders. Am J 87. Zarifi M, Tzika A, Astrakas L, et al. Magnetic resonance spectroscopy and
Neuroradiol 2005;26:442–446. magnetic resonance imaging findings in Krabbe’s disease. J Child Neurol
61. Eichler F, Grodd W, Grant PE, et al. Metachromatic leukodystrophy: a 2001;16:522–526.
scoring system for brain MR imaging observations. Am J Neuroradiol 88. Brockmann K, Dechent P, Wilken B, et al. Proton MRS profile of cerebral
2009;30:1893–1897. metabolic abnormalities in Krabbe disease. Neurology 2003;60:819–825.
62. Kim TS, Kim I-O, Kim WS, et al. MR of childhood metachromatic leu- 89. Kingsley PB, Shah TC, Woldenberg R. Identification of diffuse and focal
kodystrophy. Am J Neuroradiol 1997;18:733–738. brain lesions by clinical magnetic resonance spectroscopy. NMR Biomed
63. Maia Jr ACM, Da Rocha AJ, da Silva CJ. Multiple cranial nerve enhance- 2006;19:435–462.
ment: a new MR imaging finding in metachromatic leukodystrophy 90. Mosser J, Douar AM, Sarde CO, et al. Putative X-linked adrenoleukodystro-
(letter). Am J Neuroradiol 2007;28:999. phy gene shares unexpected homology with ABC transporters. Nature
64. Sener R. Metachronatic leukodystrophy: diffusion MR imaging findints. 1993;361:726–730.
Am J Neuroradiol 2002;23:1424–1426. 91. Mosser J, Lutz Y, Stoeckel ME, et al. The gene responsible for adrenoleu-
65. Engelbrecht V, Schere A, Rassek M, et al. Diffusion-weighted MR imaging kodystrophy encodes a peroxisomal membrane protein. Hum Mol Genet
in the brain in children: findings in the normal brain and in the brain with 1994;3:265–271.
white matter diseases. Radiology 2002;222:410–418. 92. Moser HW, Loes DJ, Melhern ER, et al. X-linked adrenoleukodystrophy:
66. van der Knaap MS, Valk J. Diffusion-weighted imaging. In: van der Knaap overview and prognosis as a function of age and brain magnetic resonance
MS, Valk J, eds. Magnetic Resonance of Myelination and Myelin Disorders, imaging abnormality. A study involving 372 patients. Neuropediatrics
3rd ed. Berlin: Springer, 2005:839–853. 2000;31:227–239.

Barkovich_Chap03.indd 217 5/6/2011 12:09:35 AM


218 Pediatric Neuroimaging

93. Kemp S, Wanders RJ. X-linked adrenoleukodystrophy: very long chain 120. Suzuki Y, Iai M, Kamei A, et al. Peroxisomal acyl CoA oxidase deficiency.
fatty acid metabolism, ABC half-transporters and the complicated route J Pediatr. 2002;140:128–130.
to treatment. Mol Genet Metab 2007;90:268–276. 121. Schiffmann R, Elroy-Stein O. Childhood ataxia with CNS hypomyelina-
94. Moser HW, Raymond G, Dubey P. Adrenoleukodystrophy: new appraoches tion/vanishing white matter disease–A common leukodystrophy caused
to a neurodegenerative disease. J Am Med Assn 2005;294:3131–3134. by abnormal control of protein synthesis. Mol Genet Metab 2006;88(1):7.
95. Krasemann E, Meier V, Korneke GC, et al. Identificantion of mutations 122. van der Knaap MS, Kamphorst W, Barth PG, et al. Phenotypic varia-
in the ALD gene of 20 families with adrenoleukodystrophy/adrenomy- tion with leukoencephalopathy with vanishing white matter. Neurology
eloneuropathy. Hum Genet 1996;97:194–197. 1998;51:540–547.
96. Korenke GC, Fuchs S, Krasemann E, et al. Cerebral adrenoleukodystrohpy 123. Damon-Perriere N, Menegon P, Olivier A, et al. Intra-familial phenotypic
(ALD) in only one of monozygotic twins with an identical ALD genotype. heterogeneity in adult onset vanishing white matter disease. Clin Neurol
Ann Neurol 1996;40:254–257. Neurosurg 2008;110:1068–1071.
97. van Geel BM, Bezman L, Loes DJ, et al. Evolution of phenotypes in 124. van der Knaap MS, van Berkel CG, Herms J, et al. eIF2B-related disorders:
adult male patients with X-linked adrenoleukodystrophy. Ann Neurol antenatal onset and involvement of multiple organs. Am J Hum Genet
2001;49:186–194. 2003;73(6):1199–1207.
98. Brites P, Mooyer PA, El Mrabet L, et al. Plasmalogens participate in very- 125. Francalanci P, Eymard-Pierre E, Dionisi-Vici C, et al. Fatal infantile leu-
long-chain fatty acid-induced pathology. Brain 2009;132:482–492. kodystrophy: a severe variant of CACH/VWM syndrome, allelic to chro-
99. Eichler F, Itoh R, Barker P, et al. Proton MR spectroscopic and diffusion mosome 3q27. Neurology 2001;57:265–270.
tensor brain MR imaging in X-linked adrenoleukodystrophy: initial expe- 126. Fogli A, Wong K, Eymard-Pierre E, et al. Cree leukocephalopathy
rience. Radiology 2002;225:245–252. and CACH/VWM disease are allelic at the EIF2B5 lucis. Ann Neurol
100. Moser HW. Adrenoleukodystrophy: phenotype, genetics, pathogenesis, 2002;52:506–510.
and therapy. Brain 1997;120:1485–1508. 127. Schiffmann R, Moller JR, Trapp BD, et al. Childhood ataxia with diffuse
101. Stephenson DJ, Bezman L, Raymond GV. Acute presentation of childhood central nervous system hypomyelination. Ann Neurol 1994;35:331–340.
adrenoleukodystrophy. Neuropediatrics 2000;31:293–297. 128. Hanefeld F, Holzbach U, Kruse B, et al. Diffuse white matter disease in
102. Naidu S, Moser AE, Moser HW. Phenotypic and genotypic variability of three children: an encephalopathy with unique features on magnetic reso-
generalized peroxisomal disorders. Pediatr Neurol 1988;4:5–12. nance imaging and proton magnetic resonance spectroscopy. Neuropedi-
103. Moser HW, Mihalik SJ, Watkins PA. Adrenoleukodystrophy and other per- atrics 1993;24:244–248.
oxisomal disorders that affect the nervous system, including new observa- 129. van der Knaap MS, Barth PG, Gabreëls FJM, et al. A new leukoencephal-
tions on L-pipecolic acid oxidase in primates. Brain Dev 1989;11:80–90. opathy with vanishing white matter. Neurology 1997;48:845–855.
104. Moser HW. Adrenoleukodystrophy: from bedside to molecular biology. 130. Vermeulen G, Seidl R, Mercimek-Mahmutoglu S, et al. Fright is a pro-
J Child Neurol 1987;2:140–150. voking factor in vanishing white matter disease. Ann Neurol 2005;57(4):
105. Moser H, Moser A, Naidu S, et al. Clinical aspects of adrenoleukodystro- 560–563.
phy and adrenomyeloneuropathy. Dev Neurosci 1991;13:254–261. 131. Passemard S, Gelot A, Fogli A, et al. Progressive megalencephaly due to
106. Fatemi A, Barker P, Ulug A, et al. MRI and proton MRSI in women specific EIF2B-epsilon mutations in two unrelated families. Neurology
heterozygous for X-linked adrenoleukodystrophy. Neurology 2003;60: 2007;69:400–402.
1301–1307. 132. Pineda M, Palmero AR, Baquero M, et al. Vanishing white matter disease
107. Moser HW. Peroxisomal disorders. In: Berg BO, ed. Principles of Child associated with progressive macrocephaly. Neuropediatrics 2008;39:29–32.
Neurology. New York: McGraw-Hill, 1996:1233–1248. 133. van der Knaap MS, Wevers RA, Kure S, et al. Increased CSF glycine; a bio-
108. Loes DJ, Hite S, Moser H, et al. Adrenoleukodystrophy: a scoring method chemical marker for leukoencephalopathy with vanishing white matter.
for brain MR observations. Am J Neuroradiol 1994;15:1761–1766. J Child Neurol 1999;14:728–731.
109. Schneider J, Il’yasov K, Boltshauser E, et al. Diffusion tensor imaging in 134. Black D, Booth F, Watters G, et al. Leukoencephalopathy among
cases of adrenoleukodystrophy: preliminary experience as a marker for native Indian infants in northern Quebec and Manitoba. Ann Neurol
early demyelination? Am J Neuroradiol 2003;24:819–824. 1988;24:490–496.
110. Melhem E, Loes D, Georgiades C, et al. X-linked adrenoleukodystrophy: 135. Harder S, Gourgaris A, Frangou E, et al. Clinical and neuroimaging find-
the role of contrast-enhanced MR imaging in predicting disease progres- ings of cree leukodystrophy: a retrospective case series. Am J Neuroradiol
sion. Am J Neuroradiol 2000;21:839–844. 2010 2010:ajnr.A2108.
111. Barkovich AJ, Ferriero DM, Bass N, et al. Involvement of the pontomed- 136. van der Knaap MS, Pronk JC, Scheper GC. Vanishing white matter disease.
ullary corticospinal tracts: a useful finding in the diagnosis of X-linked Lancet Neurol 2006;5:413–423.
adrenoleukodystrophy. Am J Neuroradiol 1997;19:95–100. 137. Fogli A, Rodriguez D, Eymard-Pierre E, et al. Ovarian failure related to
112. Kumar AJ, Rosenbaum AE, Naidu S, et al. Adrenoleukodystrophy: corre- eukaryotic initiation factor 2B mutations. Am J Hum Genet 2003;72:
lating MR imaging with CT. Radiology 1987;165:497–504. 1544–1550.
113. Aubourg P, Diebler C. Adrenoleukodystrophy—its diverse CT appearances 138. Leegwater PA, Konst AA, Kuyt B, et al. The gene for leukoencephalopathy
and an evolutive or phenotypic variant. Neuroradiology 1982;24:33–42. with vanishing white matter is located on chromosome 3q27. Am J Hum
114. Hong-Magno ET, Muraki AS, Huttenlocher PR. Atypical CT scans in adre- Genet 1999;65:728–734.
noleukodystrophy. J Comput Assist Tomogr 1987;11:333–336. 139. Fogli A, Dionisi-Vici C, Deodado F, et al. A severe variant of childhood
115. MacDonald J, Stauffer A, Heitoff K. Adrenoleukodystrophy: early frontal ataxia with central hypomyelination/vanishing white matter leukoenceph-
lobe involvement on computed tomography (case report). J Comput Assist alopathy related to EIF21B5 mutation. Neurology 2002;59:1966–1968.
Tomogr 1984;8:128–130. 140. Fogli A, Schiffmann R, Hugendubler L, et al. Decreased guanine nucle-
116. Tzika A, Ball W Jr, Vigneron D, et al. Childhood adrenoleukodystrophy: otide exchange factor activity in eIF2B-mutated patients. Eur J Hum Genet
assessment with proton MR spectroscopy. Radiology 1993;189:467–480. 2004;12:561–566.
117. Bruhn H, Kruse B, Korenke GC, et al. Proton NMR spectroscopy of cere- 141. Brück W, Herms J, Brockmann K, et al. Myelinopathia centralis diffusa
bral metabolic alterations in infantile peroxisomal disorders. J Comput (vanishing white matter disease): evidence of apoptotic oligodendrocyte
Assist Tomogr 1992;16:335–344. degeneration in early lesion development. Ann Neurol 2001;50:532–536.
118. Eichler F, Barker P, Cox C, et al. Proton MR spectroscopic imaging predicts 142. Wong K, Regina CA, Kymberly AG, et al. Foamy cells with oligodendro-
lesion progression on MRI in X-linked andrenoleukodystrophy. Neurology glial phenotype in childhood ataxia with diffuse central nervous system
2002;58:901–907. hypomyelination syndrome. Acta Neuropathologica 2000;V100(6):635.
119. Eichler F, Mahmood A, Loes D, et al. Magnetic resonance imaging detec- 143. Dietrich J, Lacagnina M, Gass D, et al. EIF2B5 mutations compromise
tion of lesion progression in adult patients with X-linked adrenoleu- GFAP+ astrocyte generation in vanishing white matter leukodystrophy.
kodystrophy. Arch Neurol 2007;64:659–664. Nature Med 2005;11:277–283.

Barkovich_Chap03.indd 218 5/6/2011 12:09:35 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 219

144. Ding XQ, Georg M, Eckert B, et al. Rapidly progressive vanishing white 170. Zinnati WJ, Lazovic J, Griffin K, et al. Dual mechanism of brain injury and
matter disease in a child with previously inconspicuous brain MRI. Neu- novel treatment strategy in maple syrup urine disease. Brain 2009;132:
ropediatrics 2006;37(4):253–256. 903–918.
145. van der Knaap MS, Smit LME, Barth PG, et al. MRI in classification of 171. Chuang DT, Shih VE. Disorders of branched-chain amino acid and keto
congenital muscular dystrophies with brain abnormalities. Ann Neurol acid metabolism. In: Scriver CR, Beardet AL, Sly WS, et al., eds. The Meta-
1997;42:50–59. bolic and Molecular Basis of Inherited Disease, vol 7. New York: McGraw-
146. Tedeschi G, Schiffmann R, Barton NW, et al. Proton magnetic resonance Hill, 1995:1239–1277.
spectroscopic imaging in childhood ataxia with diffuse central nervous 172. Brismar J, Aqueel A, Brismar G, et al. Maple syrup urine disease. Am J
system hypomyelination. Neurology 1995;45:1526–1532. Neuroradiol 1990;11:1219–1228.
147. Prineas JW, Ouvrier RA, Wright RG, et al. Giant axonal neuropathy–a gen- 173. Muller K, Kahn T, Wendel U. Is demyelination a feature of maple syrup
eralized disorder of cytoplasmic microfilament formation. J Neuropathol urine disease. Pediatr Neurol 1993;9:375–382.
Exp Neurol 1976;35(4):458–470. 174. Fariello G, Dionisi-Vici C, Orazi C, et al. Cranial ultrasonography in maple
148. Flanigan K, Crawford T, Griffin J, et al. Localization of the giant axonal syrup urine disease. Am J Neuroradiol 1996;17:311–315.
neuropathy gene to chromosome 16q24. Ann Neurol 1998;43:143–148. 175. Sakai M, Inoue Y, Oba H, et al. Age dependence of diffusion weighted mag-
149. Bomont P, Cavalier L, Blondeau F, et al. The gene encoding gigaxonin, a netic resonance imaging findings in maple syrup urine disease encephal-
new member of the cytoskeletal BTB/kelch repeat family, is mutated in opathy. J Comput Assist Tomogr 2005;29:524–527.
giant axonal neuropathy. Nat Genet 2000;26:370–374. 176. Harper PAW, Healy PJ, Dennis JA. Ultrastructural findings in maple syrup
150. Berg B, Rosenberg S, Ashbury A. Giant axonal neuropathy. Pediatrics urine disease in Poll Hereford calves. Acta Neuropathol (Berl) 1986;71:
1972;49:894–899. 316–320.
151. Igisu H, Goto I, Kawamura Y, et al. Acrylamide encephaloneuropathy due 177. Cavalleri F, Bernardi A, Burlina A, et al. Diffusion-weighted MRI of maple
to well water pollution. J Neurol Neurosurg Psychiatry 1975;38:581–584. syrup urine disease encephalopathy. Neuroradiology 2002;44:499–502.
152. Richen P, Tandan R. Giant axonal neuropathy: progressive clinical and 178. Jan W, Zimmerman R, Wang Z, et al. MR diffusion imaging and MR spec-
radiologic CNS involvement. Neurology 1992;42:2220–2222. troscopy of maple syrup urine disease during acute metabolic decompen-
153. Ravishankar S, Goel G, Rautenstrauss CP, et al. Spectrum of magnetic sation. Neuroradiology 2003;45:393–399.
resonance imaging findings in a family with giant axonal neuropathy con- 179. Felber SR, Sperl W, Chemelli A, et al. Maple syrup urine disease: metabolic
firmed by genetic studies. Neurol India 2009;57:181–184. decompensation monitored by proton magnetic resonance imaging and
154. Brockmann K, Pouwels P, Dechent P, et al. Cerebral proton magnetic reso- spectroscopy. Ann Neurol 1993;33:396–401.
nance spectroscopy of a patient with giant axonal neuropathy. Brain Dev 180. Enns GM, Cowan TM, Klein O, et al. Amiinoacidemias and organic aci-
2003;25:45–50. demias. In: Swaiman KF, Ashwal S, Ferriero DM, eds. Pediatric Neurol-
155. van der Knaap MS, Valk J. Magnetic Resonance of Myelin, Myelination, and ogy: Principles and Practice, vol 1. Philadelphia: Mosby Elsevier, 2006:
Myelin Disorders, 2nd ed. Berlin: Springer, 1995. 567–602.
156. Sweetman L, Haas RH. Abnormalities of amino acid metabolism. In: Berg 181. Peterson JC, Spence JD. Vitamins and progression of atherosclerosis in
BO, ed. Neurologic Aspects of Pediatrics. Boston: Butterworth-Heinemann, hyperhomocyst(e)inaemia. Lancet 1998;24:263.
1992:3–32. 182. Yap S, Rushe H, Howard PM, et al. The intellectual abilities of early-treated
157. Scriver CR, Eisensmith RC, Woo SL, et al. The hyperphenylalaninemias of individuals with pyridoxine-nonresponsive homocystinuria due to cysta-
man and mouse. Annu Rev Genet 1994;28:141–165. thionine beta-synthase deficiency. J Inherit Metab Dis 2001;24:437–447.
158. Kahler R, Fahey M. Metabolic disorders with mental retardation. Am J 183. van den Berg M, van der Knaap MS, Boers GHJ, et al. Hyperhomocystei-
Med Genet 2003;117C:31–41. naemia; with reference to its neuroradiological aspects. Neuroradiology
159. Pey AL, Stricher F, Serrano L, et al. Predicted effects of missense mutations on 1995;37:403–411.
native-state stability account for phenotypic outcome in phenylketonuria, a 184. Sibani S, Christensen B, O’Ferrall E, et al. Characterization of six novel
paradigm of misfolding diseases. Am J Hum Genet 2007;81:1006–1024. mutations in the methylenetetrahydrofolate reductase (MTHFR) gene in
160. Valk J, van der Knaap MS. Magnetic Resonance of Myelin, Myelination, and patients with homocystinuria. Hum Mutat 2000;15:280–287.
Myelination Disorders. Berlin: Springer, 1989. 185. Goyette P, Frosst P, Rosenblatt D, et al. Seven novel mutations in the meth-
161. Pearsen KD, Gean-Marton AD, Levy HL, et al. Phenylketonuria: MRI of ylenetetrahydrofolate reductase gene and genotype/phenotype correla-
the brain with clinical correlation. Radiology 1990;177:437–440. tions in severe methylenetetrahydrofolate reductase deficiency. Am J Hum
162. Shaw DW, Maravilla KR, Weinberger E, et al. MR imaging of phenylketo- Genet 1995;56:1052–1059.
nuria. Am J Neuroradiol 1991;12:403–406. 186. Ganesan V, Prengler M, McShane MA, et al. Investigation of risk fac-
163. Pietz J, Kreis R, Schmidt H, et al. Phenylkentonuria: findings at MR imag- tors in children with arterial ischemic stroke. Ann Neurol 2003;53(2):
ing and localized in vivo H-1 MR spectroscopy of the brain in patients 167–173.
with early treatment. Radiology 1996;201:413–420. 187. Grow JL, Fliman PJ, Pipe SW. Neonatal sinovenous thrombosis associated
164. Phillips MD, McGraw P, Lowe MJ, et al. Diffusion-weighted imaging of with homozygous thermolabile methylenetetrahydrofolate reductase in
white matter abnormalities in patients with phenylkentonuria. Am J Neu- both mother and infant. J Perinatol 2002;22(2):175–178.
roradiol 2001;22:1583–1586. 188. Al Tawari A, ramadan D, Neubauer D, et al. An early onset form of methyl-
165. Kono K, Okano Y, Nakayama K, et al. Diffusion weighted MR imaging enetetrahydrofolate reductase deficiency: a report of a family from Kuwait.
in patients with phenylketonuria: relationship between serum phenylala- Brain Dev 2002;24:304–309.
nine levels and ADC values in cerebral white matter. Radiology 2005;236: 189. Pezzini A, Del Zotto E, Archetti S, et al. Plasma homocysteine concentra-
630–636. tion, C677T MTHFR genotype, and 844ins68bp CBS genotype in young
166. Pfaendner NH, Reuner G, Pietz J, et al. MR Imaging-Based Volum- adults with spontaneous cervical artery dissection and atherothrombotic
etry in Patients with Early-Treated Phenylketonuria. Am J Neuroradiol. stroke. Stroke 2002;33(3):664–669.
2005;26(7):1681–1685. 190. Strauss KA, Morton DH, Puffenberger EG, et al. Prevention of brain dis-
167. Novotny EJ, Avison MJ, Herschkowitz N, et al. In vivo measurement of ease from severe 5,10-methylenetetrahydrofolate reductase deficiency.
phenylalanine in human brain by proton nuclear magnetic resonance Molec Genet Metab 2007;91:165–175.
spectroscopy. Pediatr Res 1995;37:244–249. 191. Engelbrecht V, Rassek M, Huismann J, et al. MR and proton MR spectros-
168. Peng S, Tseng W-Y, Chien Y-H, et al. Diffusion tensor imaging in children copy of the brain in hyperhomocysteinemia caused by methylenetetrahy-
with early treated, chronic, malignant phenylketonuria: correlation with drofolate reductase deficiency. Am J Neuroradiol 1997;18:536–539.
intelligence assessment. Am J Neuroradiol 2004;25:1569–1574. 192. Rossi A, Cerone R, Biancheri R, et al. Early onset combined methylmalonic
169. Ogier de Baulny H, Saudubray JM. Branched-chain organic acidurias. aciduria and homocystinuria: neuroradiologic findings. Am J Neuroradiol
Semin Neonatol 2002;7:65–74. 2001;22:554–563.

Barkovich_Chap03.indd 219 5/6/2011 12:09:35 AM


220 Pediatric Neuroimaging

193. Biancheri R, Cerone R, Schiaffino MC, et al. Cobalamin (Cbl) C/D defi- 218. Yuksel A, Karaca E, Albayram MS. Magnetic resonance imaging, magnetic
ciency: clinical, neurophysiological and neuroradiologic findings in resonance spectroscopy, and facial dysmorphism in a case of Lowe syn-
14 cases. Neuropediatrics 2001;32:14–22. drome with novel OCRL1 gene mutation. J Child Neurol 2009;24:93–96.
194. Rosenblatt DS, Aspler AL, Shevell MI, et al. Clinical heterogeneity and 219. Cwik VA, Brooke MH. Disorders of muscle—dystrophies and myopathies.
prognosis in combined methylmalonic aciduria and homocystinuria In: Berg BO, ed. Prinicples of Child Neurology. New York: McGraw-Hill,
(cblC). J Inher Metab Dis 1997;20:528–538. 1996:1665–1702.
195. Morel CF, Lerner-Ellis JP, Rosenblatt DS. Combined methylmalonic aci- 220. Lamer S, Carlier R-Y, Pinard J-M, et al. Congenital muscular dystrophy:
duria and homocystinuria (cblC): phenotype-genotype correlations and use of brain MR imaging findings to predict merosin deficiency. Radiology
ethnic-specific observations. Molec Genet Metabol 2006;88(4):315–321. 1998;206:811–816.
196. Smith DL, Bodamer OA. Practical management of combined methylma- 221. Hillaire D, Leclerc A, Faure S. Localization of merosin-negative congenital
lonicaciduria and homocystinuria. J Child Neurol 2002;17(5):353–356. muscular dystrophy to chromosome 6q2 by homozygosity mapping. Hum
197. Watkins D, Ru M, Hwang H-Y, et al. Hyperhomocysteinemia due to Mol Genet 1994;9:1657–1661.
methionine synthase deficiency, cblG: structure of the MTR gene, geno- 222. Topaloglu H, Kale G, Yalnizoglu D, et al. Analysis of “pure” congenital
type diversity, and recognition of a common mutation, P1173L. Am J Hum muscular dystrophies in thirty-eight cases. How different is the classical
Genet 2002;71:143–153. type 1 from the occidental type of cerebromuscular dystrophy? Neurope-
198. Whitehead VM. Acquired and inherited disorders of cobalamin and folate diatrics 1994;25:94–100.
in children. Br J Haematol 2006;134:125–136. 223. Voit T. Congenital muscular dystrophies: 1997 update. Brain Dev 1998;
199. Powers JM, Rosenblatt DS, Schmidt RE, et al. Neurological and neuro- 20:65–74.
pathologic heterogeneity in two brothers with cobalamin C deficiency. 224. Jones KJ, Morgan G, Johnston H, et al. The expanding phenotype of lami-
Ann Neurol 2001;49:396–400. nin a2 chain (merosin) abnormalities: case series and review. J Med Genet
200. Longo D, Fariello G, onisi-Vici C, et al. MRI and 1H-MRS findings in 2001;38(10):649–657.
early-onset cobalamin C/D defect. Neuropediatrics 2005;36(6):366–372. 225. Brockmann K, Dechent P, Bönnemann C, et al. Quantitative proton MRS
201. Wolf B. Disorders of biotin metabolism. In: Scriver CR, Beaudet AL, Sly of cerebral metabolites in laminin alpha2 chain deficiency. Brain Dev
WS, et al., eds. The Metabolic and Molecular Bases of Inherited Disease. New 2007;29:357–364.
York: McGraw-Hill, 2001:3935–3962. 226. Leite CC, Reed UC, Otaduy MCG, et al. Congenital muscular dystrophy
202. Bousounis DP, Camfield P, Wolf B. Reversal of brain atrophy with biotin with merosin deficiency: H1 MR spectroscopy and diffusion-weighted
treatment in biotinidase deficiency. Neuropediatrics 1993;24:214–217. MR imaging. Radiology 2005;235:190–196.
203. Desai S, Ganesan K, Hegde A. Biotinidase deficiency: a reversible meta- 227. Bargal R, Avidan N, Ben-Asher E, et al. Identification of the gene causing
bolic encephalopathy. Neuroimaging and MR spectroscopic findings in a mucolipidosis type IV. Nat Genet 2000;26:118–123.
series of four patients. Pediatr Radiol 2008;38:848–856. 228. LaPlante J, Falardeau J, Sun M, et al. Identification and characteriza-
204. Soares-Fernandes JP, Magalhaes Z, Rocha JF, et al. Brain diffusion-weighted tion of the single channel function of human mucolipin-1 implicated in
and diffusion tensor imaging findings in an infant with biotinidase defi- mucolipidosis type IV, a disorder affecting the lysomal pathway. FEBS Lett
ciency. Am J Neuroradiol 2009;30(9):E128. 2002;532:183–187.
205. Mudd S, Levy H, Kraus J. Disorders of transsulferation. In: Scriver C, 229. Bindu PS, Gayathri N, Yasha T, et al. A variant form of mucolipidosis
Beaudet A, Sly W, et al., eds. The Metabolic and Molecular Bases of Inherited IV: report on 4 patients from the Indian subcontinent. J Child Neurol
Diseases. 8th ed. New York: McGraw-Hill, 2001:2007–2056. 2008;23:1443–1446.
206. Chamberlin M, Ubagai T, Mudd S, et al. Methionine adenosyltransferase 230. Frei KP, Patronas NJ, Crutchfield KE, et al. Mucolipidosis type IV: charac-
i/III deficiency: novel mutations and clinical variations. Am J Hum Genet teristic MRI findings. Neurology 1998;51:565–569.
2000;66:347–355. 231. Bonavita S, Virta A, Jeffries N, et al. Diffuse neuroaxonal involvement in
207. Stabler S, Steegborn C, Wahl M, et al. Elevated plasma total homocysteine mucolipidosis IV as assessed by proton magnetic resonance spectroscopic
in severe methionine adenosyltransferase I/III deficiency. Metabolism imaging. J Child Neurol 2003;18:443–449.
2002;51:981–988. 232. Tallaksen C, Durr A, Brice A. Recent advances in hereditary spastic para-
208. Nussbaum R, Orrison B, Jänne P, et al. Physical mapping and genomic plegia. Curr Opin Neurol 2001;14:457–463.
structure of the Lowe syndrome gene OCRL1. Hum Genet 1997;99: 233. Salinas S, Proukakis C, Crosby A, et al. Hereditary spastic paraplegia:
145–150. clinical features and pathogenetic mechanisms. Lancet Neurol 2008;7:
209. Suchy S, Olivos-glander I, Nussbaum R. Lowe syndrome, a deficiency of 1127–1138.
phosphatidyl-inositol 4,5-biphosphate 5 phosphatase in the golgi appara- 234. Coutinho P, Barros J, Zemmouri R, et al. Clinical heterogeneity of auto-
tus. Hum Mol Genet 1995;4:2245–2250. somal recessive spastic paraplegias: analysis of 106 patients in 46 families.
210. Attree O, Olivos M, Okabe I, et al. The Lowe’s oculocerebrorenal syndrome Arch Neurol 1999;56(8):943–949.
gene encodes a protein highly homolgous to inositol polyphosphate-5- 235. Franca Jr MC, D’Abreu A, Maurer-Morelli CV, et al. Prospective neuroim-
phosphatase. Nature 1992;358:239–242. aging study in hereditary spastic paraplegia with thin corpus callosum.
211. Choudhury R, Diao A, Zhang F, et al. Lowe Syndrome Protein OCRL1 Mov Disord 2007;22(11):1556–1562.
Interacts with Clathrin and Regulates Protein Trafficking between 236. Murillo FM, Kobayashi H, Pegoraro E, et al. Genetic localization of a new
Endosomes and the Trans-Golgi Network. Mol Biol Cell 2005;16(8): locus for recessive familial spastic paraparesis to 15q13–15. Neurology
3467–3479. 1999;53(1):50.
212. Charnas L, Bernar J, Pezeshkpour GH, et al. MRI findings and peripheral 237. Hehr U, Bauer P, Winner B, et al. Long-term course and mutational
neuropathy in Lowe’s syndrome. Neuropediatrics 1988;19:7–9. spectrum of spatacsin-linked spastic paraplegia. Ann Neurol 2007;62(6):
213. Lowe CU, Terrey M, MacLachlan EA. Organic-aciduria, decreased renal 656–665.
ammonia production, hydrophthalmos, and mental retardation: a clinical 238. Schüle R, Schlipf N, Synofzik M, et al. Frequency and phenotype of SPG11
entity. Am J Dis Child 1952;83:164–184. and SPG15 in complicated hereditary spastic paraplegia. J Neurol Neuro-
214. Gropman A, Levin S, Yao L, et al. Unusual renal features of Lowe syndrome surg Psychiatry 2009;80(12):1402–1404.
in a mildly affected boy. Am J Med Genet 2000;95:461–466. 239. Ölmez A, Uyanik G, Özgül RK, et al. Further clinical and genetic charac-
215. O’Tauma DA, Lasker DW. Oculocerebrorenal syndrome: case report with terization of SPG11: hereditary spastic paraplegia with thin corpus cal-
CT and MR correlates. Am J Neuroradiol 1987;8:555–557. losum. Neuropediatrics 2006;2:59–66.
216. Carroll WJ, Woodruff WW, Cadman TE. MR findings in oculocerebrore- 240. Stevanin G, Azzedine H, Denora P, et al. Mutations in SPG11 are frequent
nal syndrome. Am J Neuroradiol 1993;14:449–451. in autosomal recessive spastic paraplegia with thin corpus callosum,
217. Schneider JF, Boltshauser E, Neuhaus TJ, et al. MRI and proton spectros- cognitive decline and lower motor neuron degeneration. Brain 2007:
copy in Lowe Syndrome. Neuropediatrics 2001;32:45–48. awm293.

Barkovich_Chap03.indd 220 5/6/2011 12:09:35 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 221

241. Hourani R, El-Hajj T, Barada WH, et al. MR imaging findings in auto- localized dynamic susceptibility-weighted contrast-enhanced perfusion
somal recessive hereditary spastic paraplegia. Am J Neuroradiol 2009;30: MR imaging measurements. Am J Neuroradiol 2009;30:552–558.
936–940. 266. Gaensler EH, Dillon WP, Edwards MSB, et al. Radiation-induced telangi-
242. Iannaccone ST, Rosenberg RN. Principles of molecular genetics and neu- ectasia in the brain similates cryptic vascular malformations at MR imag-
rologic diseases. In: Berg BO, ed. Principles of Child Neurology. New York: ing. Radiology 1994;193:629–636.
McGraw-Hill, 1996:461–606. 267. Young-Poussaint T, Siffert J, Barnes PD, et al. Hemorrhagic vasculopathy
243. Lossos A, Khoury M, Rizzo WB, et al. Phenotypic variability among adult after treatment of central nervous system neoplasia in childhood: diagno-
siblings with Sjogren-Larsson syndrome. Arch Neurol 2006;63:278–280. sis and follow-up. Am J Neuroradiol 1995;16:693–699.
244. De Laurenzi VD, Rogers GR, Hamrock DJ, et al. Sjögren-Larsson syn- 268. Koike S, Aida N, Hata M, et al. Asymptomatic radiation-induced telangi-
drome is caused by mutations in the fatty aldehyde degydrogenase gene. ectasia in children after cranial irradiation: frequency, latency, and dose
Nature Genet 1996;12:52–57. relation. Radiology 2004;230(1):93–99.
245. Sylvester PE. Pathological findings in Sjögren-Larsson syndrome. J Ment 269. Ciricillo SF, Cogen PH, Edwards MSB. Pediatric cryptic vascular mal-
Defic Res 1969;13:267–275. formations: presentation, diagnosis and treatment. Pediatr Neurosurg
246. Willemsen M, van der Graaf M, van der Knaap MS, et al. MR imaging and 1994;20:137–147.
proton MR spectroscopic studies in Sjogren-Larsson syndrome: character- 270. Scott R, Barnes P, Kupsky W, et al. Cavernous angiomas of the central ner-
ization of the leukoencephalopathy. Am J Neuroradiol 2004;25:649–657. vous system in children. J Neurosurg 1992;76:38–46.
247. Mano T, Ono J, Kaminga T, et al. Proton MR spectroscopy of Sjögren- 271. Wright TL, Bresnan MJ. Radiation induced cerebrovascular disease in
Larsson’s syndrome. Am J Neuroradiol 1999;20:1671–1673. children. Neurology 1976;26:540–543.
248. Sijens PE, Westerlaan HE, de Groot JC, et al. MR spectroscopy and diffu- 272. Painter MJ, Chutorian AM, Hilal SK. Cerebrovasculopathy following irra-
sion tensor imaging of the brain in Sjögren-Larsson syndrome. Mol Genet diation in dhildhood. Neurology 1975;25:189–194.
Metab 2009;98:367–71. 273. Omura M, Aida N, Sekido K, et al. Large intracranial vessel occlusive
249. Willemsen M, Ijlst L, Steijlen P, et al. Clinical, biochemical and molecular vasculopathy after radiation therapy in children: clinical features and
genetic characteristics of 19 patients with the Sjogren-Larsson syndrome. usefulness of magnetic resonance imaging. Int J Radiat Oncol Biol Phys
Brain 2001;124:1426–1437. 1997;38:241–249.
250. Paris F, Fuks Z, Kang A, et al. Endothelial apoptosis as the primary lesion 274. Keene D, Johnston D, Grimard L, et al. Vascular complications of cranial
initiating intestinal radiation damage in mice. Science 2001;293:293–297. radiation. Child Nervous Sys 2006;22:547–555.
251. Folkman J, Camphausen K. What does radiotherapy do to endothelial 275. Cappelli C, Grill J, Raquin M, et al. Long term follow up of 69 patients
cells? Science 2001;293:227. treated for optic pathway tumours before the chemotherapy era. Arch Dis
252. Sundgren PC. MR spectroscopy in radiation injury. Am J Neuroradiol Child 1998;79:334–338.
2009;30:1469–1476. 276. Rea D, Brandsema JF, Armstrong D, et al. Cerebral arteriopathy in children
253. Marks JE, Baglan RJ, Prassad SC, et al. Cerebral radionecrosis: incidence with neurofibromatosis type 1. Pediatrics 2009;124:e476–e483.
and risk in relation to dose, time, fractionation, and volume. Int J Radiat 277. Maher CO, Raffel C. Early vasculopathy following radiation in a child with
Oncol Biol Phys 1981;7:243–252. medulloblastoma. Pediatr Neurosurg 2000;32:255–258.
254. Pollard JM, Gatti RA. Clinical radiation sensitivity with DNA repair disor- 278. Johnson D, McCabe MA, Nicholson HS, et al. Quality of long-term sur-
ders: an overview. Int J Radiat Oncol Biol Phys 2009;74(5):1323–1331. vival in young children with medulloblastoma. J Neurosurg 1994;80:
255. McKinnon PJ, Caldecott KW. DNA strand break repair and human genetic 1004–1010.
disease. Ann Rev Genom Hum Genet 2007;8(1):37–55. 279. Kaschten B, Flandroy P, Reznik M, et al. Radiation-induced gliosarcoma.
256. Ball WS Jr, Prenger EC, Ballard ET. Neurotoxicity of radio/chemotherapy J Neurosurg 1995;83:154–162.
in children: pathologic and MR correlation. Am J Neuroradiol 1992;13: 280. Pettorini BL, Park Y, Caldarelli M, et al. Radiation-induced brain tumours
761–776. after central nervous system irradiation in childhood: a review. Childs
257. Miot E, Hoffschir D, Alpetite C, et al. Experimental MR study of cerebral Nerv Syst 2008;24:793–805.
radiation injury: quantitative T2 changes over time and histopathologic 281. Fouladi M, Langston J, Mulhern R, et al. Silent lacunar lesions detected by
correlation. Am J Neuroradiol 1995;16:79–85. magnetic resonance imaging of children with brain tumors: a late sequela
258. Armstrong CL, Gyato K, Awadalla AW, et al. A critical review of the clinical of therapy. J Clin Oncol 2000;18(4):824.
effects of therapeutic irradiation damage to the brain: the roots of contro- 282. Kitajima M, Hirai T, Maruyama N, et al. Asymptomatic cystic changes in
versy. Neuropsychol Rev 2004;14:65–86. the brain of children after cranial irradiation: frequency, latency, and rela-
259. Burger PC, Boyko OB. The pathology of central nervous system radiation tionship to age. Neuroradiology 2007;49(5):411–417.
injury. In: Gutin PH, Liebel SA, Sheline GF, eds. Radiation Injury to the 283. Edmister WB, Lane JI, Gilbertson JR, et al. Tumefactive cysts: a delayed
Nervous System. New York: Raven, 1991:191–208. complication following radiosurgery for cerebral arterial venous malfor-
260. Valk PE, Dillon WP. Radiation injury of the brain. Am J Neuroradiol mations. Am J Neuroradiol 2005;26:1152–1157.
1991;12:45–62. 284. Berg BO. Principles of Child Neurology. New York: McGraw-Hill, 1996.
261. Sundgren PC, Fan X, Weybright P, et al. Differentiation of recurrent brain 285. Pääkkö E, Talvensaari K, Pyhtinen J, et al. Decreased pituitary gland height
tumor versus radiation injury using diffusion tensor imaging in patients after radiation treatment to the hypothalamic-pituitary axis evaluated by
with new contrast-enhancing lesions. Magn Reson Imaging 2006;24: MR. Am J Neuroradiol 1994;15:537–541.
1131–1142. 286. Stevens SK, Moore SG, Kaplan ID. Early and late bone marrow changes
262. Sundgren PC, Nagesh V, Elias A, et al. Metabolic alterations: a biomarker after irradiation: MR evaluation. Am J Roentgenol 1990;154:745–750.
for radiation-induced normal brain injury-an MR spectroscopy study. 287. Cavenagh E, Weinberger E, Shaw D, et al. Hematopoietic marrow regen-
J Magn Reson Imaging 2009;29:291–297. eration in pediatric patients undergoing spinal irradiation: MR depiction.
263. Chan MSM, Roebuck DJ, Yuen M-P, et al. MR imaging of the brain in Am J Neuroradiol 1995;16:461–467.
patients cured of acute lymphblastic leukemia—the value of gradient echo 288. Orlandini G, Ruggiero C, Gulisano M, et al. MR evaluation of bone mar-
imaging. Am J Neuroradiol 2006;27:548–552. row in vertebral bodies. Arch Ital Anat Embriol 1991;96:93–100.
264. Khong P, Kwong D, Chan G, et al. Diffusion-tensor imaging for the 289. Stevens S, Moore S, Amylon M. Repopulation of marrow after transplanta-
detection and quantification of treatment-induced white matter injury tion: MR imaging with pathologic correlation. Radiology 1990;175:213–218.
in children with medulloblastoma: a pilot study. Am J Neuroradiol 290. Glass JP, Lee YY, Bruner J, et al. Treatment-related leukoencephalopathy.
2003;24:734–740. Medicine 1986;65:154–162.
265. Hu L, Baxter L, Smith K, et al. Relative cerebral blood volume values to 291. Lien HH, Blomlie V, Saeter G, et al. Osteogenic sarcoma: MR signal abnor-
differentiate high-grade glioma recurrence from posttreatment radiation malities of the brain in asymptomatic patients treated with high dose
effect: direct correlation between image-guided tissue histopathology and methotrexate. Radiology 1991;179:547–550.

Barkovich_Chap03.indd 221 5/6/2011 12:09:35 AM


222 Pediatric Neuroimaging

292. Mahoney DH Jr, Shuster JJ, Nitschke R, et al. Acute neurotoxicity in 314. Henneke M, Preuss N, Engelbrecht V, et al. Cystic leukoencephalopathy
children with B-precursor acute lymphoid leukemia: an association with without megalencephaly: a distinct disease entity in 15 children. Neurology
intermediate-dose intravenous methotrexate and intrathecal triple therapy— 2005;64:1411–1416.
a Pediatric Oncology Group study. J Clin Oncol 1998;16(5):1712–1722. 315. Grosso S, Cerase A, De Stefano N, et al. Non-progressive leukoencephal-
293. Rollins N, Winick N, Bash R, et al. Acute methotrexate neurotoxicity: find- opathy with bilateral anterior temporal cysts: a case report and review of
ings on diffusion-weighted imaging and correlation with clinical outcome. the literature. Brain Develop 2005;27(1):73.
Am J Neuroradiol 2004;25(10):1688–1695. 316. Aicardi J, Goutieres F. A progressive familial encephalopathy in infancy
294. Fisher MJ, Khademian ZP, Simon EM, et al. Diffusion-weighted MR imag- with calcifications of the basal ganglia and chronic cerebrospinal fluid
ing of early methotrexate-related neurotoxicity in children. Am J Neurora- lymphocytosis. Ann Neurol 1984;15:49–54.
diol 2005;26(7):1686–1689. 317. Alarcon-Riquelme ME. Nucleic acid by-products and chronic inflamma-
295. Brandsma D, Stalpers L, Taal W, et al. Clinical features, mechanisms, and tion. Nat Genet 2006;38(8):866–867.
management of pseudoprogression in malignant gliomas. Lancet Oncol 318. Crow YJ, Leitch A, Hayward BE, et al. Mutations in genes encoding ribo-
2008;9:453–461. nuclease H2 subunits cause Aicardi-Goutieres syndrome and mimic con-
296. Ebner F, Ranner G, Slavc I, et al. MR findings in methotrexate-induced genital viral brain infection. Nat Genet 2006;38(8):910–916.
CNS abnormalities. Am J Neuroradiol 1989;10:959–964. 319. Crow YJ, Rehwinkel J. Aicardi-Goutieres syndrome and related pheno-
297. Wilson CA, Nitschke R, Bowman ME, et al. Transient white matter changes types: linking nucleic acid metabolism with autoimmunity. Hum Mol
on MR images in children undergoing chemotherapy for acute lympho- Genet 2009;18:R130–R136.
cytic leukemia: Correlation with neuropsychologic deficiencies. Radiology 320. Goutières F. Aicardi-Goutières syndrome. Brain Dev 2005;27(3):201–206.
1991;180:205–209. 321. Uggetti C, La Piana R, Orcesi S, et al. Aicardi-Goutieres syndrome: Neuro-
298. Rubnitz JE, Relling MV, Harrison PL, et al. Transient encephalopathy fol- radiologic findings and follow-up. Am J Neuroradiol 2009;30:1971–1976.
lowing high-dose methotrexate treatment in childhood acute lympho- 322. Vivarelli R, Grosso S, Cioni M, et al. Pseudo-TORCH syndrome or
blastic leukemia. Leukemia 1998;12(8):1176–1181. Baraitser-Reardon syndrome: diagnostic criteria. Brain Dev 2001;23:18–23.
299. Reddick WE, Glass JO, Helton KJ, et al. Prevalence of leukoencephalopa- 323. Reardon W, Hockey A, Silberstein P, et al. Autosomal recessive congenital
thy in children treated for acute lymphoblastic leukemia with high-dose intrauterine infection-like syndrome of microcephaly, intracranial calcifi-
methotrexate. Am J Neuroradiol 2005;26:1263–1269. cation, and CNS disease. Am J Med Genet 1994;52:58–65.
300. Sandoval C, Kutscher M, Jayabose S, et al. Neurotoxicity of intrathecal 324. Baraitser M, Brett EM, Piesowicz AT. Microcephaly and intracranial calci-
methotrexate: MR imaging findings. Am J Neuroradiol 2003;24:1887–1890. fication in two brothers. J Med Genet 1983;20:210–212.
301. Chu WC, Chik KW, Chan YL, et al. White matter and cerebral metabolite 325. Henning KA, Li L, Iyer N, et al. The Cockayne syndrome group A gene
changes in children undergoing treatment for acute lymphoblastic leuke- encodes a WD repeat protein that interacts with CSB protein and a sub-
mia: longitudinal study with MR imaging and 1H MR spectroscopy. Radi- unit of RNA polymerase II TFIIH. Cell 1995;82(4):555.
ology 2003;229(3):659–669. 326. Cleaver JE, Lam ET, Revet I. Disorders of nucleotide excision repair: the genetic
302. Oka M, Terae S, Kobayashi R, et al. MRI in methotrexate-related leukoen- and molecular basis of heterogeneity. Nat Rev Genet. 2009;10:756–768.
cephalopathy: Disseminated necrotising leukoencephalopathy in compari- 327. Guzezetta F. Cockayne-Neill-Dingwall syndrome. In: Vinken PJ, Bruyn
son with mild leukoencephalopathy. Neuroradiology 2003;45(7):493–497. GW, eds. Handbook of Clinical Neurology. vol 13. Amsterdam: North
303. Brown MS, Simon JH, Stemmer SM, et al. MR and proton spectroscopy Holland, 1972:431–440.
of white matter disease induced by high dose chemotherapy with bone 328. Brumback RA. Cockayne’s syndrome diagnosis. Neurology 1984;34:
marrow transplant in advanced breast carcinoma. Am J Neuroradiol 842–843.
1995;16:2013–2020. 329. Demaerel P, Kendall BE, Kingsley D. Cranial CT and MRI in diseases with
304. Van Tassel P, Bruner J, Maor M, et al. MR of toxic effects of accelerated DNA repair defects. Neuroradiology 1992;34:117–121.
fractionation radiation therapy and carboplatin chemotherapy for malig- 330. Demaerel P, Wilms G, Verdru P, et al. MRI in the diagnosis of Cockayne’s
nant gliomas. Am J Neuroradiol 1995;16:715–726. syndrome. One case. J Neuroradiol 1990;17(2):157–160.
305. Kay HEM, Knapton PJ, O’Sullivan JP, et al. Encephalopathy in acute 331. Boltshauser E, Yalcinkaya C, Wichmann W, et al. MRI in Cockayne syn-
leukemia associated with methotrexate therapy. Arch Dis Child 1972;47: drome type I. Neuroradiology 1989;31:276–277.
344–354. 332. Bi WL, Baehring JM, Lesser RL. Evolution of brain imaging abnormalities
306. van der Knaap MS, Barth PG, Stroink H, et al. Leukoencephalopathy with in mitochondrial encephalomyopathy with lactic acidosis and stroke-like
swelling and a discrepantly mild clinical course in eight children. Ann episodes. J Neuro Ophthalmol 2006;26(4):251–256.
Neurol 1995;37:324–334. 333. Chen CC, Chiu PC, Shieh KS. Type 1 GM1 gangliosidosis with basal gan-
307. Topçu M, Saatci I, Topcuoglu MA, et al. Megalencephaly and leukodystro- glia calcification: a case report. Chung Hua i Hsueh Tsa Chih—Chinese
phy with mild clinical course: a report on 12 new cases. Brain Dev Med J 1999;62(1):40–45.
1998;20:142–153. 334. Schulz PE, Weiner SP, Belmont JW, et al. Basal ganglia calcifications in a
308. Topçu M, Gartioux C, Ribierre F, et al. Vacuolating megalencephalic leu- case of biotinidase deficiency. Neurology 1988;38(8):1326–1328.
koencephalopathy with subcortical cysts, mapped to chromosome 22qtel. 335. Billard G, Dulac O, Bouloche J, et al. Encephalopathy with calcifications of
Am J Hum Genet 2000;66:733–739. the basal ganglia in children. Neuropediatrics 1989;20:12–19.
309. Leegwater P, Yuan B, van der Steen J, et al. Mutations of MLC1 (KIAA0027), 336. Boltshauser E, Steinlin M, Boesch CH, et al. MRI in infantile encephal-
encoding a putative membrane protein, cause megalencephalic leukoen- opathy with cerebral calcification and leukodystrophy. Neuropediatrics
cephalopathy with subcortical cysts. Am J Hum Genet 2001;68:831–838. 1991;22:33–35.
310. Duarri A, Teijido O, López-Hernández T, et al. Molecular pathogenesis of 337. Razavi-Encha FJ, Larroche J-C, Gaillard D. Infantile familial encephal-
megalencephalic leukoencephalopathy with subcortical cysts: mutations opathy with cerebral calcifications and leukodystrophy. Neuropediatrics
in MLC1 cause folding defects. Hum Mol Genet 2008;17:3728–3739. 1988;19:72–79.
311. van der Knaap M, Barth P, Vrensen G, et al. Histopathology of an infantile- 338. Snodgrass J, Anhuf D, Schuler H, et al. Abnormalities of carbohydrate
onset spongiform leukoencephalopathy with a discrepantly mild clinical metabolism. In: Berg B, ed. Neurologic Aspects of Pediatrics. Boston, MA:
course. Acta Neuropathol (Berl) 1996;1996:206–212. Butterworth-Heinemann, 1992:93–124.
312. Miles L, deGrauw T, Dinopoulos A, et al. Megalencephalic leukoenceph- 339. Donnell GN, Collado M, Koch R. Growth and development of children
alopathy with subcortical cysts: a third confirmed case with literature with galactosemia. J Pediatr 1961;58:836–839.
review. Pediatr Dev Pathol 2009;12:180–186. 340. Lo W, Packman S, Nash S, et al. Curious neurologic sequelae in galac-
313. Gelal F, Calli C, Apaydin M, et al. van der Knaap’s leukoencephalopathy: tosemia. Pediatrics 1964;73:309–312.
report of five new cases with emphasis on diffusion-weighted MRI find- 341. Nelson MD Jr, Wolff JA, Cross CA, et al. Galactosemia: evaluation with
ings. Neuroradiology 2002;44:625–630. MR imaging. Radiology 1992;184:255–261.

Barkovich_Chap03.indd 222 5/6/2011 12:09:35 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 223

342. Möller HE, Ulrich K, Vermathen P, et al. In vivo study of brain metabolism 368. Gow A, Lazzarini RA. A cellular mechanism governing the severity of
in galactosemia by H-1 and P-31 magnetic resonance spectroscopy. Eur J Pelizaeus-Merzbacher disease. Nature Genetics 1996;13:422–428.
Pediatr 1995;154(Suppl 2):S8–S13. 369. Hobson GM, Huang Z, Sperle K, et al. A PLP splicing abnormality is
343. Wang ZJ, Berry GT, Dreha SF, et al. Proton magnetic resonance spectros- associated with an unusual presentation of PMD. Ann Neurol 2002;52(4):
copy of brain metabolites in galactosemia. Ann Neurol 2001;50:266–269. 477–488.
344. Moolenaar SH, van der Knaap MS, Engelke UFH, et al. In vivo and in vitro 370. Barnes DM, Enzmann D. The evolution of white matter diseases as seen
NMR spectroscopy reveal a putative novel inborn error involving polyol on computed tomography. Radiology 1981;138:379–383.
metabolism. NMR Biomed 2001;14(3):167–176. 371. Journel H, Roussey M, Gandon Y, et al. MR imaging in Pelizaeus-
345. Barkovich AJ. Myelin mishaps. Ann Neurol 2007;62:107–109. Merzbacher disease. Neuroradiology 1987;29:403–405.
346. Seitelberger F. Pelizaeus-Merzbacher disease. In: Vinken P, Bruyn G, eds. 372. Penner MW, Li KC, Gebarski SS, et al. MR imaging of Pelizaeus-Merzbacher
Handbook of Clinical Neurology. vol 10. Amsterdam: Elsevier-North Hol- disease. J Comput Assist Tomogr 1987;11:591–593.
land, 1970:150–202. 373. Wang P-J, Young C, Liu H-M, et al. Neurophysiologic studies and MRI in
347. Lazzarini A, Schwarz KO, Jiang S, et al. Pelizaeus-Merzbacher-like disease: Pelizaeus-Merzbacher disease: comparison of classic and connatal forms.
exclusion of the proteolipid protein locus and documrntation of a new Pediatr Neurol 1995;12:47–53.
locus on Xq. Neurology 1997;49:824–832. 374. Takanashi J, Sugita K, Tanabe Y, et al. MR-revealed myelination in the
348. Bugiani M, Al Shahwan S, Lamantea E, et al. GJA12 mutations in chil- cerebral corticospinal tract as a marker for Pelizaeus-Merzbacher’s dis-
dren with recessive hypomyelinating leukoencephalopathy. Neurology ease with proteolipid protein gene duplication. Am J Neuroradiol 1999;20:
2006;67(2):273–279. 1822–1828.
349. Dabbagh O, Swaiman KF. Cockayne syndrome: MRI correlates of hypo- 375. Hanefeld FA, Brockmann K, Pouwels PJW, et al. Quantitative proton MRS
myelination. Pediatr Neurol 1988;4(2):113–116. of Pelizaeus-Merzbacher disease. Evidence of dys- and hypomyelination.
350. Aula P, Autio S, Raivio KO. “Salla disease”: a new lysosomal storage disor- Neurology 2005;65:701–706.
der. Arch Neurol 1979;36:88–94. 376. Pizzini F, Fatemi A, Barker P, et al. Proton MR spectroscopic imaging in
351. Østergaard JR, Christensen T. The central nervous system in Tay syn- Pelizaeus-Merzbacher disease. Am J Neuroradiol 2003;24:1683–1689.
drome. Neuropediatrics 1996;27:326–330. 377. Menichella DM, Goodenough DA, Sirkowski E, et al. Connexins are critical
352. Tolmie JL, de Berker D, Dawber R, et al. Syndromes associated with tricho- for normal myelination in the CNS. J Neurosci 2003;23(13):5963–5973.
thiodystrophy. Clin Dysmorphol 1994;3:1–14. 378. Sijens PE, Rodiger LA, Meiners LC, et al. 1H magnetic resonance spectros-
353. Chen E, Cleaver JE, Weber CA, et al. Trichothiodystrophy: clinical spec- copy in monocarboxylate transporter 8 gene deficiency. J Clin Endocrinol
trum, central nervous system imaging, and biochemical characterization Metab 2008;93(5):1854–1859.
of two siblings. J Invest Dermatol 1994;103:154S–158S. 379. Friesema EC, Jansen J, Heuer H. Mechanisms of disease: psychomotor
354. Sasaki M, Takanashi J, Tada H, et al. Diffuse cerebral hypomyelination retardation and high T3 levels caused by mutations in monocarboxylate
with cerebellar atrophy and hypoplasia of the corpus callosum. Brain Dev transporter 8. Nat Clin Pract Endocrinol Metab 2006;2:512–523.
2009;31:582–587. 380. Toelle SP, Valsangiacomo E, Boltshauser E. Trichothiodystrophy with
355. Sistermans EA, de Coo RFM, De Wijs IJ, et al. Duplication of the pro- severe cardiac and neurologic involvement in two sisters. Eur J Pediatr
teolipid protein gene is the major cause of Pelizaeus-Merzbacher disease. 2001;160(12):728–731.
Neurology 1998;50:1749–1754. 381. Harreld JH, Smith EC, Prose NS, et al. Trichothiodystrophy with dysmyeli-
356. Inoue K, Osaka H, Sugiyama N, et al. A duplicated PLP causing Pelizaeus- nation and central osteosclerosis. Am J Neuroradiol 2010;31:129–130.
Merzbacher disease detected by comparative meltiplex PCR. Am J Hum 382. Rebora A, Crovato F. PIBI(D)S syndrome - trichothiodystriphy with
Genet 1996;59:32–39. xeroderma pigmentosum (group D) mutation. J Am Acad Dermatol
357. Koeppen AH, Ronca NA, Greenfield E, et al. Defective biosynthesis of 1987;16:940–947.
proteolipid protein in Pelizaeus-Merzbacher disease. Ann Neurol 1987;21: 383. Battistella PA, Peserico A. Central nervous system dysmyelination in
159–170. PIBI(D)S syndrome: a further case. Child Nerv Syst 1996;12:110–113.
358. Mattei MG, Alliel PM, Dautigny A, et al. The gene encoding for the major 384. Peserico A, Battistella PA, Bertoli P. MRI of a very rare hereditary ectoder-
brain proteolipid (PLP) maps on the q-22 band of the human X chromo- mal dysplasia: PIBI(D)S. Neuroradiology 1992;24:316–317.
some. Hum Genet 1986;72:352–353. 385. Yoon H-K, Sargent M, Prendiville J, et al. Cerebellar and cerebral atrophy
359. Pham Dinh D, Popot JL, Boespflug Tanguy O, et al. Pelizaeus-Merzbacher dis- in trichothiodystrophy. Pediatr Radiol 2005;35(10):1019.
ease: a valine to phenylalanine point mutation in a putative extracellular loop 386. Porto L, Weis R, Schulz C, et al. Tay’s syndrome: MRI. Neuroradiology
of myelin proteolipid. Proc Natl Acad Sci U S A 1991;88(17):7562–7566. 2000;42:849–851.
360. Inoue K. PLP1 related inherited dysmyelinating disorders: Pelizaeus- 387. Nakayama J, Hamano K, Shimakura Y, et al. Abnormal myelination in a
Merzbacher disease and spastic paraplegia type 2. Neurogenetics 2005;1: patient with ring chromosome 18. Neuropediatrics 1997;28:335–337.
1–16. 388. Lancaster JL, Cody JD, Andrews T, et al. Myelination in children with par-
361. Ziereisen F, Dan B, Christiaens F, et al. Connatal Pelizaeus-Merzbacher tial deletions of chromosome 18q. Am J Neuroradiol 2005;26:447–454.
disease in two girls. Pediatr Radiol 2000;30:435–438. 389. Linnankivi T, Autti T, Pihko S, et al. 18q-syndrome: brain MRI shows poor
362. Begleiter MM, Harris DJ. Autosomal recessive form of connatal Pelizaeus- differentiation of gray and white matter on T2-weighted images. J Magn
Merzbacher disease. Am J Med Genet 1989;33:311–313. Reson Imaging 2003;18:414–419.
363. Koeppen AH, Robitaille Y. Pelizaeus-Merzbacher disease. J Neuropathol 390. Bekiesinska-Figatowska M, Walecki J. MRI of the hypophysis in a patient
Exp Neurol 2002;61:747–759. with the 18q- syndrome. Neuroradiology 2001;43:875–876.
364. Inoue K, Osaka H, Imaizumi K, et al. Proteolipid protein gene duplica- 391. Hausler M, Anhuf D, Schuler H, et al. White matter disease in 18q deletion
tions causing Pelizaeus-Merzbacher disease: molecular mechanism and syndrome: magnetic resonance spectroscopy indicated demyelination or
phenotypic manifestations. Ann Neurol 1999;45:624–632. increased myelin turnover rather than dysmyelination. Neuroradiology
365. Takanashi J, Inoue K, Tomita M, et al. Brain N-acetylaspartate is ele- 2005;47:83–86.
vated in Pelizaeus-Merzbacher disease with PLP1 duplication. Neurology 392. Schleutker J, Laine AP, Haataja L, et al. Linkage disequilibrium utilized to
2002;58:237–241. establish a refined genetic position of the Salla disease locus on 6q14-q15.
366. Inoue K, Osaka H, Kawanish IC, et al. Mutations in the proteolipid protein Genomics 1995;27(2):286–292.
gene in Japanese families with Pelizaeus-Merzbacher disease. Neurology 393. Renlund M, Tietze F, Gahl WA. Defective sialic acid egress from isolated
1997;48:283–285. fibroblast lysosomes ofpatients with Salla disease. Science 1986;232:
367. Wolf NI, Sistermans EA, Cundall M, et al. Three or more copies of the 759–762.
proteolipid protein gene PLP1 cause severe Pelizaeus-Merzbacher disease. 394. Schleutker J, Leppanen P, Mansson JE, et al. Lysosomal free sialic acid stor-
Brain 2005;128(Pt 4):743–751. age dosorders with different phenotypic presentations—infantile form

Barkovich_Chap03.indd 223 5/6/2011 12:09:35 AM


224 Pediatric Neuroimaging

sialic acid storage disease and Salla disease—represent allelic disorders in 420. Enns GM, Barkovich AJ, van Kuilenburg ABP, et al. Head imaging
6q14–15. Am J Hum Genet 1995;57:893–901. abnormalities in dihydropyrimidine dehydrogenase deficiency. J Inherit
395. Hancock LW, Thaler MM, Horwitz AL, et al. Generalized Metab Dis 2004;27:513–522.
N-acetylneuraminic acid storage disease: quantitation and identification 421. Gibson KM, Breuer J, Nyhan WL. 3-hydroxy-3-methylglutaryl-
of the monosaccharide accumulating in brain and other tissues. J Neuro- coenzyme A lyase deficiency: review of 18 reported patients. Eur J Pediatr
chem 1982;38:803–809. 1988;148:180–186.
396. Mochel F, Yang B, Barritault J, et al. Free sialic acide storage disease with- 422. van der Knaap MS, Bakker HD, Valk J. MR imaging and proton spectros-
out sialuria. Ann Neurol 2009;65:753–757. copy in 3-hydroxy-3-methylglutaryl coenzyme A lyase deficiency. Am J
397. Linnankivi T, Lonnqvist T, Autti T. A case of Salla disease with involve- Neuroradiol 1998;19:378–382.
ment of the cerebellar white matter. Neuroradiology 2003;45:107–109. 423. Wang Z, Zimmerman RA, Sauter R. Proton MR spectroscopy of the
398. Robinson R, Fensom A, Lake B. Salla disease – rare or underdiagnosed? brain: clinically useful information obtained in assessing CNS diseases in
Dev Med Child Neurol 1997;39:153–157. children. Am J Roentgenol 1996;167:191–199.
399. Varho T, Komu M, Sonninen P, et al. A new metabolite contributing 424. Barkovich AJ, Al Ali F, Rowley HA, et al. Imaging patterns of neonatal
to N-acetyl signal in H-1 MRS of the brain in Salla disease. Neurology hypoglycemia. Am J Neuroradiol 1998;19:523–528.
1999;52:1668–1672. 425. Reid E. Science in motion: common molecular pathological themes
400. Rossi A, Biancheri R, Zara F, et al. Hypomyelination and Congenital Cata- emerge in the hereditary spastic paraplegias. J Med Genet 2003;40:81–86.
ract: Neuroimaging Features of a Novel Inherited White Matter Disorder. 426. Al-Yahyaee S, Al-Gazali LI, De Jonghe P, et al. A novel locus for heredi-
Am J Neuroradiol. 2008;29(2):301–305. tary spastic paraplegia with thin corpus callosum and epilepsy. Neurology
401. Willems PJ, Gatti R, Darby JK, et al. Fucosidosis revisited: a review of 2006;66:1230–1234.
77 patients. Am J Med Genet 1991;38:111–131. 427. Fink JK. Hereditary spastic paraplegia. Curr Neurol Neurosci Rep
402. Prietsch V, Arnold S, Kraegeloh-Mann I, et al. Severe hypomyelination as 2006;6:65–76.
the leading neuroradiological sign in a patient with fucosidosis. Neurope- 428. Gould RM, Brady ST. Neuropathology: many paths lead to hereditary
diatrics 2008;39:51–54. spastic paraplegia. Curr Biol 2004;14:R903–R904.
403. Langan T, Pueschel S. Nonketotic hyperglycinemia: clinical, biochemical, 429. Renoux C, Vukusic S, Confavreux C. The natural history of multiple scle-
and therapeutic considerations. Curr Probl Pediatr 1983;13:1–30. rosis with childhood onset. Clin Neurol Neurosurg 2008;110:897–904.
404. Hamosh A, Johnston MV. Nonketotic hyperglycinemia. In: Scrivner CR, 430. Renoux C, Vukusic S, Mikaeloff Y, et al. Natural history of Multiple
Beaudet AL, Sly WS, et al., eds. The Metabolic and Molecular Bases of Inher- Sclerosis with Childhood Onset. N Engl J Med 2007;356:2603–2613.
ited Disease, 8th ed. vol II. New York: McGraw-Hill, 2001:2065–2078. 431. Han JS, Pohl D, Rensel M, et al. Differential diagnosis and evaluation in
405. Rossi S, Daniele I, Bastrenta P, et al. Early myoclonic encephalopathy and pediatric multiple sclerosis. Neurology 2007;68(Suppl 2):S13–S22.
nonketotic hyperglycinemia. Pediatr Neurol 2009;41:371–374. 432. Belman AL, Chitnis T, Renoux C, et al. Challenges in the classification of
406. Barshop BA, Haas RH. Abnormalities of amino acid metabolism. In: pediatric multiple sclerosis and future directions. Neurology 2007;68( (16
Berg BO, ed. Principles of Child Neurology. New York: McGraw-Hill, 1996: Suppl 2) ):S70–S74.
997–1048. 433. Hanefeld F, Bauer HJ, Christen H-J, et al. Multiple sclerosis in childhood:
407. Applegarth DA, Toone JR. Nonketotic hyperglycinemia (glycine encephal- report of 15 cases. Brain Dev 1991;13:410–416.
opathy): laboratory diagnosis. Molec Genet Metab 2001;74:139–146. 434. Callen DJA, Shroff MM, Branson HM, et al. MRI in the diagnosis of pedi-
408. Korman SH, Boneh A, Ichinohe A, et al. Persistent NKH with transient atric multiple sclerosis. Neurology 2009;72:961–967.
or absent symptoms and a homozygous GLDC mutation. Ann Neurol 435. McDonald W, Compston A, Edan G, et al. Recommended diagnostic
2004;56:139–143. criteria for multiple sclerosis: guidelines from the International Panel on
409. Valvanis A, Schubiger O, Hayek J. Computed tomography in nonketotic the diagnosis of multiple sclerosis. Ann Neurol 2001;50:121–127.
hyperglycinemia. Comput Radiol 1981;5:265–270. 436. Hahn CD, Shroff MM, Blaser SI, et al. MRI criteria for multiple sclerosis:
410. Press GA, Barshop BA, Haas RH, et al. Abnormalities of the brain in evaluation in a pediatric cohort. Neurology 2004;62:806–808.
nonketotic hyperglycinemia: MR manifestations. Am J Neuroradiol 437. Mehta R, Pike GB, Enzmann DR. Improved detection of enhancing and
1989;10:315–321. nonenhancing lesions of multiple sclerosis with magnetization transfer.
411. Viola A, Chabrol B, Nicoli F, et al. Magnetic resonance spectroscopy Am J Neuroradiol 1995;16:1771–1778.
study of glycine pathways in nonketotic hyperglycinemia. Pediatr Res 438. Loevner LA, Grossman RI, Cohen JA, et al. Microscopic disease in
2002;52:292–300. normal-appearing white matter on conventional MR images in patients
412. Khong PL, Lam BC, Chung BH, et al. Diffusion weighted MR imaging in with multiple sclerosis: assessment with magnetization-transfer methods.
nonketotic hyperglycinemia. Am J Neuroradiol 2003;24:1181–1183. Radiology. 1995;196:511–515.
413. Mourmans J, Majoie CB, Barth PG, et al. Sequential MR imaging changes 439. Cercignami M, Inglese M, Pagani E, et al. Mean diffusivity and fractional
in nonketotic hyperglycinemia. Am J Neuroradiol 2006;27:208–211. anisotropy histograms of patients with multiple sclerosis. Am J Neurora-
414. Heindel W, Kugel H, Roth B. Noninvasive detection of increased glycine diol 2001;22:952–958.
content by proton MR spectroscopy in the brains of two infants with non- 440. Rovaris M, Gawne-Cain ML, Wang L, et al. A comparison of conventional
ketotic hyperglycinemia. Am J Neurorad 1993;14:629–635. and fast spin echo sequences for the measurement of lesion load in mul-
415. Kuilenburg ABPV, Vreken P, Abeling NGGM, et al. Genotype and pheno- tiple sclerosis using a semi-automated contour technique. Neuroradiology
type in patients with dihydropyrimidine dehydrogenase deficiency. Hum 1997;39:161–165.
Genet 1999;V104(1):1. 441. Keiper MD, Grossman RI, Brunson JC, et al. The low sensitivity of fluid-
416. Braakhekke J, Renier W, gabreels F, et al. Dihydroprymidine dehydroge- attenuated inversion-recovery MR in the detection of multiple sclerosis of
nase deficiency. Neurological aspects. J Neurol Sci 1987;78:71–77. the spinal cord. Am J Neuroradiol 1997;18:1035–1039.
417. Christensen E, Cezanne I, Kjaergaard S, et al. Clinical variability in three 442. Cotton F, Weiner H, Jolesz F, et al. MRI contrast uptake in new lesions in
Danish patients with dihydropyrimidine dehydrogenase deficiency all relapsing-remitting MS followed at weekly intervals. Neurology 2003;60:
homozygous for the same mutation. J Inherit Metab Dis 1998;21(3): 640–646.
272–275. 443. Ebner F, Millner MM, Justich E. Multiple sclerosis in children: value of serial
418. Van Gennip A, Abeling N, Vreken P, et al. Inborn errors of pyrimidine MR studies to monitor patients. Am J Neuroradiol 1990;11:1023–1027.
degradation: clinical, biochemical and molecular aspects. J Inherit Metab 444. Osborn AG, Harnsberger HR, Smoker WR, et al. Multiple sclerosis in ado-
Dis 1997;20:203–213. lescents: CT and MR findings. Am J Neuroradiol 1990;11:489–494.
419. Franco DA, Greenberg HS. 5-FU multifocal inflammatory leukoenceph- 445. Waubant E, Chabas D, Okuda DT, et al. Difference in Disease Burden and
alopathy and dihydropyrimidine dehydrogenase deficiency. Neurology. Activity in Pediatric Patients on Brain Magnetic Resonance Imaging at
2001;56(1):110–112. Time of Multiple Sclerosis Onset vs Adult. Arch Neurol 2009;66:967–971.

Barkovich_Chap03.indd 224 5/6/2011 12:09:35 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 225

446. Glasier CM, Robbins MB, Davis PC, et al. Clinical, neurodiagnostic, and 471. Johnson RT, Griffin DE, Gendelman HE. Postinfectious encephalomyelitis.
MR findings in children with spinal and brain stem multiple sclerosis. Semin Neurol 1985;5:180–190.
AJNR: Am J Neuroradiol 1995;16:87–95. 472. Nasralla CAW, Pay N, Goodpasture HC, et al. Postinfectious encephalopa-
447. Cianfoni A, Niku S, Imbesi SG. Metabolite findings in tumefactive demy- thy in a child following Campylobacter jejuni enteritis. Am J Neuroradiol
elinating lesions utilizing short echo time proton magnetic resonance 1993;14:444–448.
spectroscopy. Am J Neuroradiol 2007;28:272–277. 473. Kaye E, van der Knaap MS. Disorders primarily of white matter. In:
448. Cha S, George A. How much asymmetry should be considered normal Swaiman KF, Ashwal S, Ferriero DM, eds. Pediatric Neurology: Principles
variation or within normal range in asymmetrical frontal horns of the lat- and Practice. Philadelphia: Mosby Elsevier, 2006:1345–1373.
eral ventricles noted during CT brains scans without evidence of midline 474. DiMario FJ, Younkin D. Disorders of the respiratory system. In: Berg B,
shift or any other significant lesion? Am J Roentgenol 2002;178:240. ed. Neurologic Aspects of Pediatrics. Boston: Butterworth-Heinemann,
449. Kidd D, Barkhof F, McConnell R, et al. Cortical lesions in multiple sclero- 1992:339–356.
sis. Brain 1999;122:17–26. 475. Sriram S, Steinman L. Postinfectious and postvaccinial encephalomyelitis.
450. Peterson JW, Bo L, Mork S, et al. Transected neurites, apoptotic neurons, Neurol Clin 1984;2:341–353.
and reduced inflammation in cortical multiple sclerosis lesions. Ann 476. Idrissova Z, Boldyreva M, Dekonenko E, et al. Acute disseminated enceph-
Neurol 2001;50:389–400. alomyelitis in children: clinical features and HLA-DR linkage. Eur J Neurol
451. Guo A, Jewells V, Provenzale JM. Analysis of normal-appearing white mat- 2003;10:537–546.
ter in multiple sclerosis: comparison of diffusion tensor MR imaging and 477. Dale rC, de Sousa C, Chong WK, et al. Acute disseminated encephalomy-
magnetization transfer imaging. Am J Neuroradiol 2001;22:1893–1900. elitis, multiphasic disseminated encephalomyelitis and multiple sclerosis
452. Guo A, MacFall J, Provenzale J. Multiple sclerosis: diffusion tensor MR in children. Brain 2000;123:2407–2422.
imaging for evaluation of normal-appearing white matter. Radiology 478. Krupp L, MacAllister W, International Pediatric MS Study Group. Con-
2002;222:729–736. sensus definitions of acquired CNS demyelinating disorders of childhood.
453. Rovaris M, Gass A, Bammer R, et al. Diffusion MRI in multiple sclerosis. Mult Scler 2006;16(Suppl 1):S23.
Neurology 2005;65:1526–1532. 479. Hartung H, Grossman RI. ADEM: distinct disease or part of the MS spec-
454. Ge Y. Multiple sclerosis: the role of MR imaging. Am J Neuroradiol trum? Neurology 2001;56:1257–1260.
2006;27:1165–1176. 480. Mikaeloff Y, Suissa S, Vallee L, et al. First episode of acute CNS inflamma-
455. Redpath TW, Smith FW. Technical note: use of a double inversion recovery tory demyelination in childhood: prognostic factors for multiple sclerosis
pulse sequence to image selectively grey or white brain matter. Br J Radiol and disability. J Pediatr 2004;144(2):246–252.
1994;67:1258–1263. 481. Okuno T, Fuseya Y, Ito M, et al. Reversible multiple hypodense areas in
456. Wattjes MP, Lutterbey GG, Gieseke J, et al. Double inversion recovery white matter diagnosed as acute disseminated encephalomyelitis. J Com-
brain imaging at 3T: diagnostic value in the detection of multiple sclerosis put Assist Tomogr 1981;5:119–121.
lesions. Am J Neuroradiol 2007;28:54–59. 482. Atlas SW, Grossman RI, Goldberg HI, et al. MR diagnosis of acute dissemi-
457. Mezzapesa D, Rocca M, Falini A, et al. A preliminary diffusion tensor and nated encephalomyelitis. J Comput Assist Tomogr 1986;10(5):798–801.
magnetization transfer magnetic resonance imaging study of early-onset 483. Baum P, Barkovich AJ, Koch T, et al. Deep gray matter involvement in
multiple sclerosis. Arch Neurol 2004;61:366–368. children with acute disseminated encephalomyelitis. Am J Neuroradiol
458. Vishwas MS, Chitnis T, Pienaar R, et al. Tract-based analysis of callosal, 1994;15:1275–1283.
projection, and association pathways in pediatric patients with multiple 484. Ohtaki E, Murakami Y, Komori H, et al. Acute disseminated encephalo-
sclerosis. Am J Neuroradiol 2010;31:121–128. myelitis after Japanese B encephalitis vaccination. Pediatr Neurol 1992;8:
459. Dousset V, Grossman RI, Ramer KN, et al. Experimental allergic encepha- 137–139.
lomyelitis and multiple sclerosis: lewion characterization with magnetiza- 485. Murthy J. Acute disseminated encephalomyelitis. Neurol India 2002;50:
tion transfer imaging. Radiology 1992;182:483–491. 238–243.
460. Rocca MA, Mastronardo G, Rodegher M, et al. Long term changes of 486. Caldmeyer KS, Harris TM, Smith RR, et al. Gadolinium enhance-
magnetization transfer derived measures from patients with relapsing- ment in acute disseminated encephalomyelitis. J Comput Assist Tomogr
remitting and secondary progressive multiple sclerosis. Am J Neuroradiol 1991;15:673–675.
1999;20:821–827. 487. Honkaniemi J, Dastidar P, Kähärä V, et al. Delayed MR imaging changes
461. Sicotte NL, Voskuhl RR, Bouvier S, et al. comparison of multiple sclerosis in acute disseminated encephalomyelitis. Am J Neuroradiol 2001;22:
lesions at 1.5 and 3.0 Tesla. Invest Radiol 2003;38:423–427. 1117–1124.
462. Geurts JJG, Pouwels PJW, Uitdehaag BMJ, et al. Intracortical lesions in 488. Steinlin MI, Blaser SI, Gilday DL, et al. Neurologic manifestations of pedi-
multiple sclerosis: improved detection with 3D double inversion-recovery atric systemic lupus erythematosus. Pediatr Neurol 1995;13:191–197.
MR imaging. Radiology 2005;236:254–260. 489. Duprez TPJ, Grandin CBG, Bonnier C, et al. Whipple disease confined
463. Grossman RI, McGowan JC. Perspectives on multiple sclerosis. Am J Neu- to the central nervous system in childhood. Am J Neuroradiol 1996;17:
roradiol 1998;19:1251–1265. 1589–1591.
464. Saindane A, Cha S, Law M, et al. Proton MR spectroscphy of tumefactive 490. Simnad VI, Pisani DE, Rose JW. Multiple sclerosis presenting as transverse
demyelinating lesions. Am J Neuroradiol 2002;23:1378–1386. myelopathy: clinical and MRI features. Neurology 1997;48:65–73.
465. Bitsch A, Bruhn H, Vougioukas V, et al. Inflammatory CNS demyelination: 491. Hofer M, Weber A, Haffner K, et al. Acute hemorrhagic leukoencephalitis
histopathologic correlation with in vivo quantitative proton MR spectros- (Hurst’s disease) linked to Epstein-Barr virus infection. Acta Neuropathol
copy. Am J Neuroradiol 1999;20:1619–1627. 2005;109:226–230.
466. Lotze TE, Northrop JL, Hutton GJ, et al. Spectrum of pediatric neuromy- 492. Kabakus N, Gurgoze M, Yildirim H, et al. Acute hemorrhagic leukoen-
elitis optica. Pediatrics 2008;122:e1039–e1037. cephalitis manifesting as intracerebral hemorrhage associated with herpes
467. Dale RC, Brilot F, Banwell B. Pediatric central nervous system inflam- simplex virus type I. J Trop Pediatr 2005;51:245–249.
matory demyelination: acute disseminated encephalomyelitis, clinically 493. Geerts Y, Dehaene I, Lammens M. Acute hemorrhagic leukoencephalitis.
isolated syndromes, neuromyelitis optica, and multiple sclerosis. Curr Acta Neurol Belg 1991;91(4):201–211.
Opin Neurol 2009;22:233–240. 494. Leake JA, Billman GF, Nespeca MP, et al. Pediatric acute hemorrhagic
468. Hazin R, Khan F, Bhatti MT. Neuromyeliticus optica: current concepts and leukoencephalitis: report of a surviving patient and review. Clin Infect Dis
prospects for future management. Curr Opin Ophthalmol 2009;20:434–439. 2002;34:699–703.
469. Wingerchuk DM, Lennon VA, Lucchinetti CF, et al. The spectrum of neu- 495. Reich H, Lin SR, Goldblatt D. Computerized tomography in acute hemor-
romyelitis optica. Lancet Neurol 2007;6:805–815. rhagic leukoencephalopathy: case report. Neurology 1979;29:255–258.
470. Pittock SJ, Lennon VA, Krecke K, et al. Brain abnormalities in neuromyelitis 496. Lahita RG. Gender and age in lupus. In: Lahita RG, ed. Systemic Lupus
optica. Arch Neurol 2006;63:390–396. Erythematosus, 3rd ed. New York: Churchill Livingstone, 1999:129–144.

Barkovich_Chap03.indd 225 5/6/2011 12:09:35 AM


226 Pediatric Neuroimaging

497. Meislin AG, Rothfield N. Systemic lupus erythematosus in childhood. 525. Kawahara I, Tokunaga Y, Ishizaka S, et al. Reversible clinical and magnetic
Analysis of 42 cases with comparative data of 200 adult cases followed resonance imaging of central pontine myelinolysis following surgery for
concurrently. Pediatrics 1968;42:37–49. craniopharyngioma: serial magnetic resonance imaging studies. Neurol
498. Glidden RS, Mantzouranis EC, Borel Y. Systemic lupus erythematosus Med Chir (Tokyo) 2009;49:120–123.
in childhood: clinical manifestations and improved survival in fifty-five 526. Cramer SC, Stegbauer KC, Schneider A, et al. Decreased diffusion in cen-
patients. Clin Immunol Immunopathol 1983;29:196–210. tral pontine myelinolysis. Am J Neuroradiol 2001;22:1476–1479.
499. Parikh S, Swaiman KF, Kim Y. Neurologic characteristics of childhood 527. Ramaekers VT, Ruel J, Kusenbach G, et al. Central pontine myelinoly-
lupus erythematosus. Pediatr Neurol 1995;13:198–201. sis associated with acquired folate depletion. Neuropediatrics 1997;28:
500. Yancy CL, Doughty RA, Athreya BH. Central nervous system involve- 126–130.
ment in childhood systemic lupus erythematosus. Arthritis Rheum 528. Niehaus L, Kulozik A, Lehmann R. Reversible central pontine and extra-
1981;24:1389–1395. pontine myelinolysis in a 16-year-old girl. Childs Nerv Syst 2001;17:294–
501. Spiro AJ, Pack DR. Disorders of immunologic dysfunction. In: Berg B, ed. Neu- 296.
rologic Aspects of Pediatrics. Boston: Butterworth-Heinemann, 1992:501–527. 529. Haspolat S, Duman O, Senol U, et al. Extrapontine myelinolysis in infancy:
502. Kaell AT, Shetty M, Lee BCP, et al. The diversity of neurologic events in sys- report of a case. J Child Neurol 2004;19:913–915.
temic lupus erythematosus: prospective clinical and computed tomographic 530. Calakos N, Fischbein N, Baringer J, et al. Cortical MRI findings associated
classification of 82 events in 71 patients. Arch Neurol 1986;43:273–276. with rapid correction of hyponatremia. Neurology 2000;55:1048–1051.
503. DeMarco PJ, Szer IS. Systemic Lupus Erythematosus in Childhood. In: 531. Mangat K, Sherlala H. Cerebellar peduncle myelinolysis: case report.
Lahita RG, ed. Systemic Lupus Erythematosus, 3rd ed. New York: Churchill Neuroradiology 2002;44:768–769.
Livingstone, 1999:361–382. 532. Brunberg JA, Jacquemont S, Hagerman RJ, et al. Fragile X premuta-
504. Szer IS. The diagnosis and management of systemic lupus erythematosus tion carriers: characteristic MR imaging findings of adult male patients
in childhood. Pediatr Ann 1986;15:596–604. with progressive cerebellar and cognitive dysfunction. Am J Neuroradiol
505. Elby C. Antiphospholipid syndrome review. Clin Lab Med 2009;29:305–319. 2002;23:1757–1766.
506. Rojas-Villarraga A, Toro CE, Espinosa G, et al. Factors influencing 533. Okamoto, K, Tokiguchi S, Furusawa T, et al. MR features of diseases
polyautoimmunity in systemic lupus erythematosus. Autoimmun Rev involving bilateral middle cerebellar peduncles. Am J Neuroradiol 2003;24:
2010;9:229–232. 1946–1954.
507. Lalani TA, Kanne JP, Hatfiled GA, et al. Imaging findings in systemic lupus 534. Vinken PJ, Bruyn GW. Intoxications of the Central Nervous System. vol
erythematosus. Radiographics 2004;24(4):1069–1086. 36,37. New York: North Holland, 1979.
508. Oku K, Atsumi T, Furukawa S, et al. Cerebral imaging of magnetic reso- 535. Valk J, van der Knaap MS. Toxic encephalopathy. Am J Neuroradiol
nance imaging and single photon emission computed tomography in 1992;13:747–760.
systemic lupus erythmatosus with central nervous system involvement. 536. Lorenzo AV, Jolesz FA, Wallman JK, et al. Proton magnetic resonance stud-
Rheumatology (Oxford) 2003;42:773–777. ies of triethyltin-induced edema during perinatal brain development in
509. Hashimoto N, Handa H, Taki W. Ruptured cerebral aneurysms in patients rabbits. J Neurosurg 1989;70:432–440.
with systemic lupus erythematosus. Surg Neurol 1986;26:512–516. 537. Sempere AP, Posada I, Ramo C, et al. Spongform leucoencephalopathy
510. Yaffe K, Ferriero DM, Barkovich AJ, Rowley HA. Reversible MRI abnor- after inhaling heroin. Lancet 1991;338:320.
malities following seizures. Neurology 1995;45:104–108. 538. Tan TP, Algra PR, Valk J, et al. Toxic leukoencephalopathy after inhalation
511. Minagar A, DeToledo J, Falcone S. Cortical-subcortical lesions in “revers- of poisoned heroin: MR findings. Am J Neuroradiol 1994;15:175–178.
ible posterior leukoencephalopathy syndrome.” Encephalopathy or 539. Kriegstein AR, Armitage BA, Kim PY. Heroin inhalation and progressive
seizures? J Neurol 2001;248:537–540. spongiform leukoencephalopathy. N Eng J Med 1997;336:589–590.
512. Hinchey J, Chaves C, Appignani B, et al. A reversible posterior leukoen- 540. Dietemann J-L, Botelho C, Nogueira T, et al. Imagerie des encéphalopa-
cephalopathy syndrome. N Engl J Med 1996;334:494–500. thies toxiques aiguës. J Neuroradiol 2004;31:313–326.
513. Mukherjee P, McKinstry R. Reversible posterior leukoencephalopathy 541. Pappas CL, Quisling RG, Ballinger WE, et al. Lead encephalopathy: symp-
syndrome: evaluation with diffusion-tensor MR imaging. Radiology toms of a cerebellar mass lesion and obstructive hydrocephalus. Surg
2001;219:756–765. Neurol 1986;26:391–394.
514. Antunes N, Small T, George D, et al. Posterior leukoencephalopathy syn- 542. Tüzün M, Tüzün D, Salan A, et al. Lead encephalopathy: CT and MR find-
drome may not be reversible. Pediatr Neurol 1999;20:241–243. ings. J Comput Assist Tomogr 2002;26:479–481.
515. Lim MK, Suh CH, Kim JK, et al. Systemic lupus erythematosus: brain 543. Harrington JF, Mapstone TB, Selman WR, et al. Lead encephalopathy pre-
MR imaging and single-voxel hydrogen 1 MR spectroscopy. Radiology senting as a posterior fossa mass. J Neurosurg 1986;65:713–715.
2000;217:43–49. 544. Aydin K, Sencer S, Demir T, et al. Cranial MR findings in chronic toluene
516. Mortilla M, Ermini M, Nistri M, et al. Brain study using magnetic reso- abuse by inhalation. Am J Neuroradiol 2002;23:1173–1179.
nance imaging and proton MR spectroscopy in pediatric onset systemic 545. Xiong L, Matthes JD, Li J, et al. MR imaging of “spray heads”: toluene
lupus erythematosus. Clin Exp Rheumatol 2003;21:129–135. abuse via aerosol paint inhalation. Am J Neuroradiol 1993;14:1195–1199.
517. Adams rD, Victor M, Mancall EL. Central pontine myelinolysis. Arch 546. Yamanouchi N, Okada S, Kodama K, et al. White matter changes caused by
Neurol Psychiatry 1971;81:154. chronic solvent abuse. Am J Neuroradiol 1995;16:1643–1649.
518. Burcar PJ, Norenberg MD, Yarnell PR. Hyponatremia and central pontine 547. Kamran S, Bakshi R. MRI in chronic toluene abuse: low signal in the cere-
myelinolysis. Neurology 1977;27:223–227. bral cortex on T2 weighted images. Neuroradiology 1998;40:519–521.
519. Kumar S, Fowler M, Gonzalez-Toledo E, et al. Central pontine myelinoly- 548. Hallervorden J, Spatz H. Eigenartige: Erkrankung im extrapyramidallen
sis, an update. Neurol Res 2006;28:360–366. System mit besonderer Beteiligung des Globus pallidus und der Substantia
520. Cadman TE, Rorke LB. Central pontine myelinolysis in childhood and nigra. Z Neurol Psychiatry 1922;79:254–302.
adolescence. Arch Dis Child 1969;44:342–345. 549. Bindu PS, Desai S, Shehanaz KE, et al. Clinical heterogeneity in Haller-
521. Valsamis MP, Peress NS, Wright LD. Central pontine myelinolysis in child- vorden-Spatz syndrome: a clinicoradiological study in 13 patients from
hood. Arch Neurol 1971;25:307–310. South India. Brain Dev 2006;28:343–347.
522. Kato T, Hattori H, Nagato M, et al. Subclinical central pontine myelinoly- 550. Swaiman KF. Hallervorden-Spatz syndrome. Pediatr Neurol 2001;25:102–108.
sis following liver transplantation. Brain Dev 2002;24:179–182. 551. Taylor TD, Litt M, Kramer P, et al. Homozygosity mapping of Haller-
523. Sivaswamy L, Karia S. Extrapontine myelinolysis in a 4 year old with dia- vorden-Spatz syndrome to chromosome 20p12.3-p13. Nature Genet
betic ketoacidosis. Eur J Paediatr Neurol 2007;11:389–393. 1996;14:479–481.
524. Cardenas JF, Bodensteiner JB. Osmotic demyelination syndrome as a con- 552. Zhou B, Westaway SK, Levinson B, et al. A novel pantothenate kinase
sequence of treating hyperammonemia in a patient with ornithine tran- gene (PANK2) is defective in Hallervorden-Spatz syndrome. Nat Genet
scarbamylase deficiency. J Child Neurol 2009;24:884–886. 2001;28:345–349.

Barkovich_Chap03.indd 226 5/6/2011 12:09:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 227

553. Wu Z, Li C, Zhou B. Pantothenate kinase-associated neurodegeneration: 578. Vockley J, Parimoo B, Tanaka K. Molecular characterization of four dif-
insights from a Drosphila model. Hum Mol Genet 2009;18:3659–3672. ferent classes of mutations in the isovaleryl-CoA dehydrogenase gene
554. Hayflick S, Hartman M, Coryell J, et al. Brain MRI in neurodegeneration responsible for isovaleric acidemia. Am J Hum Genet 1991;49:147–157.
with brain iron accumulation with and without PANK2 mutations. Am J 579. Tanaka K, Budd MA, Efron ML, et al. Isovaleric acidemia: a new genetic
Neuroradiol 2006;27:1230–1233. defect of leucine metabolism. Proc Nat Acad Sci 1966;56:236–242.
555. Hayflick S, Westaway S, Levinson B, et al. Genetic, clinical and radiographic 580. Sweetman L, Williams J. Branched chain organic acidurias. In: Scriver C,
delineation of Hallervorden-Spatz syndrome. N Engl J Med 2003;348:33– Beaudet A, Sly W, et al., eds. The Metabolic and Molecular Bases of Inherited
40. Diseases. New York: McGraw-Hill, 2001:2125–2163.
556. Wigboldus JM, Bruyn GW. Hallervorden-Spatz disease. In: Vinken PJ, 581. Sogut A, Acun C, Aydin K, et al. Isovaleric acidaemia: cranial CT and MRI
Bruyn GW, eds. Handbook of Clinical Neurology: Diseases of Basal Ganglia. findings. Pediatr Radiol 2004;34(2):160–162.
vol 6. Amsterdam: North Holland, 1968:604–631. 582. Gibson K, Sweetman L, Kozich V, et al. Unusual enzyme findings in five
557. Rutledge JN, Hilal SK, Silver AJ, et al. Study of movement disorders and patients with metabolic profiles suggestive of succinic semialdehyde dehy-
brain iron by MR. Am J Neuroradiol 1987;8:397–411. drogenase deficiency (4-hydroxybutyric aciduria). J Inherit Metab Dis
558. Savoiardo M, Halliday WC, Nardocci N, et al. Hallervorden-Spatz disease: 1998;23:255–261.
MR and pathologic findings. Am J Neuroradiol 1993;14:155–162. 583. Chambliss K, Hinson D, Trettel F, et al. Two exon-skipping mutations as
559. Schaffert DA, Johhnsen SD, Johnson PC, et al. Magnetic resonance imag- the molecular basis of succinic semialdehyde dehydrogenase deficiency
ing in pathologically proven Hallervorden-Spatz disease. Neurology (4-hydroxybutyric aciduria). Am J Hum Genet 1998;63:399–408.
1989;39:440–442. 584. Akaboshi S, Hogema BM, Novelletto A, et al. Mutational spectrum of the
560. Hayflick SJ, Penzien JM, Michl W, et al. Cranial MRI changes may pre- succinate semialdehyde dehydrogenase (ALDH5A1) gene and functional
cede symptoms in Hallervorden-Spatz syndrome. Pediatr Neurol 2001;25: analysis of 27 novel disease-causing mutations in patients with SSADH
166–169. deficiency. Hum Mutat 2003;22:442–450.
561. Zolkipli Z, Dahmoush H, Saunders DE, et al. Pantothenate kinase 2 muta- 585. Ziyeh S, Berlis A, Korinthenberg R, et al. Selective involvement of the glo-
tion with classic pantothenate-kinase-associated neurodegeneration bus pallidus and dentate nucleus in succinic semialdehyde dehydrogenase
without ‘eye-of-the-tiger’ sign on MRI in a pair of siblings. Pediatr Radiol deficiency. Pediatr Radiol 2002;32:598–600.
2006;36:884–886. 586. Pearl PL, Novotny EJ, Acosta MT, et al. Succinic semialdehyde dehydrogenase
562. Valentino P, Annesi G, Cirò Candiano I, et al. Genetic heterogeneity in deficiency in children and adults. Ann Neurol 2003;54(Suppl 6):S73–S80.
patients with pantothenate kinase-associated neurodegeneration and 587. Blasi P, Palmerio F, Caldarola S, et al. Succinic semialdehyde dehydroge-
classic magnetic resonance imaging eye-of-the-tiger pattern. Mov Disord nase deficiency: clinical, biochemical and molecular characterization of
2006;21:252–254. a new patient with severe phenotype and a novel mutation (Letter). Clin
563. Baumeister FA, Auer D, Hortnagel K, et al. The eye of the tiger sign is not a Genet 2006;69:294–296.
reliable disease marker for Hallervorden-Spatz syndrome. Neuropediatrics 588. Yalçinkaya C, Gibson K, Gunduz E, et al. MRI findings in succinic semial-
2005;36:221–222. dehyde dehydrogenase deficiency. Neuropediatrics 2000;31:45–46.
564. Ginsberg LE, Jewett T, Grub R, et al. Oculodental digital dysplasia: neu- 589. Lee W-T, Weng W-C, Peng S-F, et al. Neuroimaging findings in chil-
roimaging in a kindred. Neuroradiology 1996;38:84–86. dren with paediatric neurotransmitter diseases. J Inherit Metabol Dis
565. Awasthi R, Gupta RK, Trivedi R, et al. Diffusion tensor MR imaging in chil- 2009;32(3):361–370.
dren with pantothenate kinase-associated neurodegeneration with brain iron 590. Stöckler S, Hanefeld F, Frahm J. Creatine replacement in guanidinoacetate
accumulation and their siblings. Am J Neuroradiol. 2010;31(3):442–447. methyltransferase deficiency, a novel inborn error of metabolism. Lancet
566. Sarper N, Akansel G, Aydogan M, et al. Neuroimaging abnormalities in 1996;348:789–790.
Griscelli’s disease. Pediatr Radiol 2002;32(12):875–878. 591. Item C, Stockler-Ipsiroglu S, Stromberger C, et al. Arginine: glycine ami-
567. Saillour Y, Zianni G, des Portes V, et al. Mutations in the AP1S2 gene dinotransferase deficiency: the third inborn error of creatine metabolism
encoding the sigma 2 subunit of the adaptor protein 1 complex AP1S2 are in humans. Am J Hum Genet 2001;69:1127–1133.
associated with syndromic X-linked mental retardation with hydrocepha- 592. van der Knaap MS, Verhoeven N, Maaswinkel-Mooij P, et al. Mental retar-
lus and calcifications in basal ganglia. J Med Genet 2007;44:739–744. dation and behavioral problems as presenting signs of a creatine synthesis
568. Goncalves FG, de Melo MB, de L. et al. Amygdalae and striatum calcifica- defect. Ann Neurol 2000;47:540–543.
tion in lipoid proteinosis. Am J Neuroradiol 2010;31(1):88–90. 593. Ganesan V, Johnson A, Connelly A, et al. Guanidinoacetate methyltrans-
569. Group HDCR. A novel gene containing a trinucleotide repeat that is ferase deficiency: new clinical features. Pediatr Neurol 1997;17:155–157.
expanded and unstable on Huntinton’s disease chromosomes. Cell 594. Schulze A, Hess T, Wevers R, et al. Creatine deficiency syndrome caused by
1993;72:971–983. guanidinoacetate methyltransferase deficiency: diagnostic tools for a new
570. Kremer B, Almqvist E, Theilmann J, et al. Sex-dependent mechanisms for inborn error of metabolism. J Pediatr 1997;131:626–631.
expansions and contractions of the CAG repeat on affected Huntington 595. Schulze A. Creatine deficiency syndromes. Molec Cell Biochem 2003;244:
disease chromosomes. Am J Hum Genet 1995;57:343–350. 143–150.
571. Telenius H, Kremer HP, Theilmann J, et al. Molecular analysis of juvenile 596. Vodopiutz J, Item CB, Häusler M, et al. Severe speech delay as the present-
Huntington disease: the major influence on (CAG)n repeat length is the ing symptom of guanidinoacetate methyltransferase deficiency. J Child
sex of the affected parent. Hum Mol Genet 1993;2:1535–1540. Neurol 2007;22:773–774.
572. Ho V, Chuang S, Rovira M, et al. Juvenile Huntington disease: CT and MR 597. Battini R, Alessandri MG, Leuzzi V, et al. Arginine:glycine amidinotrans-
features. Am J Neuroradiol 1995;16:1405–1412. ferase (AGAT) deficiency in a newborn: early treatment can prevent phe-
573. Gonzalez-Alegre P, Affifi AK. Clinical characteristics of childhood-onset notypic expression of the disease. J Pediatr 2006;148:828–830.
(juvenile) Huntington disease: report of 12 patients and review of the lit- 598. Bianchi MC, Tosetti M, Battini R, et al. Treatment monitoring of brain
erature. J Child Neurol 2006;21:223–229. creatine deficiency syndromes: A 1H- and 31P-MR spectroscopy study.
574. Gambardella A, Muglia M, Labate A, et al. Juvenile Huntingon’s disease Am J Neuroradiol 2007;28(3):548–554.
presenting as progressive myoclonic epilepsy. Neurology 2001;57:708–711. 599. Benedict SL. Diffusion-weighted imaging in diagnosing neurological dis-
575. Harris GJ, Pearlson GD, Peyser CE, et al. Putamen volume reduction on orders in children: a pediatric neurologist’s perspective. Pediatr Radiol
magnetic resonance imaging exceeds caudate changes in mild Hunting- 2007;37:734–738.
ton’s disease. Ann Neurol 1992;31:69–73. 600. Bianchi MC, Tosetti M, Fornai F, et al. Reversible brain creatine deficiency in
576. Kuhl DE. Cerebral metabolism and atrophy in Huntington’s disease deter- two sisters with normal blood creatine level. Ann Neurol 2000;47:511–513.
mined by 18FDG and CT scan. Ann Neurol 1982;12:425–431. 601. Salomons G, van Dooren S, Verhoeven N, et al. X-linked creatine-
577. Schapiro M, Cecil K, Doescher J, et al. MR imaging and spectroscopy in transporter gene (SLC6A8) defect: a new creatine-deficiency syndrome.
juvenile Huntington disease. Pediatr Radiol 2004;34:640–643. Am J Hum Genet 2001;68:1497–1500.

Barkovich_Chap03.indd 227 5/6/2011 12:09:36 AM


228 Pediatric Neuroimaging

602. Degrauw T, Salomons G, Cecil K, et al. Congenital creatine transporter 629. Castillo M, Kwock L, Arbelaez A. Sydenham’s chorea: MRI and proton
deficiency. Neuropediatrics 2002;33:232–238. spectroscopy. Neuroradiology 1999;41:943–945.
603. Battini R, Chilosi A, Mei D, et al. Mental retardation and verbal dyspraxia 630. Genovese E, Maghnie M, Maggiore G, et al. MR imaging of CNS involve-
in a new patient with de novo creatine transporter (SLC6A8) mutation. ment in children affected by chronic liver disease. Am J Neuroradiol
Am J Med Genet 2007;143A:1771–1774. 2000;21:845–851.
604. Clark A, Rosenberg E, Almeida L, et al. X-linked creatine transporter 631. Dyken P, Wisniewski K. Classification of the neuronal ceroid lipofuscino-
(SLC6A8) mutations in about 1% of males with mental retardation of ses: expansion of the atypical form. Am J Med Genet 1995;57:150–154.
unknown etiology. Hum Genet 2006;119:604–610. 632. Lauronen L, Santavuori P, Hirvasniemi A, et al. Northern epilepsy syn-
605. Newmeyer A, deGrauw T, Clark J, et al. Screening of male patients with drome (NES, CLN8)—MRI and electrophysiological studies. Eur J Pae-
autism spectrum disorder for creatine transporter deficiency. Neuropedi- diatr Neurol 2001;5(Suppl A):167–173.
atrics 2007;38:310–312. 633. Steinfeld R, Reinhardt K, Schreiber K, et al. Cathepsin D deficiency is
606. Vasconcelos MM, Silva KP, Vidal G, et al. Early diagnosis of pediatric Wer- associated with a human neurodegenerative disorder. Am J Hum Genet
nicke’s encephalopathy. Pediatr Neurol 1999;20:289–294. 2006;78:988–998.
607. Zuccoli G, Gallucci M, Capellades J, et al. Wernicke encephalopathy: MR 634. Järvelä I, Schleurker J, Haataja L, et al. Infantile neuronal ceroid lipofusci-
findings at clinical presentation in twenty-six alcoholic and nonalcoholic nosis (INCL, CLN1) maps to the short arm of chromosome 1. Genomics
patients. Am J Neuroradiol 2007;28:1328–1331. 1991;8:170–173.
608. Harter SB, Nokes SR. Gadolinium-enhanced MR findings in a pediatric 635. Liu C, Sleat D, Donnelly R, Lobel P. Structural organization and sequence
case of Wernicke encephalopathy. Am J Neuroradiol 1995;16:700–702. of CLN2, the defective gene in classical late infantile neuronal ceroid lipo-
609. Shogry MEC, Curnes JT. Mamillary body enhancement on MR as the only fuscinosis. Genomics 1998;50:206–212.
sign of acute Wernicke encephalopathy. Am J Neuroradiol 1994;15:172–174. 636. Savukoski M, Klockars T, Holmberg V, et al. CLN5, a novel gene encoding
610. Oka M, Terae S, Kobayashi R, et al. Diffusion weighted MR findings in a putative transmembrane protein mutated in Finnish variant late infan-
a reversible case of acute Wernicke encephalopathy. Acta Neurol Scand tile neuronal ceroid lipofuscinosis. Nat Genet 1998;19:286–288.
2001;104:178–181. 637. Wheeler R, Sharp J, Schultz R, et al. The gene mutated in variant late-
611. Coe M, Carfagnini F, Tani G, Ambrosetto P. Wernicke’s encephalopathy in infantile neuronal ceroid lipofuscinosis (CLN6) and in nclf mutant mice
a child: case report and MR findings. Pediatr Radiol 2001;31:167–168. encodes a novel predicted transmembrane protein. Am J Hum Genet
612. Brown W. Osmotic demyelination disorders: central pontine and extra- 2002;70:537–542.
pontine myelinolysis. Curr Opin Neurol 2000;13:691–697. 638. Siintola E, Topcu M, Aula N, et al. The novel neuronal ceroid lipofusci-
613. Brown W, Caruso J. Extrapontine myelinolysis with involvement of the nosis gene MFSD8 encodes a putative lysosomal transporter. Am J Hum
hippocampus in three children with sever hypernatremia. J Child Neurol Genet 2007;81:136–146.
1999;14:428–433. 639. Ranta S, Zhang Y, Ross B, et al. The neuronal ceroid lipofuscinoses in
614. Gianantonio CA, Vitaccio M, Mendilaharzu F, et al. The hemolytic uremic human EPMR and mnd mutant mice are associated with mutations in
syndrome. Nephron 1973;11:174–192. CLN8. Nat Genet 1999;23:233–236.
615. Sheth KJ, Swick HM, Haworth N. Neurological involvement in the hemo- 640. Callen DF, Baker E, Lane S, et al. Regional mapping of the Batten dis-
lytic uremic syndrome. Ann Neurol 1986;19:90–93. ease locus (CLN3) to human chromosome 16p12. Am J Hum Genet
616. Eriksson KJ, Boyd SG, Tasker RC. Acute neurology and neurophysology of 1991;49:1372–1377.
haemolytic-uraemic syndrome. Arch Dis Child 2001;84:434–435. 641. Kim S-J, Zhang Z, Sarkar C, et al. Palmitoyl protein thioesterase-1 defi-
617. Schmidt S, Gudinchet F, Meagher-Villemure K, et al. Brain involvement ciency impairs synaptic vesicle recycling at nerve terminals, contributing to
in Haemolytic-uraemic syndrome: MRI features of coagulative necrosis. neuropathology in humans and mice. J Clin Invest 2008;118:3075–3086.
Neuroradiology 2001;43:581–585. 642. Hoffmann SL, Peltonen L. The neuronal ceroid lipofuscinoses. In: Scriver
618. Loirat C, Noris M, Fremeaux-Bacchi V. Complement and the atypi- CR, Beaudet AL, Sly WS, et al., eds. The Metabolic and Molecular Basis of
cal hemolytic uremic syndrome in children. Pediatr Nephrol 2008;23: Inherited Disease. New York: McGraw-Hill, 2001:3877–3894.
1957–1972. 643. Kohlschütter A, Schulz A. Towards understanding the neuronal ceroid
619. Sherwood JW, Wagle WA. Hemolytic uremic syndrome: MR findings of lipofuscinoses. Brain Dev 2009;31:499–502.
CNS complications. Am J Neuroradiol 1991;12:703–704. 644. Siintola E, Partanen S, Stromme P, et al. Cathepsin D deficiency under-
620. Steinborn M, Leiz S, Rudisser K, et al. CT and MRI in haemolytic ura- lies congenital human neuronal ceroid-lipofuscinosis. Brain 2006;129:
emic syndrome with central nervous system involvement: distribu- 1438–1445.
tion of lesions and prognostic value of imaging findings. Pediatr Radiol 645. Nardocci N, Verga M, binelli S, et al. Neuronal ceroid lipofuscinosis: a clinical
2004;34:805–810. and morphological study of 19 patients. Am J Med Genet 1995;57:123–141.
621. Hager A, Staudt M, Klare B, et al. Hemolytic-uremic syndrome with 646. Williams R, Aberg L, Autti T, et al. Diagnosis of the neuronal ceroid lipo-
involvement of basal ganglia and cerebellum. Neuropediatrics 1999;30: fuscinoses: an update. Biochim Biophys Acta 2006;1762:865–872.
210–213. 647. Autti T, Raininko R, Vanhanen SL, et al. MRI of neuronal ceroid lipofusci-
622. Schor NF, Sheets R, Szer IS, et al. Neurologic manifestations of rheumatic nosis. I. Cranial MRI of 30 patients with juvenile neuronal ceroid lipofus-
disorders of childhood. In: Swaiman KF, Ashwal S, Ferreiro DM, eds. cinosis. Neuroradiology 1996;38:476–482.
Pediatric Neurology: Principles and Practice. Philadelphia: Mosby Elsevier, 648. Autti T, Raininko R, Launes J, et al. Jansky-Bielschowsky variant disease:
2006:1803–1833. CT, MRI, and SPECT findings. Pediatr Neurol 1992;8:121–126.
623. Gordon N. Sydenham’s chorea, and its complications affecting the ner- 649. Vanhanen S-L, Raininko R, Autti T, et al. MRI evaluation of the brain
vous system. Brain Dev 2009;31:11–14. in infantile neuronal ceroid-lipofuscinoses. Part 2: MRI findings in
624. Buchanan D. Pathological changes in chorea. Am J Dis Child 1941;62: 21 patients. J Child Neurol 1995;10:444–450.
443–445. 650. Vanhanen SL, Puranen J, Autti T, et al. Neuroradiological findings (MRS,
625. Emery E, Vieco P. Sydenham Chorea: magnetic resonance imaging reveals MRI, SPECT) in infantile neuronal ceroid-lipofuscinosis (infantile CLN1)
permanent basal ganglia injury. Neurology 1997;48:531–533. at different stages of the disease. Neuropediatrics 2004;35:27–35.
626. Kienzle G, Breger R, Chun R, et al. Sydenham chorea: MR manifestations 651. Autti T, Raininko R, Santavuori P, et al. MRI of neuronal ceroid lipofus-
in two cases. Am J Neuroradiol 1991;12:73–76. cinosis. II. Postmortem MRI and histopathological study of the brain in
627. Traill Z, Pike M, Byrne J. Sydenham’s chorea: a case showing reversible 16 cases of neuronal ceroid lipofuscinosis of juvenile or late infantile type.
striatal abnormalities on CT and MRI. Dev Med Child Neurol 1995;37: Neuroradiology 1997;39:371–377.
270–273. 652. Järvelä I, Autti T, Lamminranta S, et al. Clinical and magnetic resonance
628. Giedd J, Rapoport J, Kruesi M, et al. Sydenham’s chorea: magnetic reso- imaging findings in Batten disease: analysis of the major mutation (1.02-kb
nance imaging of the basal ganglia. Neurology 1995;45:2199–2202. deletion). Ann Neurol 1997;42:799–802.

Barkovich_Chap03.indd 228 5/6/2011 12:09:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 229

653. Seitz D, Grodd W, Schwab A, et al. MR imaging and localized proton MR 677. Rett A. Über ein zerebral-atrophisches Syndrom bei Hyperammonämie.
spectroscopy in late infantile neuronal ceroid lipofuscinosis. Am J Neuro- Wien: Brüder Hollinek, 1966.
radiol 1998;19:1373–1377. 678. Hagberg BA, Aicardi J, Dias K, et al. A progressive syndrome of autism,
654. Autti T, Raininko R, Vanhanen S-L, et al. Magnetic resonance techniques demantia, ataxia, and loss of purposeful hand use in girls: Rett syndrome.
in neuronal ceroid lipofuscinoses and some other lysosomal diseases Report of 35 cases. Ann Neurol 1983;14:471–479.
affecting the brain. Curr Opinion Neurol 1997;10:519–524. 679. Amir RE, van den Veyver IB, Wan M, et al. Rett syndrome is caused by
655. Brockmann K, Pouwels PJW, Christen H-J, et al. Localized proton magnetic mutations in X-linked MECP2. Nat Genet 1999;23:185–188.
resonance spectroscopy of cerebral metabolic disturbances in children 680. Kerr AM, Nomura Y, Armstrong D, et al. Guidelines for reporting clinical
with neuronal ceroid lipofuscinosis. Neuropediatrics 1996;27:242–248. features in cases with MECP2 mutations. Brain Dev 2001;23:208–211.
656. Iannetti P, Messa C, Spalice A, et al. Positron emission tomography in neu- 681. Clayton-Smith J, Watson P, Ramsden S, et al. Somatic mutation in MECP2
ronal ceroid lipofuscinosis (Jansky-Bielschowsky disease): a case report. as a nonfatal neurodevelopmental disorder in males. Lancet 2000;356:
Brain Dev 1994;16:459–462. 830–831.
657. Vanhanen S-L, Liewendahl K, Raininko R, et al. Brain perfusion SPECT in 682. Moog U, Smeets EEJ, van Roozendaal KEP, et al. Neurodevelopmental
infantile neuronal ceriod-liopfuscinosis (INCL). Comparison with clini- disorders in males related to the gene causing Rett syndrome in females
cal manifestations and MRI findings. Neuropediatrics 1996;27:76–83. (MECP2). Eur J Paediatr Neurol 2003;7:5–12.
658. Launes J, Heiskala H, Nikkinen P, et al. Brain perfusion SPECT in juvenile 683. Kerr A. Early clinical signs in the Rett disorder. Neuropediatrics 1995;16:
neuronal ceriod lipofuscinosis. Neuropediatrics 1996;27:84–87. 67–71.
659. Jenner F, Pollit R. Large quantities of 2-acetamido-1(beta-L-aspartamido)- 684. Witt-Engerstrom I. Age-related occurrence of signs and symptoms in the
1,2-dideoxyglucose in the urine of mentally retarded siblings. Biochem J Rett syndrome. Brain Dev 1992;14:S11-S20.
1967;103:148–150. 685. Armstrong DD. The neuropathology of Rett syndrome—overview 1994.
660. Ikonen E, Peltonen L. Mutations causing aspartylglucosaminuria (AGU): Neuropediatrics 1995;26:100–104.
a slysosomal accumulation disease. Hum Mutat 1992;1:361–365. 686. Watson P, Black G, Ramsden S, et al. Angelman syndrome phenotype asso-
661. Autti T, Raininko R, Haltia M, et al. Aspartylglucosaminuria: radiologic ciated with mutations in MECP2, a gene encoding a methyl CpG binding
course of the disease with histopathologic correlation. J Child Neurol protein. J Med Genet 2001;38:224–228.
1997;12:369–375. 687. Hoffbuhr K, Devaney J, LaFleur B, et al. MeCP2 mutations in children with
662. Arvio MA, Peippo MM, Arvio PJ, et al. Dysmorphic facial features in aspar- and without the phenotype of Rett syndrome. Neurology 2001;56:1486–
tylglucosaminuria patients and carriers. Clin Dysmorphol 2004;13:11–15. 1495.
663. Autti T, Lönnqvist T, Joensuu R. Bilateral pulvinal signal intensity decrease 688. Schwartzman J, Bernadino A, Nishimura A, Gomes R, Zatz M. Rett syn-
on T2 weighted images in patiens with aspartylglucosaminuria. Acta drome in a boy with a 47, XXY karyotype confirmed by a rare mutation in
Radiol 2008;49:687–692. the MECP2 gene. Neuropediatrics 2001;32:162–164.
664. Autti T, Rapola J, Santavuori P, et al. Bone marrow transplantation in 689. Villard L, Kpebe A, Cardoso C, Chelly P, Tardieu P, Fontes M. Two affected
aspartylglucosaminuria—histopathological and MRI study. Neuropediat- boys in a Rett syndrome family: clinical and molecular findings. Neurology
rics 1999;30:283–288. 2000;55:1188–1193.
665. Dalmau J, Tuzun E, Wu HY, et al. Paraneoplastic anti-N-methyl-D- 690. Meloni I, Bruttini M, Longo I, et al. A mutation in the rett syndrome gene,
aspartase receptor encephalitis associated with ovarian teratoma. Ann MECP2, causes X-linked mental retardation and progressively spasticity
Neurol 2007;61:25–36. in males. Am J Hum Genet 2000;67:982–985.
666. Dale RC, Irani SR, Brilot F, et al. N-methyl-D-aspartate receptor antbodies in 691. Geerdink N, Rotteveel J, Lammens M, et al. MECP2 mutation in a boy
pediatric dyskinetic encephalitis lethargica. Ann Neurol 2009;66:704–709. with severe neonatal encephalophathy: clinical, neuropathological and
667. Shimazaki H, Ando Y, Nakano I, et al. Reversible limbic encephalitis with molecular findings. Neuropediatrics 2002;33:33–36.
antibodies against the membranes of neurones of the hippocampus. 692. Subramaniam B, Naidu S, Reiss AL. Neuroanatomy in Rett syndrome:
J Neurol Neurosurg Psychiatry 2007;78:324–325. cerebral cortex and posterior fossa. Neurology 1997;48:399–407.
668. Kurian MA, Morgan NV, MacPherson L, et al. Phenotypic spectrum 693. Hanefeld F, Christen H-J, Holzbach U, et al. Cerebral proton magnetic
of neurodegeneration associated with mutations in the PLA2G6 gene resonance spectroscopy in Rett syndrome. Neuropediatrics 1995;26:
(PLAN). Neurology 2008;70:1623–1629. 126–127.
669. Morgan NV, Westaway SK, Morton JEV, et al. PLA2G6, encoding a phos- 694. Naidu S, Kaufmann W, Abrams M, et al. Neuroimaging studies in Rett
pholipase A2, is mutated in neurodegenerative disorders with high brain syndrome. Brain Dev 2001;23:S62-S71.
iron. Nature Genet 2006;38:752–754. 695. van der Knaap MS, Valk J. Toxic encephalopathy. In: van der Knaap MS,
670. Gregory A, Polster BJ, Hayflick S. Clinical and genetic delineation of neuro- Valk J, eds. Magnetic Resonance of Myelin, Myelination, and Myelin Disor-
degeneration with brain iron accumulation. J Med Genet 2009;46:73–80. ders, 2nd ed. Berlin: Springer, 1995:350–361.
671. Seitelberger F. Neuroaxonal dystrophy: its relation to aging and neurologi- 696. Kinoshita T, Sugihara S, Matsusue E, et al. Pallidoreticular Damage in
cal disease. In: Vinken PJ, Bruin GW, Klawans HL, eds. Handbook of Clini- Acute Carbon Monoxide Poisoning: Diffusion-Weighted MR Imaging
cal Neurology. Amsterdam: Elsevier, 1986:391–495. Findings. Am J Neuroradiol 2005;26(7):1845–1848.
672. Farina L, Nardocci N, Bruzzone MG, et al. Infantile neuroaxonal dystro- 697. O’Donnell P, Buxton P J, Pitkin A, et aL. The magnetic resonance imag-
phy: neuroradiological studies in 11 patients. Neuroradiology 1999;41: ing appearances of the brain in acute carbon monoxide poisoning. Clin
376–380. Radiol 2000;55(4):273.
673. Mader I, Krägeloh-Mann I, Seeger U, et al. Proton MR spectroscopy reveals 698. Rachinger J, Fellner FA, Stieglbauer K, et al. MR changes after acute cya-
lactate in infantile neuroaxonal dystrophy. Neuropediatrics 2001;32:97–100. nide intoxication. Am J Neuroradiol 2002;23(8):1398–1401.
674. Takahashi T, Suchi M, Desnick R, et al. Identification and expression of 699. Lapresle J, Fardeau M. The central nervous system and carbon monoxide
five mutations in the human acid sphingomyelinase gene causing types poisoning. II. Anatonical study of brain lesions following intoxication
A and B Niemann-Pick disease. Molecular evidence for genetic hetero- with carbon monoxide (22 cases). Prog Brain Res 1967;24:31–74.
geneity in the neuronopathic and non-neuronopathic forms. J Biol Chem 700. Ginsberg M, Myers R, McDonahg B. Experimental carbon monoxide
1992;267:12552–12558. encephalopathy in the primate. II. Clinical aspects, neuropathyology, and
675. Greer WL, Riddell DC, Murty S, et al. Linkage disequilibrium mapping physiologic correlation. Arch Neurol 1974;30:209–216.
of the Nova Scotia variant of Niemann-Pick disease. Clin Genet 1999;55: 701. Kumar S, Mattan NS, de Vellis J. Canavan disease: a white matter disorder.
248–255. Ment Retard Dev Disabil Res Rev 2006;12:157–165.
676. Pastores GM, Kolodny EH. Lysosome storage diseases. In: Swaiman KF, 702. Matalon R, Michals K, Sebesta D, et al. Aspartoacylase deficiency and
Ashwal S, Ferriero DM, eds. Pediatric Neurology: Principles and Practice, N-acetylaspartic aciduria in patients with Canavan disease. Am J Med
4th ed. Philadelphia: Mosby Elsevier; 2006:659–714. Genet 1988;29:463–471.

Barkovich_Chap03.indd 229 5/6/2011 12:09:36 AM


230 Pediatric Neuroimaging

703. Matalon R, Michals-Matalon K. Recent advances in Canavan disease. Adv 729. Quinlan R, Brenner M, Goldman J, et al. GFAP and its role in Alexander
Pediatr 1999;46:493–506. disease. Exp Cell Res 2007;313:2077–2087.
704. Baslow M, Suckow R, Sapirstein V, et al. Expression of aspartoacylase 730. van der Voorn JP, Pouwels PJW, Salomons GS, et al. Unraveling pathology
activity in cultured rat macroglial cells is limited to oligodendrocytes. in juvenile Alexander disease: serial quantitative MR imaging and spec-
J Mol Neurosci 1999;13:47–53. troscopy of white matter. Neuroradiology 2009;51:669–675.
705. Traka M, Wollman RL, Cerda SR, et al. Nur7 is a nonsense mutation in the 731. Kyllerman M, Rosengren L, Wiklund LM, et al. Increased levels of GFAP
mouse aspartoacylase gene that causes spongy degeneration of the CNS. in the cerebrospinal fluid in three subtypes of genetically confirmed
J Neurosci 2008;28:11537–11549. Alexander disease. Neuropediatrics 2005;36:319–323.
706. Baslow M, Guilfoyle D. Are astrocytes the missing link between lack of 732. Farrell K, Chuang S, Becker LE. Computed tomography in Alexander’s
brain aspartoacylase activity and the spongiform leukodystrophy in Cana- disease. Ann Neurol 1984;15:605–609.
van disease? Neurochem Res 2009;34(9):1523–1534. 733. Trommer BL, Naidich TP, Del Cento MC, et al. Noninvasive CT diagnosis
707. Volpe JJ. Neurology of the Newborn. Philadelphia: Saunders, 1987. of infantile Alexander disease: pathologic correlation. J Comput Assist
708. Gordon N. Canavan disease: a review of recent developments. Eur J Pae- Tomogr 1983;7:509–512.
diatr Neurol 2001;5:65–69. 734. Springer S, Erlewein R, Naegele T, et al. Alexander disease—classification
709. Yalçinkaya C, Benbir G, Salomons GS, et al. Atypical MRI findings in revisited and isolation of a neonatal form. Neuropediatrics 2000;31:
Canavan disease: a patient with a mild course. Neuropediatrics 2005;36: 86–92.
336–339. 735. Farina L, Pareyson D, Minati L, et al. Can MR imaging diagnose adult-
710. Janson C, Kolodny E, Zeng B, et al. Mild-onset presentation of Canavan’s onset Alexander disease? Am J Neuroradiol 2008;29:1190–1196.
disease associated with novel G212A point mutation in aspartoacylase 736. McKusick VA, Neufeld EF. The mucopolysaccharide storage diseases. In:
gene. Ann Neurol 2006;59:428–431. Stanbury JB, ed. The Metabolic Basis of Inherited Disease. New York, NY:
711. Tacke U, Olbrich H, Sass JO, et al. Possible genotype-phenotype correla- McGraw-Hill, 1983:751–823.
tions in children with mild clinical course of Canavan disease. Neuropedi- 737. Butler IJ. Disorders of connective tissue and bone. In: Berg BO, ed. Neu-
atrics 2005;36:252–255. rologic Aspects of Pediatrics. Boston: Butterworth-Heinemann, 1992:468–
712. Breitbach-Faller N, Schrader K, Rating D, et al. Ultrasound findings in 483.
follow-up investigations in a case of aspartoacylase deficiency (Canavan 738. Vougioukas V, Berlis A, Kopp M, et al. Neurosurgical interventions in
disease). Neuropediatrics 2003;34(2):96–99. children with Maroteaux-Lamy syndrome. Case report and review of the
713. Brismar J, Brismar G, Gascon G, et al. Canavan disease: CT and MR imag- literature. Pediatr Neurosurg 2001;35:35–38.
ing of the brain. Am J Neuroradiol 1990;11:805–810. 739. Dickerman RD, Colle KO, Bruno Jr CA, et al. Craniovertebral instability
714. Topçu M, Erdem G, Saatci I, et al. Clinical and magnetic resonance imag- with spinal cord compression in a 17-month-old boy with Sly syndrome
ing features of L-2-hydroxyglutaric acidemia: report of three cases in com- (mucopolysaccharidosis type VII): a surgical dilemma. Spine 2004;29:
parison with Canavan disease. J Child Neurol 1996;11:373–377. E92–E94.
715. Sener RN. Canavan disease: diffusion magnetic resonance imaging find- 740. Mut M, Cila A, Varli K, Akalan N. Multilevel myelopathy in Maroteaux-
ings. J Comput Assist Tomogr 2003;27:30–33. Lamy syndrome and review of the literature. Clin Neurol Neurosurg
716. Grodd W, Krageloh-Mann I, Petersen D, et al. In vivo assessment of 2005;107:230–235.
N-acetylaspartate in brain in spongy degeneration (Canavan;s disease) by 741. Finn CT, Vedolin L, Schwartz IV, et al. Magnetic resonance imaging find-
proton spectroscopy (Letter). Lancet 1990;2:437–438. ings in Hunter syndrome. Acta Paediatr 2008;97(Suppl 457):61–68.
717. Velinov M, Zellers N, Styles J, et al. Homozygosity for mutation G212A of 742. Lee C, Dineen TE, Brack M, et al. Mucopolysaccharidoses: characteriza-
the gene for aspartoacylase is associated with atypical form of Canavan’s tion by cranial MR imaging. Am J Neuroradiol 1993;14:1285–1292.
disease (Letter). Clin Genet 2008;73:288–289. 743. Murata R, Nakajima S, Tanaka A, et al. MR imaging of the brain in patients
718. Bluml S, Moreno A, Hwang JH, et al. 1-(13)C glucose magnetic reso- with mucopolysaccharidosis. Am J Neuroradiol 1989;10:1165–1170.
nance spectroscopy of pediatric and adult brain disorders. NMR Biomed 744. Watts RWE, Spellacy E, Kendall BE, et al. CT studies on patients with
2001;14(1):19–32. mucopolysaccharidoses. Neuroradiology 1981;21:9–23.
719. Alexander WS. Progressive fibrinoid degeneration of fibrillary astro- 745. Barone R, Parano E, Trifiletti R, et al. White matter changes mimicking a
cytes associated with mental retardation in a hydrocephalic infant. Brain leukodystrophy in a patient with Mucopolysaccharidosis: characterization
1949;72:373–381. by MRI. J Neurol Sci 2002;195:171–175.
720. Pridmore CL, Baraitser M, Harding B, et al. Alexander’s disease: clues to 746. Barone R, Nigro F, Triulzi F, et al. Clinical and neuroradiological follow-up
diagnosis. J Child Neurol 1993;8:134–144. in mucopolysaccaridosis type III (Sanfilipo syndrome). Neuropediatrics
721. Isaacs A, Baker M, Hutton M. Determination of the gene structure of 1999;30:270–274.
human GFAP and absence of coding region mutations associated with 747. Hughes DG, Chadderton RD, Cowie RA, et al. MRI of the brain and
frontotemporal dementia with Parkinsonism linked to chromosome 17. craniocervical junction in Morquio’s disease. Neuroradiology 1997;39:
Genomics 1998;51:152–154. 381–385.
722. Sawaichi Y. Review of Alexander disease: beyond the classical concept of 748. Hurko O, Uemotsu S. Neurosurgical consideration in skeletal dysplasia.
leukodystrophy. Brain Dev 2009;31:493–498. Neurosurg Quart 1993;3:192–217.
723. van der Knaap MS, Ramesh V, Schiffmann R, et al. Alexander disease: ven- 749. Petitti N, Holder CA, Williams III DW. Mucopolysaccharidosis III (San-
tricular garlands and abnormalities of the medulla and spinal cord. Neu- filippo syndrome) type B: cranial imaging in two cases. J Comput Asst
rology 2006;66:494–498. Tomogr 1997;21:897–899.
724. Niinikoski H, Haataja L, Brander A, et al. Alexander disease as a cause of 750. Kulkarni MV, Williams JC, Yeakley JW. MR in the diagnosis of the cranio-
nocturnal vomiting in a 7-year-old girl. Pediatr Radiol 2009;39:872–875. cervical manifestations of the mucopolysaccharidoses. Magn Res Imaging
725. van der Knaap MS, Naidu S, Breiter SN, et al. Alexander disease: diagnosis 1987;5:317–323.
with MR imaging. Am J Neuroradiol 2001;22:541–552. 751. Tzika AA, Ball WS Jr, Vigneron D, et al. Clinical proton MR spectroscopy of
726. van der Knaap MS, Salomons GS, Li R, et al. Unusual variants of Alexan- neurodegenerative disease in childhood. Am J Neuroradiol 1993;14:1267–
der’s disease. Ann Neurol Mar 2005;57(3):327–338. 1281.
727. Li R, Johnson AB, Salomons G, et al. Glial fibrillary acidic protein muta- 752. Takahashi Y, Sukegawa K, Aoki M, et al. Evaluation of accumulated muco-
tions in infantile, juvenile, and adult forms of Alexander disease. Ann Neu- polysaccharides in the brain of patients with mucopolysaccharidoses
rol 2005;57(3):310–326. by 1H-magnetic resonance spectroscopy before and after bone marrow
728. Pareyson D, Fancellu R, Mariotti C, et al. Adult-onset Alexander disease: transplantation. Pediatr Res 2001;49(3):349–355.
a series of eleven unrelated cases with review of the literature. Brain 753. Busche A, Hennermann JB, Bürger F, et al. Neonatal manifestation of mul-
2008;131:2321–2331. tiple sulfatase deficiency. Eur J Pediatr 2009;168:969–973.

Barkovich_Chap03.indd 230 5/6/2011 12:09:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 231

754. Zafeiriou D, Vargiami E, Papadopoulou K, et al. Serial magnetic resonance 782. Poll-The BT, Roels F, Ogier H, et al. A new peroxisomal disorder with
imaging and neurophysiological studies in multiple sulphatase deficiency. enlarged peroxisomes and a specific deficiency of acyl-CoA oxidase
Eur J Paediatr Neurol 2008;12:190–194. (pseudo-neonatal adrenoleukodystrophy). Am J Hum Genet 1988;41:
755. Mancini G, van Diggelen O, Huijmans J, Stroink H, de Coo R. Pitfalls in the 422–434.
diagnosis of multiple sulfatase deficiency. Neuropediatrics 2001;32:38–40. 783. Fournier B, Saudubray J-M, Benichou B, et al. Large deletion of the perox-
756. Raymond GV, Naidu S, Moser HW. Peroxisomal disorders. In: Swaiman isomal acyl-CoA oxidase gene in pseudoneonatal adrenoleukodystrophy.
KF, Ashwal S, Ferriero DM, eds. Pediatric Neurology: Principles and Prac- J Clin Invest 1994;94:526–531.
tive, 4th ed. Philadelphia: Mosby Elsevier; 2006:735–758. 784. Carrozzo R, Bellini C, Lucioli S, et al. Peroxisomal acyl-CoA-oxidase defi-
757. Krisans SK. The role of peroxisomes in cholsterol metabolism. Am J Resp ciency: two new cases. Am J Med Genet 2008;146A:1676–1681.
Cell Mol Biol 1992;7:358–364. 785. Ferdinandusse S, Denis S, Hogenhout EM, et al. Clinical, biochemical, and
758. Vanden Bosch H, Schutgens RBH, Wanders RJA, et al. Biochemistry of mutational spectrum of peroxisomal acyl-coenzyme A oxidase deficiency.
peroxisomes. Ann Rev Biochem 1992;61:157–197. Hum Mutat 2007;28:904–912.
759. Moser H. Genotype-phenotype correlations in peroxisomal disorders. 786. Powers J, Moser H. Peroxisomal disorders: genotype, phenotype, major
Dev Brain Dysfunction 1997;10:282–292. neuropathologic lesions, and pathogenesis. Brain Pathol 1998;8:101–120.
760. Wanders R, Vreken P, Ferdinandusse S, et al. Peroxisomal fatty acid 787. Braverman N, Steel G, Obie C, et al. Human PEX7 encodes the peroxi-
alpha- and beta- oxidation in humans: enzymology, peroxisomal metabo- somal PTS2 receptor and is responsible for rhizomelic chondrodysplasia
lite transporters and peroxisomal diseases. Biochem Soc Trans 2001;29: punctata. Nature Genet 1997;15:369–376.
250–267. 788. Motley AM, Hettema WH, Hogenhout EM, et al. Rhizomelic chondrodys-
761. Baumgartner M, Saudubray J. Peroxisomal disorders. Semin Neonatol plasia punctata is a peroxisomal protein targeting disease caused by a non-
2002;7:85–94. functional PTS2 receptor. Nature Genet 1997;15:377–380.
762. Rizzo W. Peroxisome 1, 2, 3… Ann Neurol 2000;47:281–283. 789. Purdue PE, Zhang JW, Skoneczny M, et al. Rhizomelic chondrodysplasia
763. Subramani S. Components involved in peroxisome import, biogenesis, punctata is caused by deficiency of human PEX7, a homologue of the yeast
proliferation, turnover, and movement. Physiol Rev 1998;78:171–188. PTS2 receptor. Nature Genet 1997;15:381–384.
764. Kaufmann WE, Theda C, Naidu S, et al. Neuronal migration abnormality 790. Viola A, Confort-Gouny S, Ranjeva J, et al. MR imaging and MR spec-
in peroxisomal bifunctional enzyme defect. Ann Neurol 1996;39:268–271. troscopy in rhizomelic chondrodysplasia punctata. Am J Neuroradiol
765. Naritomi K, Izumikawa Y, Ohshiro S, et al. Gene assignment of Zellweger 2002;23:480–483.
syndrome to 7q11.23: report of the second case associated with a pericen- 791. Clayton PT, Eckhardt S, Wilson J, et al. Isolated dihydroxyacetonephos-
tric inversion of chromosome 7. Hum Genet 1989;84:79–80. phate acyltransferase deficiency presenting with developmental delay.
766. Barkovich AJ, Peck WW. MR of Zellweger syndrome. Am J Neuroradiol J Inherit Metab Dis 1994;17:533–540.
1997;18:1063–1070. 792. Sztriha LS, Nork MP, Abdulrazzaq YM, et al. Abnormal myelination in
767. Volpe JJ, Adams RD. Cerebro-hepato-renal syndrome of Zellweger. An peroxisomal isloated dihydroxyacetonephosphate acyltransferase defi-
inherited disorder of neuronal migration. Acta Neuropathol 1972;20: ciency. Pediatr Neurol 1997;16:232–236.
175–198. 793. Gu M, Cooper J, Butler P, et al. Oxidative-phosphorylation defects in liver
768. Norman MG, McGillivray BC, Kalousek DK, et al. Congenital Malforma- of patients with Wilson’s disease. Lancet 2000;356:469–474.
tions of the Brain: Pathologic, Embryologic, Clinical, Radiologic and Genetic 794. Bowcock A, Farrer L, Hebert J, et al. Eight closely linked loci place the
Aspects. Oxford: Osford University Press; 1995. Wilson disease locus within 13q14-q21. Am J Hum Genet 1988;43:
769. Evrard P, Caviness VSJ, Prats-Vinas J, et al. The mechanism of arrest of 664–674.
neuronal migration in the Zellweger malformation: an hypothesis based 795. Kim TJ, Kim IO, Kim WS, et al. MR imaging of the brain in Wilson disease
upon cytoarchitectonic analysis. Acta Neuropathol 1978;41:109–117. of childhood: findings before and after treatment with clinical correlation.
770. van Straaten HL, van Tintelen JP, Trijbels JM, et al. Neonatal lactic acidosis, Am J Neuroradiol 2006;27:1373–1378.
complex I/IV deficiency, and fetal cerebral disruption. Neuropediatrics 796. Kvicala V, Vymazal J, Nevsimalova S. CT of Wilson’s disease. Am J Neuro-
2005;36:193–199. radiol 1983;4:429–430.
771. Groenendaal F, Bianchi MC, Battini R, et al. Proton magnetic resonance 797. van Wassenaer-van Hall H, van den Heuvel A, Algra A, et al. Wilson dis-
spectroscopy of the cerebrum in two young infants with Zellweger syn- ease: findings at MR imaging and CT of the brain with clinical correlation.
drome. Neuropediatrics 2001;32:23–27. Radiology 1996;198:531–536.
772. Ulrich J, Herschkowitz N, Heitz P, et al. Adrenoleukodystrophy. Preliminary 798. van Wassenaer-van Hall HN. Neuroimaging in Wilson disease. Metab
report of a connatal case. Light- and electron microscopical, immunohis- Brain Dis 1997;12:1–19.
tochemical and biochemical findings. Acta Neuropathol 1978;43:77–83. 799. Mochizuki H, Kamakura K, Masaki T, et al. Atypical MRI features of
773. Powers JM. The pathology of peroxisomal disorders with pathogenetic Wilson’s disease: high signal in globus pallidus in T1 weighted images.
considerations. J Neuropath Exp Neurol 1995;54:710–719. Neuroradiology 1997;39:171–174.
774. Shimozawa N, Imamura A, Zhang Z, et al. Defective PEX gene products 800. Kim I, Yeon K, Kim W, et al. MR imaging of the brain in Wilson disease of
correlate with the protein import, biochemical abnormalities, and phe- childhood. Radiology 1993;189(P):195.
notypic heterogeneity in peroxisome biogenesis disorders. J Med Genet 801. Sinha S, Taly AB, Ravishankar S, et al. Wilson’s disease: cranial MRI obser-
1999;36:779–781. vations and clinical correlation. Neuroradiology 2006;48:613–621.
775. Wanders R, Schutgens R, Barth P. Peroxisomal disorders: a review. J Neu- 802. Kitzberger R, Madl C, Ferenci P. Wilson disease. Metab Brain Dis 2005;
ropath Exp Neurol 1995;54:726–739. 20:295–302.
776. Percy A, Rutledge S. Adrenoleukodystrophy and related disorders. Ment 803. Aisen AM, Martel W, Gabrielsen TO, et al. CT of Wilson’s disease. Am J
Retard Dev Disabil Res Rev 2001;7:179–189. Neuroradiol 1985;4:429–430.
777. Choksi V, Hoeffner E, Karaarslan E, et al. Infantile refsum disease: case 804. Starosta-Rubenstein S, Young AB, Kluin K, et al. Clinical assessment of
report. Am J Neuroradiol 2003;24:2082–2084. 31 patients with Wilson’s disease: correlations with structural changes on
778. Cakirer S, Savas MR. Infantile Refsum disease: serial evaluation with MRI. magnetic resonance imaging. Arch Neurol 1987;44:365–370.
Pediatr Radiol 2005;35:212–215. 805. Prayer L, Wimberger D, Kramer J, et al. Cranial MRI in Wilson’s disease.
779. Wanders RJ, Waterham HR. Biochemistry of mammalian peroxisomes Neuroradiology 1990;32:211–212.
revisited. Annu Rev Biochem 2006;75:295–332. 806. Jayasundar R, Sahani A, Gaikwad S, et al. Proton MR spectroscopy of basal
780. Watkins PA, Chen WW, Harris CJ, et al. Peroxisomal bifunctional enzyme ganglia in Wilson’s disease: case report and review of literature. Magn
deficiency. J Clin Invest 1989;83:771–777. Reson Imaging 2002;20:131–135.
781. Goldfischer S, Collins J, Rapin I, et al. Pseudo-Zellweger syndrome: deficien- 807. Kraft E, Trenkwalder C, Then Bergh F, et al. Magnetic resonance proton
cies in several peroxisomal oxidative activities. J Pediatr 1986;108:25–32. spectroscopy of the brain in Wilson’s disease. J Neurol 1999;246:693–699.

Barkovich_Chap03.indd 231 5/6/2011 12:09:36 AM


232 Pediatric Neuroimaging

808. Alanen A, Komu M, Pettinen M, et al. Magnetic resonance imaging and pro- 835. Inao S, Marmarou A, Clarke G, et al. Production and clearance of lactate
ton MR spectroscophy in Wilson’s disease. Br J Radiol 1999;72:749–756. from brain tissue, cerebrospinal fluid, and serum following experimental
809. Lucato LT, Otaduy MCG, Barbosa ER, et al. Proton MR spectroscopy in brain injury. J Neurosurg 1988;69:736–744.
Wilson disease: analysis of 36 cases. Am J Neuroradiol 2005;26:1066–1071. 836. Pavlakis SG, Phillips PC, DiMauro S, et al. Mitochondrial myopathy,
810. DiMauro S, Bonilla E, Zeviani M, et al. Mitochondrial Myopathies. Ann encephalopathy, lactic acidosis, and stroke-like episodes: a distinctive
Neurol 1985;17:521–538. clinical syndrome. Ann Neurol 1984;16:481–488.
811. DiMauro S, Bonilla E, Lombes A, et al. Mitochondrial encephalomyopa- 837. Rowland LP, Blake DM, Hirano M, et al. Clinical syndromes associated
thies. Neurol Clin 1990;8:483–506. with ragged red fibers. Rev Neurol 1991;147:467–473.
812. Petty RKH, Harding AE, Morgan-Hughes JA. The clinical features of 838. Haas RH, Nyhan WL. Disorders of organic acids. In: Berg BO, ed. Neuro-
mitochondrial myopathy. Brain 1986;109:915–923. logic Aspects of Pediatrics. Boston: Butterworth-Heinemann; 1992:47–91.
813. Holt IJ, Harding AE, Cooper JM, et al. Mitochondrial myopathies: clinical 839. DiMauro S, Moraes CT, Shanske S, et al. Mitochondrial encephalomyopa-
and biochemical features of 30 patients with major deletions of muscle thies: biochemical approach. Rev Neurol 1991;147:443–449.
mitochondrial DNA. Ann Neurol 1989;26(6):699–708. 840. Allard JC, Tilak S, Carter AP. CT and MR of MELAS syndrome. Am J Neu-
814. Wallace DC. Mitochondrial genetics: a paradigm for aging and degenera- roradiol 1988;9:1234–1238.
tive diseases? Science 1992;256:628–632. 841. van Hellenberg Hubar JLM, Gabreels FJM, Rultenbeek W, et al. MELAS
815. Munnich A, Rustin P. Clinical spectrum and diagnosis of mitochondrial syndrome: report of two patients, and comparison with data of 24 patients
disorders. Am J Med Genet 2001;106:4–17. derived from the literature. Neuropediatrics 1991;22:10–14.
816. Moslemi AR, Darin N. Molecular genetic and clinical aspects of mito- 842. Sproule DM, Kaufmann P. Mitochondrial encephalopathy, lactic acidosis,
chondrial disorders in children. Mitochondrion 2007;7:241–252. and strokelike episodes: basic concepts, clinical phenotype, and thera-
817. Skladal D, Halliday J, Thorburn D. Minimum birth prevalence of mito- peutic management of MELAS syndrome. Ann NY Acad Sci 2008;1142:
chondrial respiratory chanin disorders in children. Brain 2003;126:1905– 133–158.
1912. 843. Rosen L, Phillips S, Enzmann D. Magnetic resonance imaging in MELAS
818. Gire c, Girard N, Nicaise C, et al. Clinical features and neuroradiologi- syndrome. Neuroradiology 1990;32:168–171.
cal findings of mitochondrial pathology in six neonates. Childs Nerv Syst 844. Tam E, A F, Addis J, et al. A novel mitochondrial DNA mutation in COX1
2002;18:621–628. leads to strokes, seizures, and lactic acidosis. Neuropediatrics 2008;39:
819. De Vivo DC, DiMauro S. Mitochondrial diseases. In: Swaiman KF, Ash- 328–334.
wal S, Ferriero DM, eds. Pediatric Neurology: Prinicples & Practice, 4th ed. 845. Barkovich AJ, Good W, Koch TK, et al. Mitochondrial disorders: analysis of
Philadelphia: Mosby Elsevier, 2006:715–734. their clinical and imaging characteristics. Am J Neuroradiol 1993;14:1119–
820. Visapää I, Fellman V, Vesa J, et al. GRACILE syndrome, a lethal metabolic 1137.
disorder with iron overload, is caused by a point mutation in BCS1L. Am 846. Koo B, Becker LE, Chuang SH, et al. Mitochondrial encephalopathy, lactic
J Hum Genet 2002;71(4):863–876. acidosis, stroke-like episodes (MELAS): clinical radiologic, pathological,
821. DiMauro S, Bonilla E. Mitochondrial encephalomyopathies. In: Engel and genetic observations. Ann Neurol 1993;34:25–32.
AG, Franzini-Armstrong E, eds. Myology, Vol. 2. New York: McGraw-Hill, 847. Ducreux D, Nasser G, Lacroix C, et al. MR diffusion tensor imaging, fiber
2004:1623–1662. tracking, and single-voxel spectroscopy findings in an unusual MELAS
822. Wolf N, Smeitink J. Mitochondrial disorders: a proposal for consensus case. Am J Neuroradiol 2005;26(7):1840–1844.
diagnostic criteria in infants and children. Neurology 2002;59:151–159. 848. Sue C, Crimmins D, Soo Y, et al. Neuroradiological features of six kindreds
823. Bernier F, Boneh A, Dennett X, et al. Diagnostic criteria for respiratory with MELAS tRNA(Leu) A2343G point mutation: implications for patho-
chain disorders in adults and children. Neurology 2002;59:1406–1411. genesis. J Neurol Neurosurg Psychiatry 1998;65:233–240.
824. Tulinius MH, Holme E, Kristiansson B, et al. Mitochondrial encephalomy- 849. Abe K, Invi T, Hirono N, et al. Fluctuating MR images with mitochon-
opathies in childhood. II. Clinical manifestations and syndromes. J Pediatr drial encephalomyopathy, lactic acidosis, stroke-like syndrome (MELAS).
1991;119(2):251–259. Neuroradiol 1990;32:77–80.
825. Berenberg RA, Pellock JM, DiMauro S, et al. Lumping or splitting? “Oph- 850. Iizuka T, Sakai F, Suzuki N, et al. Neuronal hyperexcitability in stroke-like
thalmoplegia-plus” or Kearns-Sayre syndrome? Ann Neurol 1977;1:37–54. episodes of MELAS syndrome. Neurology 2002;59:816–824.
826. Morgan-Hughes JA. The mitochondrial myopathies. In: Engel AG, Banker 851. Detre JA, Wang Z, Bogdan AR, et al. Regional variation in brain lactate in
BQ, eds. Myology, Vol. 2. New York: McGraw-Hill, 1986:1709–1743. Leigh syndrome by localized 1-H magnetic resonance spectroscopy. Ann
827. Truong DD, Harding AE, Scaravilli F, et al. Movement disorders in mito- Neurol 1991;29:218–221.
chondrial myopathies. A study of nine cases with two autopsy studies. Mov 852. Grodd W, Krageloh-Mann I, Klose U, et al. Metabolic and destructive
Disord 1990;5:109–117. brain disorders in children: findings with localized proton MR spectros-
828. Darin N, Oldfors A, Moslemi A-R, et al. The incidence of mitochondrial copy. Radiology 1991;181:173–181.
encephalomyopathies in childhood: clinical features and morphological, 853. Dujin JH, Matson GB, Maudsley AA, et al. Human brain infarction: pro-
biochemical, and DNA abnormalities. Ann Neurol 2001;49:377–383. ton MR spectroscopy. Radiology 1992;183:711–718.
829. Ventura F, Ruiter J, Ijlst L, et al. Inhibition of oxidative phosphorylation 854. Wilchowski E, Pouwels P, Frahm J, et al. Quantitative proton magnetic
by palmitoyl-CoA in digitonin permeabilized fibroblasts: implications resonance spectroscopy of cerebral metabolic disturbances in patients
for long-chain fatty acid beta-oxidation disorders. Biochim Biophys Acta with MELAS. Neuropediatrics 1999;30:256–263.
1995;1272:14–20. 855. Yonemura K, Hasegawa Y, Kimura K, et al. Diffusion weighted MR imag-
830. Rupar C, Gillett J, Gordon B, et al. Isolated sulfite oxidase deficiency. ing in a case of mitochondrial myopathy, encephalopathy, lactic acidosis,
Neuropediatrics 1996;27:299–304. and strokelike episodes. Am J Neuroradiol 2001;22:269–272.
831. Valanne L, Ketonen L, Majander A, et al. Neuroradiologic findings in chil- 856. Majoie CB, Akkerman EM, Blank C, et al. Mitochondrial encephalo-
dren with mitochondrial disorders. Am J Neuroradiol 1998;19:369–377. myopathy: comparison of conventional MR imaging with diffusion-
832. Moroni I, Bugiani M, Bizzi A, et al. Cerebral white matter involve- weighted and diffusion tensor imaging: case report. Am J Neuroradiol
ment in children with mitochondrial encephalopathies. Neuropediatrics 2002;23(5):813–816.
2002;33:79–85. 857. Oppenheim C, Galanaud D, Samson Y, et al. Can diffusion weighted mag-
833. Scaglia F, Wong L-J, Vladutiu GD, et al. Predominant cerebellar volume netic resonance imaging help differentiate stroke from stroke-like events
loss as a neuroradiologic feature of pediatric respiratory chain defects. Am in MELAS? J Neurol Neurosurg Psychiatry. 2000;69(2):248–250.
J Neuroradiol 2005;26:1675–1680. 858. Takahashi S, Tohgi H, Yonezawa H, et al. Cerebral blood flow and oxygen
834. Lin D, Crawford T, Barker P. Proton MR spectroscopy in the diagnostic metabolism before and after a stroke-like episode in patients with mito-
evaluation of suspected mitochondrial disease. Am J Neuroradiol chondrial myopathy and encephalopathy, lactic acidosis and stroke-like
2003;24:33–41. episodes (MELAS). J Neurol Sci 1998;158:58–64.

Barkovich_Chap03.indd 232 5/6/2011 12:09:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 233

859. Gropen T, Prohovnik I, Tatemichi T, et al. Cerebral hyperemia in MELAS. 886. Schon EA, DiMauro S. Mitochondrial mutations: genotype to phenotype.
Stroke 1994;25:1873–1876. Novartis Found Symp 2007;287:214–225.
860. Nariai T, Ohno K, Akimoto H, et al. Cerebral blood flow, vascular response 887. Dahl H. Pyruvate dehydrogenase E1 alpha deficiency: males and females
and metabolism in patients with MELAS syndrome—xenon CT and PET differ yet again. Am J Hum Genet 1995;56:553–557.
study. Keio J Med 2000;49(Suppl 1):A68–A70. 888. DeVivo DC, Haymond MW, Obert KA, et al. Defective activation of the
861. Chen R, Huang C, Lee C, et al. Overlapping syndrome of MERRF and pyruvate dehydrogenase complex in subacute necrotizing encephalomyel-
MELAS: molecular and neuroradiological studies. Acta Neurol Scand opathy. Ann Neurol 1979;6:483–494.
1993;87:494–498. 889. Otero L, Brown G, Silver K, et al. Association of cerebral dysgenesis and
862. Kearns T, Sayre GP. Retinitis pigmentosa, external ophthalmoplegia, and lactic acidemia with X-linked PDH E1 alpha subunit mutations in females.
complete heart block. Arch Ophthalmol 1958;60:280–289. Pediatr Neurol 1995;13:327–332.
863. Guggenheim MA, Becker LE, Jagadha V. CPC: muscle weakness in an ado- 890. Nissenkorn A, Michelson M, Ben-Zeev B, et al. Inborn errors of metabolism:
lescent male. Pediatr Neurosci 1985–86;12:320–325. a cause of abnormal brain development. Neurology 2001;56:1265–1272.
864. DiMauro S, Lombes A, Nakase H, et al. Cytochrome c oxidase deficiency. 891. Darin N, Moslemi AR, Lebon S, et al. Genotypes and clinical pheno-
Ped Res 1990;28:536–541. types in children with cytochrome-c oxidase deficiency. Neuropediatrics
865. Maceluch JA, Niedziela M. The clinical diagnosis and molecular genetics 2003;34(6):311–317.
of Kearns-Sayre syndrome: a complex mitochondrial encephalomyopathy. 892. van Riesen AKJ, Antonicka H, Ohlenbusch A, et al. Maternal segmental
Pediatr Endocrinol Rev 2006;4:117–137. disomy in Leigh syndrome with cytochrome c oxidase deficiency caused
866. Bastiaensen LAK, Joosten EMG, De Rooij JAM, et al. Ophthalmoplegia by homozygous SURF1 mutation. Neuropediatrics 2006;37:88–94.
plus, a real nosological entity. Acta Neurol Scand 1978;58:9–34. 893. Tiranti V, Jaksch M, Hofmann S, et al. Loss of function mutations of
867. Leutner C, Layer G, Zierz S, et al. Cerebral MR in ophthalmoplegia plus. SURF-1 are specifically associated with Leigh syndrome with cytochrome
Am J Neuroradiol 1994;15:681–687. c oxidase deficiency. Ann Neurol 1999;46:161–166.
868. Seigel RS, Seeger JF, Gabrielsen TO, et al. Computed tomography 894. Tiranti V, Hoertnagel K, Carrozzo R, et al. Mutations of SURF-1 in Leigh
in oculocraniosomatic disease (Kearns-Sayre syndrome). Radiology disease associated with cytochrome c oxidase deficiency. Am J Hum Genet
1979;130:159–164. 1998;63:1609–1621.
869. Pellock JM, Behrens M, Lewis L, et al. Kearns-Sayre syndrome and 895. Zhu Z, Yao J, Johns T, et al. SURF1, encoding a factor involved in the bio-
hypoparathyroidism. Ann Neurol 1978;3:455–458. genesis of cytochrome c oxidase, is mutated in Leigh syndrome. Nat Genet
870. Demange P, Gia HP, Kalifa G, et al. MR of Kearns-Sayre Syndrome. Am J 1998;20:337–343.
Neuroradiol 1989;10:S91. 896. Rossi A, Biancheri R, Bruno C, et al. Leigh syndrome with COX deficiency
871. Saneto RP, Friedman SD, Shaw DW. Neuroimaging of mitochondrial dis- and SURF1 gene mutations: MR imaging findings. Am J Neuroradiol
ease. Mitochondrion 2008;8:396–413. 2003;24:1188–1191.
872. Sakai Y, Kira R, Torisu H, et al. Persistent diffusion abnormalities in the 897. Farina L, Chiapparini L, Uziel G, et al. MR findings in Leigh syndrome with
brain stem of three children with mitochondrial diseases. Am J Neurora- COX deficiency and SURF-1 mutations. Am J Neuroradiol 2002;23:1095–
diol 2006;27:1924–1926. 1100.
873. Duning T, Deppe M, Keller S, et al. Diffusion tensor imaging in a case 898. Salviati L, Freehauf C, Sacconi S, et al. Novel SURF1 mutation in a child
of Kearns-Sayre syndrome: striking brainstem involvement as a possible with subacute encephalopathy and without the radiological features of
cause of oculomotor symptoms. J Neurol Sci 2009;281:110–112. Leigh syndrome. Am J Med Genet 2004;128A:195–198.
874. Hirano M, Silvestri G, Blake DM, et al. Mitochondrial neurogastrointes- 899. Savoiardo M, Zeviani M, Uziel G, et al. MRI in Leigh Syndrome with
tinal encephalomyopathy (MNGIE): clinical, biochemical, and genetic SURF1 gene mutation. Ann Neurol 2001;51:138–139.
features of an autosomal recessive mitochondrial disorder. Neurology 900. Xie S, Xiao JX, Qi ZY, et al. Heterogeneity of magnetic resonance imaging
1994;44:721–727. in Leigh syndrome with SURF1 gene 604G—>C mutation. Clin Imaging
875. Hirano M, Nishigaki Y, Marti R. Mitochondrial neurogastrointestinal 2009/2// 2009;33(1):1–6.
encephalomyopathy (MNGIE): a disease of two genomes. The Neurologist 901. Santorelli FM, Shanske S, Macaya A, et al. The mutation at nt 8993 of
2004;10(1):8–17. mitochondrial DNA is a common cause of Leigh’s syndrome. Ann Neurol
876. Hirano M, Garcia-de-Yebenes J, Jones AC, et al. Mitochondrial neuro- 1993;34:827–834.
gastrointestinal encephalomyopathy syndrome maps to chromosome 902. Shoffner JM, Fernhoff PM, Krawiecki NS, et al. Subacute necrotiz-
22q13.32-qter. Am J Hum Genet 1998;63:526–533. ing encephalopathy: oxidative phosphorylation defects and the ATPase
877. Van Goethem G, Schwartz M, Lofgren A, et al. Novel POLG mutations in 6 point mutation. Neurology 1992;42:2168–2174.
progressive external ophthalmoplegia mimicking mitochondrial neuro- 903. Pastores G, Santorelli F, Shanske S, et al. Leigh syndrome and hypertrophic
gastrointestinal encephalomyopathy. Eur J Hum Genet 2003;11:547–549. cardiomyopathy in an infant with a mitochondrial DNA point mutation
878. Nishigaki Y, Marti R, Hirano M. ND5 is a hot-spot for multiple atypi- (T8993G). Am J Med Genet 1994;50:265–271.
cal mitochondrial DNA deletions in mitochondrial neurogastrointestinal 904. DeVries DD, Van Engelen B, Gabrells F, et al. A second missense muta-
encephalomyopathy. Hum Mol Genet 2004;13:91–101. tion in the mitochondrial ATPase 6 gene in Leigh’s syndrome. Ann Neurol
879. Labauge P, Durant R, Castelnovo G, et al. MNGIE: diarrhea and leukoen- 1993;34:410–412.
cephalopathy. Neurology 2002;25:58. 905. Akagi M, Inui K, Tsukamoto H, et al. A point mutation of mitochondrial
880. Millar WS, Lignelli A, Hirano M. MRI of five patients with mitochon- ATPase 6 gene in Leigh syndrome. Neuromuscul Disord 2002;12(1):53.
drial neurogastrointestinal encephalomyopathy. Am J Roentgenol 906. Moslemi AR, Darin N, Tulinius M, et al. Two new mutations in the MTATP6
2004;182:1537–1541. gene associated with Leigh syndrome. Neuropediatrics 2005;36:314–318.
881. Leigh D. Subacute necrotizing encephalomyelopathy in an infant. J Neurol 907. Thyagarajan D, Shanske S, Vasquez-Memije M, et al. A novel mitochno-
Neurosurg Psychiatry 1951;14:216–221. drial ATPase 6 point mutation in familial bilateral striatal necrosis. Ann
882. Walter GF, Brucher JM, Martin JJ, et al. Leigh’s disease—several nosologi- Neurol 1995;38:468–472.
cal entities with an identical histopathological complex? Neuropath Appl 908. De Meirleir L, Seneca S, Lissens W, et al. Bilateral striatal necrosis with a
Neurobiol 1986;12:95–107. novel point mutation in the mitochondrial ATPase 6 gene. Pediatr Neurol
883. Robinson J, Norman MG. CPC: Feeding problems and lactic acidosis in a 1995;13:242–246.
10-week-old boy. Pediatr Neurosci 1989;15:28–35. 909. Holt IJ, Harding AE, Petty RK, et al. A new mitochondrial disease associated
884. Friede RL. Developmental neuropathology, 2nd ed. Berlin: Springer-Verlag; with mitochondrial DMA heteroplasmy. Am J Hum Genet 1990;46:428–433.
1989. 910. Morris aAM, Leonard JV, Brown GK, et al. Deficiency of respira-
885. DiMauro S, De Vivo DC. Genetic heterogeneity in Leigh syndrome. Ann tory chain complex I is a common cause of Leigh disease. Ann Neurol
Neurol 1996;40:5–7. 1996;40:25–30.

Barkovich_Chap03.indd 233 5/6/2011 12:09:36 AM


234 Pediatric Neuroimaging

911. Triepels RH, van den Heuvel LP, Loeffen JLCM, et al. Leigh syndrome 937. De Paepe A, Loeys B, Devriendt K, et al. Occipital horn syndrome in a
associated with a mutation in the NDUFS7 (PSST) nuclear encoded sub- 2-year-old boy. Clin Dysmorphol 1999;8:179–183.
unit of complex I. Ann Neurol 1999;45:787–790. 938. Bankier A. Menkes disease. J Med Genet 1995;32:213–215.
912. Kirby DM, Kahler SG, Freckmann M-L, et al. Leigh disease caused by the 939. Wesenberg RL, Gwinn JL, Barnes GR Jr. Radiological findings in the kinky
mitochondrial DNA G14459A mutation in unrelated families. Ann Neurol hair syndrome. Radiology 1969;92:500–506.
2000;48:102–104. 940. Seay AR, Bray PE, Wing SD, et al. CT scan in Menkes disease. Neurology
913. Moslemi AR, Darin N, Tulinius M, et al. Progressive encephalopathy and 1979;29:304–312.
complex I deficiency associated with mutations in MTND1 Neuropediat- 941. Faerber EN, Grover WD, DeFilipp GJ, et al. Cerebral MR of Menkes kinky-
rics 2008;39:24–28. hair disease. Am J Neuroradiol 1989;10(1):190–192.
914. Zafeiriou DI, Rodenburg RJT, Scheffer H, et al. MR spectroscopy and 942. Blaser SI, Berns DH, Ross JS, et al. Serial MR studies in Menkes disease.
serial magnetic resonance imaging in a patient with mitochondrial cystic J Comput Assist Tomogr 1989;13(1):113–115.
leukoencephalopathy due to complex I deficiency and NDUFV1 muta- 943. Johnsen DE, Coleman L, Poe L. MR of progressive neurodegen-
tions and mild clinical course. Neuropediatrics 2008;39:172–175. erative change in treated Menkes’ kinky hair disease. Neuroradiology
915. Hoefs SJG, Dieteren CEJ, Distelmaier F, et al. NDUFA2 complex I muta- 1991;33(2):181–182.
tion leads to Leigh disease. Am J Hum Genet 2008;82:1306–1315. 944. Ichihashi K, Yano S, Kobayashi S, et al. Serial imaging of Menkes disease.
916. Singleton CK, Martin PR. Molecular mechanism of thiamine utilization. Neuroradiol 1990;32:56–59.
Curr Mol Med 2001;1:197–207. 945. Strauss KA, Morton DH. Type I glutaric aciduria, part 2: a model of acute
917. Kornreich L, Bron-Harlev E, Hoffmann C, et al. Thiamine Deficiency in striatal necrosis. Am J Med Genet C Semin Med Genet 2003;121(1):53–70.
Infants: MR Findings in the Brain. Am J Neuroradiol 2005;26(7):1668– 946. Hedlund G, Longo N, Pasquali M. Glutaric adidemia type 1. Am J Med
1674. Genet 2006;142C:86–94.
918. Wyalt DT, Michael JN, Hillman RE. Infantile beriberi presenting as sub- 947. Hoffmann GF, Athanassopoulos S, Burlina AB, et al. Clinical course, early
acute necrotizing encephalomyelopathy. J Pediatr 1987;110:888–891. diagnosis, treatment, and prevention of disease in glutaryl-CoA dehydro-
919. Zuccoli G, Siddiqui N, Bailey A, et al. Neuroimaging findings in pediatric genase deficiency. Neuropediatrics 1996;27:115–123.
Wernicke encephalopathy: a review. Neuroradiology 2010;52(6):523–529. 948. Gordon N. Glutaric aciduria types I and II. Brain Dev 2006;28:136–140.
920. Brockmann K, Bjornstad A, Dechent P, et al. Succinate in dystrophic white 949. Bähr O, Mader I, Zachocke J, et al. Adult onset glutaric aciduria type I
matter: A proton magnetic resonance spectroscopy finding characteristic presenting with a leukoencephalopathy. Neurology 2002;59:1802–1804.
for complex II deficiency. Ann Neurol 2002;52(1):38–46. 950. Mellerio C, Marignier S, Roth P, et al. Prenatal cerebral ultrasound and
921. Bourgeron T, Rustin P, Chretien D, et al. Mutation of a nuclear succinate MRI findings in glutaric aciduria Type 1: a de novo case. Ultrasound Obstet
dehydrogenase gene results in mitochnodrial respiratory chain deficiency. Gynecol 2008;31:712–714.
Nature Genet 1995;11:144–149. 951. Altman NR, Rovira MJ, Bauer M. Glutaric aciduria type I: MR findings in
922. Krägeloh-Mann I, Grodd W, Schöning M, et al. Proton spectroscopy in two cases. Am J Neuroradiol 1991;12:966–968.
five patients with Leigh’s disease and mitochondrial enzyme deficiency. 952. Brismar J, Ozand PT. CT and MR of the brain in glutaric acidemia type I:
Dev Med Child Neurol 1993;35:769–776. a review of 59 published cases and a report of 5 new patients. Am J Neuro-
923. Alpers BJ. Progressive cerebral degeneration in infancy. J Nerv Ment Dis radiol 1995;16:675–683.
1960;130:442–448. 953. Martinez-Lage JF, Casas C, Fernandez M, et al. Macrocephaly, dystonia
924. Alpers BJ. Diffuse progressive degeneration of the grey matter of the cere- and bilateral temporal arachnoid cysts: glutaric aciduria type 1. Child’s
brum. Arch Neurol Psychiat 1931;25:469–505. Nerv Syst 1994;10:198–203.
925. Harding BN. Progressive neuronal degeneration of childhood with liver 954. McErlean A, Abdalla K, Donoghue V, et al. The dentate nucleus in children:
disease (Alpers-Huttenlocher syndrome): a personal review. J Child Neurol normal development and patterns of disease. Pediatr Radiol 2010;40:326–
1990;5(4):273–287. 339.
926. Naviaux RK, Nyhan WL, Barshop BA, et al. Mitochondrial DNA poly- 955. Oguz KK, Ozturk A, Cila A. Diffusion-weighted MR imaging and MR spec-
merase gamma deficiency and mtDNA depletion in a child with Alpers’ troscopy in glutaric aciduria, type 1. Neuroradiology 2005;47:229–234.
syndrome. Ann Neurol1999;45(1):54–58. 956. Awaad Y, Shamato H, Chugani H. Hemidystonia improved by beclofen
927. Naviaux RK, Nguyen KV. POLG mutations associated with Alpers’ syndrome and PET scan findings in a patient with glutaric aciduria type 1. J Child
and mitochondrial DNA depletion. Ann Neurol 2004;55(5):706–712. Neurol 1996;11:167–169.
928. Menkes JH. Genetic disorders of mitochondrial function. J Pediatr 957. Amendt B, Rhead W. The multiple acyl-coenzyme A dehydrogenation
1987;110(2):255–259. disorders, glutaric aciduria type II and ethylmalonic-adipic aciduria.
929. Egger J, Harding BN, Boyd SG, et al. Progressive neuronal degeneration of Mitochondrial fatty acid oxidation, acyl-coenzyme A dehydrogenase,
childhood (PNDC) with lever disease. Brain 1987;26:167–173. and electron transfer flavoprotein activities in fibroblasts. J Clin Invest
930. Wiltshire E, Davidzon G, DiMauro S, et al. Juvenile Alpers disease. Arch 1986;78:205–213.
Neurol 2008;65:121–124. 958. Shevell M, Didomenicantonio G, Sylvain M, et al. Glutaric acidemia
931. Brunetti-Pierri N, Selby K, O’Sullivan M, et al. Rapidly progressive neuro- type II: neuroimaging and spectroscopy evidence for developmental
logical deterioration in a child with Alpers syndrome exhibiting a previ- encephalomyopathy. Pediatr Neurol 1995;12:350–353.
ously unremarkable brain MRI. Neuropediatrics 2008;39:179–183. 959. Dusheiko G, Kew MC, Joffe BI, et al. Recurrent hypoglycemia associated
932. Menkes JH, Alter M, Steigleder GK, et al. A sex-linked recessive disorder with glutaric aciduria type II in an adult. N Eng J Med 1979;301:1405–1409.
with retardation of growth, peculiar hair, and focal cerebral and cerebellar 960. Goodman SI, Frerman FE, Loehr JP. Recent progress in understanding
degeneration. Pediatrics 1962;29:764–779. glutaric acidemias. Enzyme 1987;38:76–79.
933. Chelly J, Turner Z, Tonnesen T, et al. Isolation of a candidate gene for 961. Stockler S, Radner H, Karpf EF, et al. Symmetric hypoplasia of the tempo-
Menkes disease that codes a potential heavy metal binding protein. Nature ral cerebral lobes in an infant with glutaric aciduria type II (myltiple acyl-
Genet 1993;3:14–19. coenzyme A dehydrogenase deficiency). J Pediatr 1994;124(4):601–604.
934. Vulpe C, Levinson B, Whitney S, et al. Isolation of a candidate gene for 962. Takanashi J, Fujii K, Sugita K, et al. Neuroradiologic findings in glutaric
Menkes disease and evidence that it encodes a copper-transport ATPase. aciduria type II. Pediatr Neurol 1999;20(2):142–145.
Nature Genet 1993;3:7–13. 963. Tiranti V, D’Adamo P, Briem E, et al. Ethylmalonic encephalopathy is
935. Kaler SG, Holmes CS, Goldstein DS, et al. Neonatal diagnosis and treat- caused by mutations in ETHE1, a gene encoding a mitochondrial matrix
ment of Menkes disease. N Engl J Med 2008;358:605–614. protein. Am J Hum Genet 2004;74:239–252.
936. Proud VK, Mussell HG, Kaler SG, et al. Distinctive Menkes disease variant 964. Burlina AB, Dionisi-Vici C, Bennett MJ, et al. A new syndrome with ethyl-
with occipital horns: delineation of natural history and clinical pheno- malonic aciduria and normal fatty acid oxidation in fibroblasts. J Pediatr
type. Am J Med Genet 1996;65:44–51. 1994;124:79–86.

Barkovich_Chap03.indd 234 5/6/2011 12:09:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 235

965. Burlina AB, Zacchello F, Dionisi-Vici C, et al. New clinical pheno- 991. Bajaj SK, Kurlemann G, Schuierer G, et al. CT and MRI in a girl with
type of brainched-chain acyl-CoA oxidation defect (Letter). Lancet late-onset ornithine transcarbamylase deficiency: case report. Neuroradi-
1991;338:1522–1523. ology 1996;38:796–799.
966. Heberle L, Al Tawari A, ramadan D, et al. Ethylmalonic encephalopathy— 992. Choi C-G, Yoo HW. Localized proton MR spectroscopy in infants with
report of two cases. Brain Dev 2006;28:329–331. urea cycle defect. Am J Neuroradiol 2001;22:834–837.
967. Zafeiriou DI, Augoustides-Savvopoulou P, Haas D, et al. Ethylmalonic 993. Kojic J, Robertson PL, Quint DJ, et al. Brain glutamine by MRS in a patient
encephalopathy: clinical and biochemical observations. Neuropediatrics with urea cycle disorder and coma. Pediatr Neurol 2005;32(2):143–146.
2007;38:78–82. 994. Ugarte M, Perez-Cerda C, Rodrigeuz-Pombo P, et al. Overview of muta-
968. Nowaczyk MJM, Blaser SI, Clarke JTR. Central nervous system malforma- tions in the PCCA and PCCB genes causing propionic acidemia. Hum
tions in ethylmalonic encephalopathy. Am J Med Genet 1998;75:292–296. Mutat 1999;14:275–282.
969. Sandhu J, Dillon WP. MR in mitochondrial encephalomyelopathy. Am J 995. Ledley F, Lumetta M, Zoghbi H, et al. Mapping of the human methyl-
Neuroradiol 1991;12:375–379. malonyl CoA mutase (MUT) locus on chromosome 6. Am J Hum Genet
970. Blaser S, Feigenbaum A, Becker L, et al. Lethal neonatal mitochondrial dis- 1988;42:839–846.
ease: a radiographic mimic of perinatal asphyxia. Paper presented at: Ameri- 996. Andreula CF, De Blasi R, Carella A. CT and MR studies of methylmalonic
can Society of Neuroradiology 31st Annual Meeting 1993; Vancouver, B.C. acidemia. Am J Neuroradiol 1991;12:410–412.
971. Lamperti C, Naini A, Hirano M, et al. Cerebellar ataxia and coenzyme Q10 997. Surtees RAH, Matthews EE, Leonard JV. Neurologic outcome of propi-
deficiency. Neurology 2003;60:1206–1208. onic acidemia. Pediatr Neurol 1992;8:333–337.
972. Musumeci O, Naini A, Slonim A, et al. Familial cerebellar ataxia with 998. Al-Essa M, Bakheet S, Patay Z, et al. 18-fluoro-2-deoxyglucose PET scan
muscle coenzyme Q10 deficiency. Neurology 2001;56:849–855. of the brain in propionic acidemia: clinical and MRI correlations. Brain
973. de Lonlay-Debeney P, von Kleist-Retzow J, Hertz-Pannier L, et al. Cerebral Dev 1999;21:312–317.
white matter disease in children may be caused by mitochondrial respira- 999. Bergman AJ, van der Knaap MS, Smeitink JA, et al. Magnetic resonance
tory chain deficiency. J Pediatr 2000;2000:209–214. imaging and spectroscopy of the brain in propionic acidemia: clinical
974. Lissens W, De Meirleir L, Seneca S, et al. Mutations in the X-linked pyruvate and biochemical considerations. Pediatr Res 1996;40:404–409.
dehydrogenase (E1) alpha subunit gene (PDHA1) in patients with a pyru- 1000. Gebarski SS, Gabrielsen TO, Knake JE, et al. Cerebral CT findings in
vate dehydrogenase complex deficiency. Hum Mutat 2000;15:209–219. methylmalonic and propionic acidemias. Am J Neuroradiol 1983;4:
975. Head RA, Brown RM, Zolkipli Z, et al. Clinical and genetic spectrum of 955–957.
pyruvate dehydrogenase deficiency: dihydrolipoamide acetyltransferase 1001. Radmanesh A, Zaman T, Ghanaati H, et al. Methylmalonic acidemia:
(E2) deficiency. Ann Neurol 2005;58(2):234–241. brain imaging findings in 52 children and a review of the literature. Pedi-
976. Brown R, Head R, Brown G. Pyruvate dehydrogenase E3 binding protein atr Radiol 2008;38:1054–1061.
deficiency. Hum Genet 2002;110:187–191. 1002. Trinh B-C, Melhem ER, Barker PB. Multislice proton MR spectroscopy
977. Dey R, Mine M, Desguerre I, et al. A new case of pyruvate dehydroge- and diffusion weighted imaging in methylmalonic acidemei: report of two
nase deficiency due to a novel mutation in the PDX1 gene. Ann Neurol cases and review of the literature. Am J Neuroradiol 2001;22:831–833.
2003;53:273–277. 1003. Suzuki Y, Sakuraba H, Oshima A. Beta galactosidase deficiency (beta-
978. De Vivo DC. Complexities of the pyruvate dehydrogenase complex. galactosidosis): GM1 gangliosidosis and Morquio B disease. In: Scriver
Neurology 1998;51:1247–1249. CR, Beaudet AL, Sly WS, eds. The Metabolic and Molecular Bases of Inher-
979. De Vivo DC. The expanding clinical spectrum of mitochondrial diseases. ited Disease. New York: McGraw-Hill, 1995:2785–2823.
Brain Dev 1993;15:1–21. 1004. Chen C-Y, Zimmerman RA, Lee C-C, et al. Neuroimaging findings in late
980. Strassburg HM, Koch J, Mayr J, et al. Acute flacid paralysis as initial symp- infantile GM1 gangliosidosis. Am J Neuroradiol 1998;19:1628–1630.
tom in 4 patients with novel E1-alpha mutations of the pyruvate dehydro- 1005. Takano T, Yamanouchi Y. Assignment of human beta-galactosidase-A
genase complex. Neuropediatrics 2006;37:137–141. gene to 3p21.33 by fluorescence in situ hybridization. Hum Genet
981. Cross JH, Connelly A, Gadian DG, et al. Clinical diversity of pyruvate 1993;92:403–404.
dehydrogenase deficiency. Pediatr Neurol 1994;10:276–283. 1006. Brunetti-Pierri N, Scaglia F. GM1 gangliosidosis: review of clinical,
982. Wada N, Matsuishi T, Nonaka M, et al. Pyruvate dehydrogenase E1 molecular, and therapeutic aspects. Mol Genet Metab 2008;94:391–396.
alpha subunit deficiency in a female patient: evidence of antenatal ori- 1007. Myerowitz R, Lawson D, Mizukami H, et al. Molecular pathophysiology
gin of brain damage and possible etiology of infantile spasms. Brain Dev in Tay-Sachs and Sandhoff diseases as revealed by gene expression profil-
2004;26:57–60. ing. Hum Mol Genet 2002;11:1343–1350.
983. Soares-Fernandes J, Teixeira-Gomes R, Cruz R, et al. Neonatal pyruvate 1008. Inui K, Grebner EE, Jackson LG, et al. Juvenile GM2 gangliosidosis (AMB
dehydrogenase deficiency due to a R302H mutation in the PDHA1 gene: variant): inability to activate hexosaminidase A by activator protein. Am
MRI findings. Pediatr Radiol 2008;38(5):559–562. J Hum Genet 1983;35:551–564.
984. Zand DJ, Simon EM, Pulitzer SB, et al. In vivo pyruvate detected by MR 1009. Koelfen W, Freund M, Jaschke W, et al. GM2 gangliosidosis (Sandhoff ’s
spectroscopy in neonatal pyruvate dehydrogenase deficiency. Am J Neuro- disease): two year follow-up by MRI. Neuroradiology 1994;36:152–154.
radiol 2003;24(7):1471–1474. 1010. Gravel R, Clarke J, Kaback M. The GMZ gangliosidoses. In: Scriver C,
985. Bosoi CR, Rose CF. Identifying the direct effects of ammonia on the brain. Beaudet A, Sly W, et al., eds. The Metabolic and Molecular Bases of Inher-
Metab Brain Dis 2009;24:95–102. ited Disease, 7th ed. New York: McGraw Hill, 1995:2839–2879.
986. Enns GM. Neurologic damage and neurocognitive dysfunction in urea 1011. Maegawa GHB, Stockley T, Tropak M, et al. The Natural History of Juve-
cycle disorders. Semin Pediatr Neurol 2008;15:132–139. nile or Subacute GM2 Gangliosidosis: 21 New Cases and Literature Review
987. Bartholomew DW, Brusilow SW. Abnormalities of urea cycle and of 134 Previously Reported. Pediatrics 2006;118(5):e1550–e1562.
ammonium. In: Berg B, ed. Neurologic Aspects of Pediatrics. Boston: 1012. Frey LC, Ringel SP, Filley CM. The natural history of cognitive dysfunc-
Butterworth-Heinemann, 1992:33–46. tion in late-onset GM2 gangliosidosis. Arch Neurol 2005;62:989–994.
988. Brusilow SW, Horwich AL. Urea cycle enzymes. In: Scrivner CR, Beaudet 1013. Kobayashi O, Takashima S. Thalamic hyperdensity on CT in infantile
AL, Sly WS, et al., eds. The Metabolic and Molecular Bases of Inherited Dis- GM1 gangliosidosis. Brain Dev 1994;16:472–474.
ease, 8th ed. New York: McGraw-Hill; 2001:1909–1963. 1014. Mugikura S, Takahashi S, Higano S, et al. MR findings in Tay-Sachs dis-
989. Brusilow SW. Inborn errors of urea synthesis: paradigm of hyperam- ease. J Comput Assist Tomogr 1996;20:551–555.
monemic encephalopathy. In: Berg BO, ed. Principles of Child Neurology. 1015. Stalker HP, Han BK. Thalamic hyperdensity: a previously unreported
New York: McGraw-Hill, 1996:979–996. sign of Sandhoff disease. Am J Neuroradiol 1989;10:S82.
990. Takanashi J, Barkovich AJ, Cheng S, et al. Brain MR imaging in acute 1016. De Grandis E, Di Rocco M, Pessagno A, et al. MR imaging findings in
hyperammonemic encephalopathy arising from late-onset ornithine tran- 2 cases of late infantile GM1 gangliosidosis. AJNR Am J Neuroradiol
scarbamylase deficiency. Am J Neuroradiol 2003;24:390–393. 2009;30:1325–1327.

Barkovich_Chap03.indd 235 5/6/2011 12:09:36 AM


236 Pediatric Neuroimaging

1017. Aydin K, Bakir B, Tatli B, et al. Proton MR spectroscopy in three children 1042. Nakano I, Otsuki N, Hasegawa A. Acute stage neuropathology of a case
with Tay-Sachs disease. Pediatr Radiol 2005;35:1081–1085. of infantile acute encephalopathy with thalamic involvement: wide-
1018. Imamura A, Miyajima H, Ito R, et al. Serial MR imaging and 1H-MR spread symmetrical fresh necorsis of the brain. Neuropathology (Kyoto)
spectroscopy in monozygotic twins with Tay-Sachs disease. Neuropediat- 1993;13:315–325.
rics 2008;39:259–263. 1043. Olgar S, Ertugrul T, Nisli K, et al. Influenza A-associated acute necrotizing
1019. Wilken B, Dechent P, Hanefeld F, et al. Proton MRS of a child with Sand- encephalopathy. Neuropediatrics 2006;37:166–168.
hoff disease reveals elevated brain hexosamine. Eur J Paediatr Neurol 1044. Oki J, Yoshida H, Tokumitsu A, et al. Serial neuroimages of acute necro-
2008;12:56–60. tizing encephalopathy associated with human herpesvirus 6 infection.
1020. Galluzzi P, Rufa A, Balestri P, et al. MR brain imaging of fucosidosis type Brain Dev 1995;17:356–359.
1. Am J Neuroradiol 2001;22:777–780. 1045. Skelton BW, Hollingshead MC, Sledd AT, et al. Acute necrotizing enceph-
1021. Provenzale JM, Barboriak D, Sims K. Neuroradiologic findings in alopathy of childhood: typical findings in an atypical disease. Pediatr
fucosidosis, a rare lysosomal storage disease. Am J Neuroradiol 1995;16: Radiol 2008;38:810–813.
809–813. 1046. Nagai T, Yagishita A, Tsuchiya Y, et al. Symmetrical thalamic lesions on
1022. Durand P, Borrone C, Della Cella G. Fucosidosis. J Pediatr 1969;75: CT in influenza A virus infection presenting with or without Reye syn-
665–674. drome. Brain Dev 1993;15:67–73.
1023. Oner AY, Cansu A, Akpek S, Serdaroglu A. Fucosidosis: MRI and MRS 1047. Neilson DE, Adams MD, Orr CMD, et al. Infection-triggered familial or
findings. Pediatr Radiol 2007;37:1050–1052. recurrent cases of acute necrotizing encephalopathy caused by muta-
1024. Topçu M, Jobard F, Halliez S, et al. L-2-hydroxyglutaric aciduria: identi- tions in a component of the nuclear pore, RANBP2. Am J Hum Genet
fication of a mutant gene C14orf160, localized on chromosome 14q22.1. 2009;84:44–51.
Hum Mol Genet 2004;13:2803–2811. 1048. Neilson DE, Feiler HS, Wilhelmsen KC, et al. Autosomal dominant
1025. D’Incerti L, Farina L, Moroni I, et al. L-2-hydroxyglutaric aciduria: MRI acute necrotizing encephalopathy maps to 2q12.1–2q13. Ann Neurol
in seven cases. Neuroradiology 1998;40:727–733. 2004;55:291–294.
1026. Barth PG, Hoffmann GF, Jaeken JJ, et al. L-2-hydroxyglutaric acidemia: a 1049. Neilson DE, Eiben RM, Waniewski S, et al. Autosomal dominant acute
novel inherited neurometabolic disease. Ann Neurol 1992;32:66–71. necrotizing encephalopathy. Neurology 2003;61:226–230.
1027. Barth PG, Hoffmann GF, Jaeken JJ, et al. L-2-hydroxyglutaric acidae- 1050. Mizuguchi M, Hayashi M, Nakano I, et al. Concentric structure of
mia: clinical and biochemical findings in 12 patients and preliminary thalamic lesions in acute necrotizing encephalopathy. Neuroradiology
report on l-2-hydroxyacid dehydrogenase. J Inherit Metab Dis 1993;16: 2002;44:489–493.
753–761. 1051. Albayram S, Bilgi Z, Selcuk H, et al. Diffusion weighted MRI imag-
1028. Barth PG. Commentary. 2-Hydroxyglutaric acidurias. Brain Dev 1995;17: ing findings of acute necrotizing encephalopathy. Am J Neuroradiol
144–145. 2004;25:792–797.
1029. Barbot C, Fineza I, Diogo L, et al. L-2-Hydroxyglutaric aciduria: clinical, 1052. van der Knaap M, Linnankivi T, Paetau A, et al. Hypomyelination with
biochemical and magnetic resonance imaging in six Portuguese pediatric atrophy of the basal ganglia and cerebellum: follow-up and pathology.
patients. Brain Dev 1997;19:268–273. Neurology 2007;69:166–171.
1030. Chen E, Nyhan WL, Jakobs C, et al. L-2-Hydroxyglutaric aciduria: neu- 1053. Mercimek-Mahmutoglu S, van der Knaap MS, Baric I, et al. Hypomy-
ropathological correlations and first report of severe neurodegenerative elination with atrophy of the basal ganglia and cerebellum (H-ABC).
disease and neonatal death. J Inherit Metab Dis 1996;19:335–343. Report of a new case. Neuropediatrics 2005;36:223–226.
1031. Larnaout A, Hentati F, Belal S, et al. Clinical and pathological study of 1054. Gunay-Aygun M. 3-Methylglutaconic aciduria: a common biochemical
three Tunisian siblings with L-2-hydroxyglutaric aciduria. Acta Neuro- marker in various syndromes with diverse clinical features. Mol Genet
pathol 1994;88:367–370. Metab 2005;84:1–3.
1032. Seijo-Martinez M, Navarro C, Castro del Rio M, et al. L-2-hydroxygl- 1055. Gibson K, Elpeleg O, Jakobs C, et al. Multiple syndromes of
utaric aciduria: clinical, neuroimaging, and neuropathological findings. 3-methylglutaconic aciduria. Pediatr Neurol. 1993;9:120–123.
Arch Neurol 2005;62:666–670. 1056. Ijlst L, Loupatty F, Ruiter J, et al. 3-Methylglutaconic aciduria type I is
1033. Steenweg M, Salomons G, Yapici Z, et al. L-2-Hydroxyglutaric acidu- caused by mutations in AUH. Am J Hum Genet 2002;71:1463–1466.
ria: pattern of MR imaging abnormalities in 56 patients. Radiology 1057. Barth PG, Valianpour F, Bowen VM, et al. X-linked cardioskeletal myo-
2009;251:856–865. pathy and neutropenia (Barth syndrome): an update. Am J Med Genet.
1034. Aydin K, Ozmen M, Tatli B, et al. Single-voxel MR spectroscopy and 2004;126A:349–354.
diffusion-weighted MRI in two patients with l-2-hydroxyglutaric acidu- 1058. Johnston J, Kelley RI, Feigenbaum A, et al. Mutation characterization and
ria. Pediatr Radiol 2003;33(12):872–876. genotype-phenotype correlation in Barth syndrome. Am J Hum Genet
1035. Aghili M, Zahedi F, Rafiee E. Hydroxyglutaric aciduria and malig- 1997;61:1053–1058.
nant brain tumor: a case report and literature review. J Neurooncol 1059. Kelley RI, Cheatham JP, Clark BJ, et al. X-linked cardiomyopathy with
2009;91:233–236. neutropenia, growth retardation and 3-methylglutaconic aciduria. J
1036. Komotar RJ, Starke RM, Sisti MB, et al. IDH1 and IDH2 mutations in Pediatr 1991;119:738–747.
gliomas and the associated induction of hypoxia-inducible factor and 1060. Costeff H, Elpeleg O, Apter N, et al. 3-Methylglutaconic aciduria in “optic
production of 2-hydroxyglutarate. Neurosurgery 2010;66:N20–N21. atrophy plus.” Ann Neurol 1993;33:103–104.
1037. Mizuguchi M. Acute necrotizing encephalopathy of childhood: a novel 1061. Chitayat D, Chemke J, Gibson KM, et al. 3-Methylglutaconic aciduria: a
form of acute encephalopathy prevalent in Japan and Taiwan. Brain Dev marker for as yet unspecified disorders and the relevance of prenatal diag-
1997;19:81–92. nosis in a “new” type (“type 4”). J Inherit Metab Dis 1992;15:204–212.
1038. Mizuguchi M, Abe J, Mikkaichi K, et al. Acute necrotizing encephalopa- 1062. Jareño N, Fernández-Mayoralas D, Silvestre C, et al. 3-methylglutaconic
thy of childhood: a new syndrome presenting with multifocal, symmetric aciduria type 4 manifesting as Leigh syndrome in 2 siblings. J Child Neu-
brain lesions. J Neurol Neurosurg Psychiatry. 1995;58:555–561. rol. 2007;22:218–221.
1039. Mizuguchi M. Acute encephalopathy with necrosis of bilateral thalami: 1063. Davey KM, Parboosingh JS, McLeod DR, et al. Mutation of DNAJC19, a
clinical aspects. Neuropathology (Kyoto) 1993;13:327–331. human homologue of yeast inner mitochondrial co-chaperones, causes
1040. Yagishita A, Nakano I, Ushioda T, et al. Acute encephalopathy with bilat- DCMA syndrome, a novel autosomal recessive Barth syndrome-like con-
eral thalamotegmental involvement in infants and children: imaging and dition. J Med Genet 2006;43:385–393.
pathology findings. Am J Neuroradiol 1995;16:439–447. 1064. Reiss J. Genetics of molybdenum cofactor deficiency. Hum Genet
1041. Wong AM, Simon EM, Zimmerman RA, et al. Acute necrotizing enceph- 2000;106:157–163.
alopathy of childhood: correlation of MR findings and clinical outcome. 1065. Reiss J, Gross-Hardt S, Christensen E, et al. A mutation in the gene for
Am J Neuroradiol 2006;27:1919–1923. the neurotransmitter receptor-clustering protein gephyrin causes a novel

Barkovich_Chap03.indd 236 5/6/2011 12:09:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 237

form of molybdenum cofactor deficiency. Am J Hum Genet 2001;68: 1090. Brett EM, Ellis RB, Haas L, et al. Late onset GM2-gangliosidosis. Clini-
218–213. cal, pathological, and biochemical studies on 8 patients. Arch Dis Child
1066. Johnson JL, Wadman SK. Molybdenum cofactor deficiency. In: Scriver 1973;48:775–785.
CR, ed. The Metabolic Basis of Inherited Disease. New York: McGraw-Hill, 1091. Streifler JY, Gornish M, Hadar H, et al. Brain imaging in late onset GM2
1989:183–190. gangliosidosis. Neurology 1993;43:2055–2058.
1067. Endres W, Shin YS, Guenther R, et al. Report on a new patient with com- 1092. Inglese M, Nusbaum AO, Pastores GM, et al. MR imaging and proton
bined deficiencies of sulfite oxidase and xanthine dehydrogenase due to spectroscopy of neuronal injury in late-onset GM2 gangliosidosis. Am J
molybdenum cofactor deficiency. Eur J Pediatr 1988;148:246–249. Neuroradiol 2005;26(8):2037–2042.
1068. Ichord RN. Perinatal metabolic encephalopathies. In: Swaiman KF, 1093. Moreira M-C, Barbot C, Tachi N, et al. The gene mutated in ataxia-
Ashwal S, Ferriero DM, eds. Pediatric Neurology: Principles and Practice, oculomotor apraxia 1 encodes the new HIT/Zn-finger protein aprataxin.
4th ed. Philadelphia: Mosby Elsevier, 2006:345–362. Nat Genet 2001;29:189–193.
1069. Roth A, Nogues C, Monnet JP, et al. Anatomopathological findings in a 1094. Kijas AW, Harris JL, Harris JM, et al. Aprataxin forms a discrete branch
case of combined deficiency of sulfite oxidase and xanthine oxidase with in the HIT (histidine triad) superfamily of proteins with both DNA/RNA
a defect of molybdenum cofactor. Virchows Arch [A] 1985;405:379–386. bindings and nucleotide hydrolase activities. J Biol Chem 2006;281:13939–
1070. Brown G, Scholem R, Croll H, et al. Sulfite oxidase deficiency: clinical, 13948.
neuroradiologic and biochemical features in two new patients. Neurology 1095. Aicardi J, Barbosa C, Andermann E, et al. Ataxia-ocular motor apraxia:
1989;39:252–257. a syndrome mimicking ataxia-telangiectasia. Ann Neurol 1988;24:
1071. Barth PG, Beemer FA, Cats BP, et al. Neuropathological findings in a case 497–502.
of combined deficiency of sulfite oxidase and xanthine dehydrogenase. 1096. Le Ber I, Moreira M-C, Rivaud-Pechoux S, et al. Cerebellar ataxia
Virchows Arch [A] 1985;408:105–106. with oculomotor apraxia type 1: clinical and genetic studies. Brain
1072. Appignani BA, Kaye EM, Wolpert SM. CT and MR appearance of the 2003;126(12):2761–2772.
brain in two children with molybdenum cofactor deficiency. Am J Neuro- 1097. Fogel BL, Perlman SL. Clinical features and moledular genetics of auto-
radiol 1996;17:317–320. somal recessive cerebellar ataxias. Lancet Neurol 2007;6:245–257.
1073. Schuierer G, Kurlemann G, Bick U, et al. Molybdenum-cofactor defi- 1098. Nemeth AH, Bochukova E, Dunne E, et al. Autosomal recessive cerebellar
ciency: CT and MR findings. Neuropediatrics 1995;26:51–54. ataxia with oculomotor apraxia (ataxia-telangiectasia-like syndrome) is
1074. Serrano M, Lizarraga I, Reiss J, et al. Cranial ultrasound and chrono- linked to chromosome 9q34. Am J Hum Genet 2000;67:1320–1326.
logical changes in molybdenum cofactor deficiency. Pediatr Radiol 1099. Le Ber I, Bouslam N, Rivaud-Pechoux S, et al. Frequency and phenotypic
2007;37(10):1043–1046. spectrum of ataxia with oculomotor apraxia 2: a clinical and genetic
1075. Graf WD, Oleinik OE, Jack RM, et al. Ahomocysteinemia in molybde- study in 18 patients. Brain 2004;127(4):759–767.
num cofactor deficiency. Neurology 1998;51:860–862. 1100. Anheim M, Monga B, Fleury M, et al. Ataxia with oculomotor apraxia
1076. Teksam O, Yurdakok M, Coskun T. Molybdenum cofactor deficiency type 2: clinical, biological and genotype/phenotype correlation study of a
presenting with severe metabolic acidosis and intracranial hemorrhage. cohort of 90 patients. Brain 2009;132:2688–2698.
J Child Neurol 2005;20:155–157. 1101. Criscuolo C, Chessa L, Di Giandomenico S, et al. Ataxia with oculomo-
1077. Topçu M, Çoskun T, Haliloglu G, et al. Molybdenum cofactor deficiency: tor apraxia type 2: A clinical, pathologic, and genetic study. Neurology
report of three cases presenting as hypoxic-ischemic encephalopathy. 2006;66:1207–1210.
J Child Neurol 2001;16:264–270. 1102. Bouchard UP, Barbeau A, Bouchard R, et al. Autosomal recessive spastic
1078. Garrett R, Bellissimo D, Rajagopalan K. Molecular cloning of human liver ataxia of Charlevoix-Saguenay. Can J Neurol Sci 1978;5:61–69.
sulfite oxidase. Biochem Biophys Acta 1995;1262:147–149. 1103. Gücüyener K, Özgül K, Paternotte C, et al. Autosomal recessive spastic
1079. Garrett R, Johnson J, Graf T, Feigenbaum A, Rajagopalan K. Human sufite ataxia of Charlevoix-Saguenay in two unrelated Turkish families. Neuro-
oxidase R160Q: identification of the mutation in a sulfite oxidase defi- pediatrics 2001;32:142–146.
cient patient and expression and characterization of the mutant enzyme. 1104. Engert J, Dore C, Mercier J, et al. Autosomal recessive spastic ataxia of
Proc Nat Acad Sci 1998;95:6394–6398. Charlevoix-Sanguenay (ARSACS): high-resolution physical and tran-
1080. Basheer SN, Waters PJ, Lam CW, et al. Isolated sulfite oxidase deficiency script map of the candidate regoin in chromosome region 13q11. Genom-
in the newborn: lactic acidaemeia and leukoencephalopathy. Neuropedi- ics 1999;62:156–164.
atrics 2007;38:38–41. 1105. Bouchard J-P, Richter A, Mathieu J, et al. Autosomal recessive spas-
1081. Lee H, Mak B, Chi C, et al. A novel mutation in neonatal isolated sulphite tic ataxia of Charlevoix-Saguenay. Neuromuscul Disord 1998;8(7):
oxidase deficiency. Neuropediatrics 2002;33:174–179. 474–479.
1082. Dublin A, Hald J, Wootton-Gorges S. Isolated sulfite oxidase deficiency: 1106. Mollet J, Delahodde A, Serre V, et al. CABC1 gene mutations cause
MR imaging features. Am J Neuroradiol 2002;23:484–485. ubiquinone deficiency with cerebellar ataxia and seizures. Am J Hum
1083. De Michele G, Di Maio L, Filla A, et al. Childhood onset of Friedreich ataxia: Genet 2008;82(3):623–630.
a clinical and genetic study of 36 cases. Neuropediatrics1996;27:3–7. 1107. Naini A, Lewis V, Hirano M, et al. Primary coenzyme Q10 deficiency and
1084. Montermini L, Andermann E, Labuda M, et al. The Friedreich ataxia the brain. Biofactors 2003;18:145–152.
GAA triplet repeat: premutation and normal alleles. Hum Mol Genet 1108. Rotig A, Appelkvist EL, Geromel V, et al. Quinone responsive multiple
1997;6:1261–1266. respiratory chain dysfunction due to widespread coenzyme Q10 defi-
1085. Harding AE. Friedreich’s ataxia: a clinical and genetic study of 90 families ciency. Lancet 2000;356:391–395.
with an analysis of early diagnostic criteria and intrafamilial clustering of 1109. Ogasahara S, Engel AG, Frens D, et al. Muscle coenzyme Q deficiency
clinical features. Brain 1981;104:589–595. in familial mitochondrial encephalomyopathy. Proc Natl Acad Sci
1086. Lamarche JB, Lemieux B, Lieu HG. The neuropathology of typical Frie- 1989;86:2379–2382.
dreich’s ataxia in Quebec. Can J Neurol Sci 1984;11:592–600. 1110. Quinzii C, Naini A, Salviati L, et al. A mutation in Para-hydroxybenzoate-
1087. Bhidayasiri R, Perlman SL, Pulst SM, et al. Late-onset Friedreich ataxia: polyprenyl transferase (COQ2) causes primary coenzyme Q10 deficiency.
phenotypic analysis, magnetic resonance imaging findings, and review of Am J Hum Genet 2006;78:345–349.
the literature. Arch Neurol 2005;62:1865–1869. 1111. Lopez LC, Schuelke M, Quinzii CM, et al. Leigh syndrome with neph-
1088. Della Nave R, Ginestroni A, Tessa C, et al. Brain white matter tracts degen- ropathy and CoQ10 deficiency due to decaprenyl diphosphate synthase
eration in Friedreich ataxia. An in vivo MRI study using tract-based spa- subunit 2 (PDSS2) mutations. Am J Hum Genet 2006;79:1125–1129.
tial statistics and voxel-based morphometry. NeuroImage 2008;40:19–25. 1112. Mollet J, Giurgea I, Schlemmer D, et al. Prenyldiphosphate synthase, sub-
1089. Argov Z, Navon R. Clinical and genetic variations in the syndrome of unit 1 (PDSS1) and OH-benzoate polyprenyltransferase (COQ2) muta-
adult GM2 gangliosidosis resulting from hexosaminidase A deficiency. tions in ubiquinone deficiency and oxidative phosphorylation disorders.
Ann Neurol 1984;16:14–20. J Clin Invest 2007;117:765–772.

Barkovich_Chap03.indd 237 5/6/2011 12:09:36 AM


238 Pediatric Neuroimaging

1113. Van Goethem G, Martin JJ, Dermaut B, et al. Recessive POLG mutations 1139. Bjursell C, Wahlstrom J, Berg K, et al. Detailed mapping of the
presenting with sensory and ataxic neuropathy in compound heterozy- phosphomannomutase 2 (PMM2) gene and mutation detection enable
gote patients with progressive external ophthalmoplegia. Neuromuscul improved analysis for Scandinavian CDG type I families. Eur J Hum
Disord 2003;13:133–142. Genet 1998;6:603–611.
1114. Winterthun S, Ferrari G, He L, et al. Autosomal recessive mitochondrial 1140. Matthijs G, Schollen E, Van Schaftingen E, et al. Lack of homozygotes for
ataxic syndrome due to mitochondrial polymerase-gamma mutations. the most frequent disease allele in carbohydrate-deficient glycoprotein
Neurology 2005;64:1204–1208. syndrome type IA. Am J Hum Genet 1998;62:542–550.
1115. Nikali K, Isosomppi J, Lonnqvist T, et al. Toward cloning of a novel ataxia 1141. Jaeken J, Matthijs G, Barone R, et al. Carbohydrate deficient glycoprotein
gene: refined assignment and physical map of the IOSCA locus (SCA8) syndrome type I. J Med Genet 1997;34:73–76.
on 10q24. Genomics 1997;39:185–191. 1142. Blennow G, Jaeken J, Wiklund L-M. Neurological findings in the carbo-
1116. Nikali K, Suomalainen A, Saharinan J, et al. Infantile onset spinocere- hydrate-deficient glycoprotein syndrome. Acta Paediatr Scand [Suppl]
bellar ataxiais caused by recessive mutations in mitochondrial proteins 1991;375:14–20.
twinkle and twinky. Hum Mol Genet 2005;14:2981–2990. 1143. Barone R, Pavone L, Fiumara A, et al. Developmental patterns and neu-
1117. Koskinen T, Valanne L, Ketonen L, et al. Infantile-onset spinocerebellar ropsychological assessment in patients with carbohydrate-deficient
ataxia: MR and CT findings. Am J Neuroradiol 1995;16:1427–1433. glycoconjugate syndrome type IA (phosphomannomutase deficiency).
1118. Conlan RV, Snead OC, Ceballos R. Olivopontocerebellar atrophy in chil- Brain Dev 1999;21:260–263.
dren: a report of seven cases in two families. Ann Neurol 1981;10:355–363. 1144. Miossec-Chauvet E, Mikaeloff Y, Heron D, et al. Neurological presenta-
1119. Bawle EV, Kupsky WJ, D’Amato CJ, et al. Familial infantile olivopontocer- tion in pediatric patients with congenital disorders of glycosylation type
ebellar atrophy. Pediatr Neurol 1995;13:14–18. Ia. Neuropediatrics 2003;34:1–6.
1120. Bertini E, des Portes V, Zanni G, et al. X-linked congenital ataxia: a clini- 1145. Kier G, Winchester BG, Clayton P. Carbohydrate deficient glycoprotein
cal and genetic study. Am J Med Genet 2000;92:53–56. syndromes: inborn errors of protein glycosylation. Ann Clin Biochem
1121. Rajab A, Mochida G, Hill A, et al. A novel form of pontocerebellar hyp- 1999;36:20–36.
oplasia maps to chromosome 7q11–21. Neurology 2003;60:1664–1667. 1146. Stibler H, Cederberg B. Diagnosis of the carbohydrate-deficient glyco-
1122. Hevner R. Progress on pontocerebellar hypoplasia. Acta Neuropathol protein syndrome by analysis of transferrin in filter blood spots. Acta
2007;114(4):401–402. Paediatr 1993;82:55–59.
1123. Barth P. Pontocerebellar hypoplasias: an overview of a group of inher- 1147. Barkovich AJ, Millen KJ, Dobyns WB. A developmental and genetic classifi-
ited neurodegenerative disorders with fetal onset. Brain Dev 1993;15: cation for midbrain-hindbrain malformations. Brain. 2009;132:3199–3230.
411–422. 1148. Drouin-Garraud V, Belgrand M, Grunewald S, et al. Neurological presen-
1124. Zelnik N, Dobyns WB, Forem SL, et al. Congenital pontocerebellar atro- tation of a congenital disorder of glycosylation CDG-Ia: implications for
phy in three patients: clinical radiologic and etiologic considerations. diagnosis and genetic counseling. Am J Med Genet 2001;101(1):46–49.
Neuroradiology 1996;38:684–687. 1149. de Lonlay P, Seta N, Barrot S, et al. A broad spectrum of clinical presenta-
1125. Muntoni F, Goodwin F, Sewry C, et al. Clinical spectrum and diagnostic tions in congenital disorders of glycosylation I: a series of 26 cases. J Med
difficulties of infantile pontocerebellar hypoplasia type 1. Neuropediatrics Genet 2001;38(1):14–19.
1999;30:243–248. 1150. Akaboshi S, Ohno K, Takeshita K. Neuroradiological findings in the carbo-
1126. Barth PG. The syndrome of autosomal recessive pontocerebellar hydrate-deficient glycoprotein syndrome. Neuroradiology 1995;37:491–495.
hypoplasia, microcephaly, and extrapyramidal dyskinesia (pontocer- 1151. Antoun H, Villeneuve N, Gelot A, et al. Cerebellar atrophy: an important
ebellar hypoplasia type 2): compiled data from 10 pedigrees. Neurology feature of carbohydrate deficient glycoprotein syndrome type 1. Pediatr
1995;45:311–317. Radiol 1999;29:194–198.
1127. Barth PG, Vrensen GF, Uylings HB, et al. Inherited syndrome of micro- 1152. Jensen PR, Hansen FJ, Skovby F. Cerebellar hypoplasia in children with
cephaly, dyskinesia, and pontocerebellar hypoplasia: a systemic atrophy the carbohydrate-deficient glycoprotein syndrome. Neuroradiology
wiht early onset. J Neurol Sci 1990;97:25–42. 1995;37:328–330.
1128. Budde BS, Namavar Y, Barth PG, et al. tRNA splicing endonuclease muta- 1153. Haas D, Kelley RI, Hoffmann GF. Inherited disorders of cholesterol bio-
tions cause pontocerebellar hypoplasia. Nat Genet 2008;40:1113–1118. synthesis. Neuropediatrics 2001;32:113–122.
1129. Ramaekers VT, Heimann G, Reul J, et al. Genetic disorders and cerebellar 1154. Hoffmann GF, Charpentier C, Mayatepek E, et al. Clinical and bio-
structural abnormalities in childhood. Brain 1997;120:1739–1751. chemical phenotype in 11 patients with mevalonic aciduria. Pediatrics
1130. Ramaekers VT. Cerebellar malformations. In: Klockgether T, ed. Handbook 1993;91:915–921.
of Ataxia Disorders. New York: Marcel Dekker, 2000:115–150. 1155. Bretón Martínez J, Cánovas Martínez A, Casaña Pérez S, et al. Mevalonic
1131. Uhl M, Pawlik H, Laubenberger Jr, et al. MR findings in pontocerebellar aciduria: report of two cases. J Inherit Metab Dis 2007;30:829.
hypoplasia. Pediatr Radiol 1998;28(7):547. 1156. Salonen R, Somer M, Haltia M, et al. Progressive encephalopathy with
1132. Barth PG, Aronica E, de Vries L, et al. Pontocerebellar hypoplasia type 2: edema, hypsarrhythmia, and optic atrophy (PEHO syndrome). Clin
a neuropathological update. Acta Neuropathol 2007;114:373–386. Genet 1991;39:287–293.
1133. Gardner RJM, Colemen LT, Mitchell LA, et al. Near total absence of the 1157. Vanhatalo S, Somer M, Barth P. Dutch patients with progresive enceph-
cerebellum. Neuropediatrics 2001;32:62–68. alopathy with edema, hypsarrhythmia, and optic atrophy (PEHO) syn-
1134. Grosso S, Mostadini R, Cioni M,et al. Pontocerebellar hypoplasia type 2. drome. Neuropediatrics 2002;33:100–104.
Further clinical characterization and evidence of positive response of 1158. Haltia M, Somer M. Infantile cerebello-optic atrophy. Neuropathology
dyskinesia to levodopa. J Neurol 2002;249:596–600. of the progressive encephalopathy syndrome with edema, hypsarrhyth-
1135. Patel MS, Becker LE, Toi A, et al. Severe, fetal-onset form of olivopon- mia and optic atrophy (the PEHO syndrome). Acta Neuropathol (Berl)
tocerebellar hypoplasia in three sibs: PCH type 5? Am J Med Genet A 1993;85:241–247.
2006;140A(6):594–603. 1159. Huisman TAGM, Klein A, Werner B, et al. Serial MR Imaging, diffusion
1136. Freeze HH. Update and perspectives on congenital disorders of glycosyla- tensor imaging, and MR spectroscopic findings in a child with progres-
tion. Glycobiology 2001;11(12):129R–143R. sive encephalopathy, edema, hypsarrhythmia, and optic atrophy (PEHO)
1137. Marquardt T, Denecke J. Congenital disorders of glycosylation: review of syndrome. Am J Neuroradiol 2006;27(7):1555–1558.
their molecular bases, clinical presentations and specific therapies. Eur J 1160. Somer M, Salonen O, Pihko H, et al. PEHO syndrome (progressive
Pediatr 2003;162:359–379. encephalopathy with edema, hypsarrhythmia and optic atrophy): neuro-
1138. Matthijs G, Schollen E, Pardon E, et al. Mutations in PMM2, a phos- radiologic findings. Am J Neuroradiol 1993;14:861–867.
phomannomutase gene on chromosome 16p13, in carbohydrate- 1161. Nagafuchi S, Yanagisawa H, Sato K, et al. Dentatorubral and pallidoluy-
deficient glycoprotein type I syndrome (Jaeken syndrome). Nat Genet sian atrophy: expansion of an unstable CAG trinucleotide on chromo-
1997;16:88–92. some 12p. Nat Genet 1994;6:14–18.

Barkovich_Chap03.indd 238 5/6/2011 12:09:36 AM


Chapter 3 • Metabolic, Toxic, and Inflammatory Brain Disorders 239

1162. Koide R, Ikeuchi T, Onodera O, et al. Unstable expansion of CAG repeat 1186. Galluzzi P, Filosomi G, Vallone I, Bardelli A, Venturi C. MRI of Wolfram
in hereditary dentatorubral-pallidoluysian atrophy (DRPLA). Nat Genet syndrome (DIDMOAD). Neuroradiology 1999;41:729–731.
1994;6:9–13. 1187. Marinesco G, Draganescu S, Vasiliu D. Nouvelle maladie familiale car-
1163. Burke JR, Wingfield MS, Lewis KE, et al. The Haw River syndrome: denta- actérisée par une cataracte congénitale et un arrêt du développement
torubropallidoluysian atrophy (DRPLA) in an African-American family. somato-neuro-physique. Encéphale 1931;26:97–109.
Nat Genet 1994;7:521–524. 1188. Sjögren T. Hereditary congenital spinocerebellar ataxia combined
1164. Maricich SM, Zoghbi HY. The cerebellum and hereditary ataxias. In: with congenital cataract and oligophrenia. Acta Psychiatr Scand
Swaiman KF, Ashwal S, Ferriero DM, eds. Pediatric Neurology, Principles 1947;46(Suppl):286–289.
and Practice, 4th ed. Philadelphia, PA: Mosby Elsevier, 2006:1241–1269. 1189. Lagier-Tourenne C, Tranebaerg L, Chaigne D, et al. Homozygosity map-
1165. Naito H, Oyanagi S. Familial myoclonus epilepsy and choreoathetosis: ping of Marinesco-Sjogren syndrome to 5q31. Eur J Hum Genet 2003;11:
hereditary dentatorubral-pallidoluysian atrophy. Neurology. 1982;32: 770–778.
798–807. 1190. Merlini L, Gooding R, Lochmuller H, et al. Genetic identity of
1166. Imamura A, Ito R, Tanaka S, et al. High intensity proton and T2 weighted Marinesco-Sjogren/myoglobinuria and CCFDN syndromes. Neurology
MR signals in the globus pallidus in juvenile type of dentatorubral and 2002;58:231–236.
pallidoluysian atrophy. Neuropediatrics 1994;25:234–237. 1191. Herva R, von Wendt L, von Wendt G, et al. A syndrome with juvenile
1167. Miyazaki M, Kato T, Hashimoto T, et al. MR of childhood onset dentato- cataract, cerebellar atrophy, mental retardation, and myopathy. Neurope-
rubral-pallidoluysian atrophy. Am J Neuroradiol 1995;16:1834–1836. diatrics 1987;18:164–169.
1168. Yamada M, Sato T, Tsuji S, et al. Oligodendrocytic polyglutamine pathology 1192. Alter M, Kennedy W. The Marinesco-Sjögren syndrome: hereditary cer-
in dentatorubral-pallidoluysian atrophy. Ann Neurol 2002;52:670–674. ebello-lental degeneration with mental retardation. Minn Med 1968;51:
1169. Prosch H, Grois N, Wnorowski M, et al. Long-term MR imaging course 901–906.
of neurodegenerative Langerhans cell histiocytosis. Am J Neuroradiol 1193. Georgy BA, Snow RD, Brogdon BG, et al. Neuroradiologic findings in
2007;28(6):1022–1028. Marinesco-Sjögren syndrome. Am J Neuroradiol 1998;19:281–283.
1170. Grois N, Barkovich AJ, Rosenau W, et al. Central nervous system disease 1194. Sasaki K, Suga K, Tsugawa S, et al. Muscle pathology in Marinesco-Sjögren
associated with Langerhans’ cell histiocytosis. Am J Ped Hematol/Oncol syndrome: a unique ultrastructural feature. Brain Dev 1996;18:64–67.
1993;15:245–254. 1195. Reinhold A, Scheer I, Lehmann R, et al. MR imaging features in
1171. Grois N, Prayer D, Prosch H, et al. Neuropathology of CNS disease in Marinesco-Sjogren syndrome: severe cerebellar atrophy is not an
Langerhans cell histiocytosis. Brain Pathol 2005;128:829–838. obligatory finding. Am J Neuroradiol 2003;24:825–828.
1172. Prayer D, Grois N, Prosch H, et al. MR imaging presentation of intracra- 1196. Illarioshkin SN, Tanaka H, Markova ED, et al. X-linked nonprogressive
nial disease associated with Langerhans cell histiocytosis. Am J Neurora- congenital cerebellar hypoplasia: clinical description and mapping to
diol 2004;25:880–891. chromosome Xq. Ann Neurol 1996;40:75–83.
1173. Martin-Duverneuil N, Idbaih A, Hoang-Xuan K, et al. MRI features 1197. Hoyeraal HM, Lamvik J, Moe PJ. Congenital hypoplastic thrombocy-
of neurodegenerative Langerhans cell histiocytosis. Eur Radiol topenia and cerebral malformations in two brothers. Acta Pediatr Scand
2006;16(9):2074–2082. 1970;59:185–191.
1174. Finsterer J. Ataxias with autosomal, X-chromosomal or maternal inheri- 1198. Hreidarsson S, Kristjansson K, Johannesson G, et al: A syndrome of
tance. Can J Neurol Sci 2009;36:409–428. progressive pancytopenia with microcephaly, cerebellar hypoplasia and
1175. Schöls L, Bauer P, Schmidt T, et al. Autosomal dominant cerebellar ataxias: growth failure. Acta Pediatr Scand 1985;77:773–775.
clinical features, genetics, and pathogenesis. Lancet Neurol 2004;3:291–304. 1199. Aalfs CM, van den Berg H, Barth PG, et al. The Høyeraal-Hreidarsson
1176. Perlman SL. Spinocerebellar degenerations: an update. Curr Neurol Neu- syndrome: fouth case of a separate entity with prenatal growth retarda-
rosci Rep 2002;2(4):331–341. tion, progressive pancytopenia and cerebellar hypoplasia. Eur J Pediatr
1177. Brusse E, Maat-Kievit JA, van Swieten JC. Diagnosis and management of 1995;154:304–308.
early- and late-onset cerebellar ataxia. Clin Genet 2007;71:12–24. 1200. Knight S, Heiss N, Vulliamy T, et al. Unexplained aplastic anaemia,
1178. Verrips A, Hoefsloot LH, Steenbergen GCH, et al. Clinical and molecular immunodeficiency, and cerebellar hypoplasia (Hoyeraal-Hreidarsson
genetic characteristics of patients with cerebrotendinous xanthomatosis. syndrome) due to mutations in the dyskeratosis congenita gene, DKC1.
Brain 2000;123:908–919. Br J Haematol 1999;107:335–339.
1179. Wolfram D, Wagener H. Diabetes mellitus and simple optic atro- 1201. Mahmood F, King M, Smyth O, et al. Familail cerebellar hypoplasia
phy among siblings: report of four cases. Proc Staff Meet Mayo Clin and pancytopenia without chromosomal breakages. Neuropediatrics
1938;13:715–718. 1998;29:302–306.
1180. Page M, Asmal A, Edwards C. Recessive inheritance of diabetes: the syn- 1202. Akaboshi S, Yoshimura M, Hara T, et al. A case of Høyeraal-Hreidarsson
drome of diabetes insipidus, diabetes mellitus, optic atrophy and deaf- syndrome: delayed myelination and hypoplasia of corpus callosum are
ness. Q J Med 1976;179:505–520. other important signs. Neuropediatrics 2000;31:141–144.
1181. Khanim F, Kirk J, Litif F, Barrett T. WFS1/wolframin mutations, Wolfram 1203. Pearson T, Curtis F, Al-Eyadhy A, et al. An intronic mutation in DKC1
syndrome, and associated diseases. Hum Mutat 2001;17:357–367. in an infant with Hoyeraal-Hreidarsson syndrome. (Letter). Am J Med
1182. El-Shanti H, Lidral A, Jarrah N, et al. Homozygosity mapping identifies Genet 2008;146A:2159–2161.
an additional locus for Wolfram syndrome on chromosome 4q. Am J 1204. Dokal I, Vulliamy T. Dyskeratosis congenital: its link to telomerase and
Hum Genet 2000;66:1229–1236. aplastic anemia. Blood Rev 2003;17:217–225.
1183. Shannon P, Becker L, Deck J. Evidence of widespread axonal pathology in 1205. Savage SA, Giri N, Baerlocher GM, et al. TINF2, a component of the shel-
Wolfram syndrome. Acta Neuropathol (Berl) 1999;98:304–308. terin telomere protection complex, is mutated in dyskeratosis congenita.
1184. Scolding NJ, Kellar-Wood HF, Shaw C, et al. Wolfram syndrome: heredi- Am J Hum Genet 2008;82:501–509.
tary diabetes mellitus with brainstem and optic atrophy. Ann Neurol 1206. Revesz T, Fletcher S, Al-Gazali LI, et al. bilateral retinopathy, aplastic
1996;39:352–360. anaemia, and central nervous system abnormalities: a new syndrome?
1185. Ito S, Sakakibara R, Hattori T. Wolfram syndrome presenting marked J Med Genet 1992;29(9):673–675.
brain MR imaging abnormalities with few neurologic abnormalities. Am 1207. Scheinfeld M, Lui Y, Kolb E, et al. The neuroradiological findings in a case
J Neuroradiol 2007;28:305–306. of Revesz syndrome. Pediatr Radiol 2007;37(11):1166–1170.

Barkovich_Chap03.indd 239 5/6/2011 12:09:36 AM


CHAPTER

Brain and Spine Injuries


4 in Infancy and Childhood
ERIN SIMON SCHWARTZ AND A. JAMES BARKOVICH

Introduction Neonatal hypoglycemia


Basic patterns of brain destruction Bilirubin encephalopathy (kernicterus)
Porencephaly Brain injury associated with congenital heart disease
Hydranencephaly Hypernatremic dehydration
Encephalomalacia CNS trauma in infancy and childhood
Hypoxic-ischemic brain injury Birth trauma
Localized infarctions Postnatal trauma
Diffuse ischemic or inflammatory brain injury Nonaccidental trauma (child abuse)
CNS injury in multiple pregnancies
Ischemic brain injury in multiple pregnancies

Before discussing specific destructive patterns, it is necessary to


INTRODUCTION understand how the developing brain reacts to injury. The immature
In this chapter, destructive processes of the brain and spine are brain responds to an insult in a different manner than the mature
discussed. This is an important group of disorders in that the injuries brain. The fetal brain has limited capacity for astrocytic reaction;
discussed in this chapter are responsible for more than 90% of cases therefore, necrotic tissue is completely reabsorbed (liquefaction necro-
of cerebral palsy (the other 9% is caused by malformations (1). In a sis), resulting is a smooth-walled, fluid-filled cavity (porencephaly,
strict sense, it is difficult to separate the brain injuries discussed in this also called a porencephalic cyst). In contrast, the mature brain reacts
chapter from many of those discussed in Chapter 3, as many metabolic to injury by significant astrocytic proliferation; the resulting lesion
and demyelinating disorders are, in fact, destroying brain cells while contains soft brain (“encephalomalacia”) consisting of astroglial cells
hypoxia, hypoglycemia, and hyperbilirubinemia might be considered and an irregular wall composed primarily of reactive astrocytes. The
metabolic disorders. However, inborn metabolic, toxic, and idiopathic neonatal and infant brains fall somewhere in between. The ability to
(autoimmune) disorders are usually progressive, whereas those result- mount an astrocytic response to injury seems to begin sometime dur-
ing from physical, hypoxic-ischemic, hypoglycemic, and hyperbiliru- ing the late second or early third trimester and progressively increases;
binemic trauma are usually static, resulting from one or two (usually) the neonatal brain has about 15% of the astrocytic response to injury
well-defined events. Therefore, separation of the disorders into two as the mature brain (2,3). Thus, the postinjury residual evolves from
chapters seems to make sense from a theoretical perspective and keeps (a) a pure cyst in the second trimester fetus to (b) a cyst with astroglial
the chapters at reasonable length. septae in the last months of gestation and the neonatal period to (c)
pure astrogliosis with no appreciable cystic component in the mature
brain.
BASIC PATTERNS OF BRAIN DESTRUCTION
Before considering specific patterns of brain injury from specific
causes, it is useful to consider the imaging appearances that result from
Porencephaly
severe diffuse brain injury in the neonate and infant. Diffuse insults to The term porencephaly has many different definitions. Pathologists
the brain, causing widespread destruction, result in different radiologic use the term to describe focal cavities with smooth walls and mini-
and pathologic appearances, depending upon the cause, the maturity mal surrounding glial reaction (2,4,5). Those cavities resulting from
of the brain at the time of insult, as well as the severity of the insult. focal brain destruction prior to approximately the 20th gestational
The imaging appearances seen in the acute and subacute stages after week are often lined by dysplastic gray matter and accompanied by
injury are discussed in subsequent sections. Specific end-stage patterns anomalies of the overlying cortex, usually polymicrogyria (5,6). Used
of tissue reaction to severe global injury are described by the terms in this sense, porencephaly is essentially synonymous with schizen-
porencephaly, multicystic encephalomalacia, and hydranencephaly. An cephaly, an anomaly that results from destruction of a portion of the
understanding of these terms will be useful in further discussions in germinal matrix and surrounding brain before the hemispheres are
this and subsequent chapters. fully formed (see Chapter 5). Other authors have designated lesions

240

Barkovich_Chap04.indd 240 5/25/2011 11:38:22 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 241

developing early in the second trimester as encephaloclastic poren- chapter on destructive disorders because the presence of remnants of
cephalies, smooth-walled cavities without any surrounding gliosis, in hemispheric tissue and reports of association with twin-twin transfu-
order to differentiate them from agenetic porencephalies and from sion (10) and congenital infection (11,12) clearly imply a destructive
cystic encephalomalacia (see discussion below), a condition charac- process. A similar condition has been induced in laboratory animals
terized by shaggy-walled cavities with considerable astroglial reaction by the occlusion of both carotid arteries in utero. In all likelihood,
and resulting from late gestational, perinatal, or postnatal injuries any of a number of diffuse insults to the developing brain, at a time
(2,7,8). when the brain reacts by liquefaction necrosis, can result in hydra-
On imaging studies, (encephaloclastic) porencephalies appear as nencephaly (2,12). Clinically, affected patients may be microcepha-
smooth-walled cavities that are isointense to CSF on all sequences, lic, normocephalic, or macrocephalic, depending upon the presence
including diffusivity maps (Fig. 4-1). The cavities have no internal and degree of associated hydrocephalus (13). Because of the nearly
structure, and the surrounding brain is of normal signal intensity. complete lack of cortical tissue, the children reach few developmental
Many regions of porencephaly (Fig. 4-2) are impossible to differen- milestones (13).
tiate from ex vacuo enlargement of the lateral ventricles secondary Imaging studies of patients with hydranencephaly show that the
to complete liquefaction and absorption of surrounding white mat- cerebral hemispheres are nearly completely replaced by CSF (2,14,15).
ter; indeed, the “cyst” may be completely integrated into the adjacent The thalami are usually preserved. The inferior medial aspects of the
ventricle. frontal lobes and the inferior medial aspects of the temporal and occip-
ital lobes may also be preserved (Fig. 4-3). The brainstem is usually
atrophic; the cerebellum is almost always normal.
Hydranencephaly
The diagnosis is best established by sonography or magnetic reso-
Hydranencephaly is a condition in which most of the brain mantle nance imaging (MRI), both of which will show the thin rim of cerebral
(cortical plate and hemispheric white matter) has been damaged, liq- tissue around dilated ventricles; MRI is probably best for fetal diag-
uefied, and resorbed (9); it can be considered as porencephaly of nearly nosis, as it is not subject to shadowing or reverberation artifacts (10).
the entire cerebrum. The cerebral hemispheres are largely replaced It is sometimes difficult to differentiate hydranencephaly from severe
by thin-walled sacs containing cerebrospinal fluid (CSF) (2,5). The hydrocephalus on computed tomography (CT) scans because the rim
membranes of the sacs are composed of an outer layer of leptomen- of tissue may be obscured by artifact generated by the overlying cal-
ingeal connective tissue and an inner layer of remnants of the cor- varium, particularly if a low radiation dose is used, as is common in
tex and white matter along with astrogliosis. Although some people pediatric studies. This distinction is significant because some hydro-
consider hydranencephaly a congenital anomaly, it is included in this cephalic children, even when the condition is extremely severe, respond

FIG. 4-1. Focal porencephaly. A. Axial T2-weighted image shows a sharply defined, homogeneously hyperintense cavity in
the posterior limb of the right internal capsule (black arrow). B. Axial Dav (average diffusivity) map at the same level shows very
high diffusivity, compatible with freely moving water molecules in a cyst.

Barkovich_Chap04.indd 241 5/25/2011 11:38:22 AM


242 Pediatric Neuroimaging

FIG. 4-2. Porencephaly secondary to presumed


venous infarction. A. Parasagittal transfontanelle
sonogram shows a large cavity (white arrows) extend-
ing from the left lateral ventricle (black arrows) into the
left frontal lobe. No internal structure is seen within
the cavity. B. Follow-up axial proton density image
shows the large cystic area (white arrows) in the left
frontal lobe at the site of prior hemorrhagic infarction.
The left basal ganglia are absent, having been destroyed
by the infarction. Diminished white matter and abnor-
mal hyperintensity (white arrowheads) is seen around
the ventricular trigones secondary to white matter
injury of prematurity. C. Coronal reconstruction of
3D-FLAIR image shows hypointensity within the cyst
(white arrows), the complete suppression of the con-
tents confirming its cystic nature. Note the difficulty
in differentiating this destructive lesion from ex vacuo
enlargement of the ventricle. Hyperintensity of the
contralateral white matter (black arrow) results from
white matter injury of prematurity.

well to CSF diversion procedures and develop normal cognitive and signal changes are poorly understood. The inherently poorer contrast
motor function if shunting is performed at an early age. In contrast, resolution of CT limits reliable differentiation of porencephaly from
children with hydranencephaly will not improve intellectually after cystic, and even noncystic, encephalomalacia (Fig. 4-4A). Ultrasound
shunting. In practice, this distinction may be purely academic because is the most sensitive modality in the detection of glial septa but is less
an abnormally enlarging head in infants with either entity dictates CSF useful in the overall evaluation of the brain.
diversion. Although CSF diversion will not improve development in Multicystic encephalomalacia results from a diffuse insult to the
infants with hydranencephaly, it will prevent the development of the brain late in gestation, during birth, or after birth (2,5). Multiple cys-
grotesquely enlarged head size that would otherwise result from the tic cavities of variable size, separated by glial septations, form in the
increased intracranial pressure. Maintenance of normal head size aids necrotic area (Figs. 4-4 to 4-7) (5). The location of the lesions varies
patient management. with the nature of the insult. If caused by thrombotic or embolic infarc-
tion (Figs. 4-4 and 4-6), the affected area will be in the distribution of
a branch of a major cerebral artery. In contrast, injury resulting from
Encephalomalacia
mild to moderate hypotension will tend to be located in the inter-
Encephalomalacia, in contradistinction to porencephaly and hydra- vascular boundary zones (see “watershed areas,” Fig. 4-5). When very
nencephaly, is characterized pathologically by astroglial proliferation severe, only the immediate periventricular white matter may be spared
and, often, septations within the area of damaged brain. MRI shows the (Fig. 4-7). Profound hypotension results in injury to the deep cerebral
reactive astrogliosis and tissue injury as areas of T1 hypointensity and nuclei and a variable amount of cortex; the precise distribution varies
T2 or FLAIR hyperintensity compared with myelinated gray and white with postconceptional age (16–18). Neonatal hypoglycemia causes dam-
matter (Fig. 4-4). In infants, images obtained in the subacute phase age in the parietal and occipital lobes (19,20). When injury is the result
after injury will often show T1 hyperintensity and T2 hypointensity of of an infection, the site of the encephalomalacia is nonspecific, reflecting
injured cortex and white matter (Fig. 4-5); the precise cause of these the region of the brain injured by the infection. Other than location,

Barkovich_Chap04.indd 242 5/25/2011 11:38:22 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 243

FIG. 4-3. Hydranencephaly. A. Sagittal T1-weighted


image shows absence of most of the cerebrum. Portions
of the inferomedial temporal and occipital lobes (white
arrows) are present. B. Axial T2-weighted image shows
complete absence of the cerebral hemispheres with the
exception of the thalami (small black arrowheads) and
some portions of the medial temporal lobes (black arrows)
and occipital lobes. The anterior falx cerebri (large black
arrowhead) is present, surrounded by a small amount of
residual frontal lobe. C. Axial T2-weighted image shows
presence of the falx (black arrows) at the level of the upper
cranial vault.

the imaging appearance does not characterize the cause. Ultrasound quite hyperintense in contrast to the CSF spaces, which are isointense
will show increased echogenicity between the second and fifth day after to the ventricular CSF. If the routine images are not definitive, diffusion
injury, with cystic degeneration developing between the 7th and 30th sequences can be used to determine diffusivity, which will be highest in
day after injury in term infants (21). The variation in the time to cavi- cystic regions, lower in microcystic regions, and still lower in noncystic
tation is presumably related to the severity of the injury, as well as the encephalomalacia; all damaged tissue will have higher diffusivity than
maturity of the brain. CT initially shows diffuse hypoattenuation in the normal brain.
affected regions of the brain; this appearance eventually evolves to one
of hypodense tissue containing cysts of various sizes (Fig. 4-8). Septa-
tions are often seen within the damaged region, and calcification may be
HYPOXIC-ISCHEMIC BRAIN INJURY
present (2). On MRI, the affected area appears as an ill-defined area of This section covers injuries of brain that are believed to be mediated by
T1 hypointensity and T2/FLAIR hyperintensity containing loculations hypoxia or ischemia. Much has been written to suggest that ultimately
of fluid (hypointense on T1, FLAIR, and diffusion-weighted images, the injury in many of these disorders, including localized infarctions,
hyperintense on T2 images and average diffusivity (Dav) images (Figs. is initiated by inflammatory factors. The authors do not dispute that
4-4, 4-6, and 4-7). Some heterogeneity is often present, as a result of the this may, indeed, be true in many cases. Nonetheless, hypoxia or isch-
combination of variably sized glial septae and CSF within the lesion. emia are likely involved in the brain injury at some level, whatever the
The heterogeneity is often most apparent on the FLAIR sequence or the initiating factors; therefore, hypoxia-ischemia seems to be a good title
first echo of the T2-weighted sequence, in which the glial strands are under which to organize these disorders.

Barkovich_Chap04.indd 243 5/25/2011 11:38:23 AM


244 Pediatric Neuroimaging

FIG. 4-4. Encephalomalacia in 12-year-old secondary to


remote MCA infarction. A. Axial noncontrast CT scan shows
heterogeneous hypodensity (white arrows) in the right posterior
temporal lobe, as well as some ex vacuo enlargement of the right
lateral ventricle. B. Coronal T1-weighted image shows better
detail of the encephalomalacic area (black arrows). Shrunken
hypointense gyri are seen separated by reduced volume of
less hypointense encephalomalacic white matter. C. Axial
T2-weighted image shows the area of encephalomalacia (white
arrows) as mostly hyperintense with residual, more normal, tis-
sue (small black arrows) as more hypointense. D. Coronal FLAIR
image shows hyperintense encephalomalacia. The central areas
of hypointensity (black arrows) are regions of fluid secondary
to cavitation.

fetal MRI has demonstrated that many infarcts occur before birth. The
Localized Infarctions
reason for this high incidence of strokes in fetuses and infants is not
Stroke is an important cause of morbidity and mortality in childhood. known. Despite these numbers, the misconception prevails that stroke
Indeed, it is one of the top ten causes of childhood death (22,23). is a rare and relatively unimportant illness in childhood. Fortunately,
Approximately 25% of all pediatric strokes occur in neonates, and the medical community has recently become more aware of this entity
approximately 50% occur in children less than 1 year of age (22,23); and its importance in pediatric health (22,24–33).

Barkovich_Chap04.indd 244 5/25/2011 11:38:24 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 245

FIG. 4-5. Encephalomalacia secondary to moderate hypotension. Axial SE T1 (A) and axial T2-weighted (B) images show cortical
thinning with abnormal T1 and T2 prolongation in the frontal watershed areas and volume loss with some T1 and T2 shortening in
the occipital lobes. T1 shortening is seen in the lateral thalami.

Manifestations and Causes of Infarctions in Children that the term ischemic perinatal stroke is now defined as “a group
Focal ischemic infarction has a variable clinical presentation in the of heterogeneous conditions in which there is a focal disruption
pediatric age group. Presenting signs and symptoms depend upon of cerebral blood flow secondary to arterial or cerebral venous
the region of brain affected and the age of the patient at the time of thrombosis or embolization between 20 weeks of fetal life through
the infarct (32,34–37). Perinatal/prenatal stroke is much more com- the 28th postnatal day, confirmed by neuroimaging or neuropatho-
mon than generally recognized, with a prevalence of 1 in 2300 to 5000 logic studies” (38). Risk factors for presumed perinatal ischemic
live births (38). Perinatal stroke has been deemed important enough stroke (PPIS) are listed in Table 4-1. When presenting acutely after

FIG. 4-6. Multicystic encephalomalacia secondary to arterial infarction. Axial SE T1 (A) and T2-weighted (B) images show a sharply
defined area of T1 and T2 prolongation in the left MCA distribution. Thin septae (arrowheads) are present within the cavity.

Barkovich_Chap04.indd 245 5/25/2011 11:38:25 AM


246 Pediatric Neuroimaging

least 1 prothrombotic risk factor compared with 44 control subjects


(24%): Factor V G1691A mutations, protein C Type I deficiency,
and elevated lipoprotein (a) levels were found more frequently in
cases than controls, while prothrombin and methylenetetrahydro-
folate reductase mutations were not significantly different between
the groups (50). In another cohort of 24 newborns with perinatal
cerebral infarction confirmed by neonatal MRI, 10 newborns (42%)
had at least 1 prothrombotic state, including heterozygosity for fac-
tor V Leiden in 5 and high factor VIIIc concentrations in 6 (51).
These prothrombotic states, in particular, the factor V Leiden muta-
tion, were associated with adverse neurodevelopmental outcomes
(51). In patients with no underlying metabolic disorders, trauma
and previous varicella zoster infection were significantly more com-
mon. A significant association was found between cerebral arterial
abnormalities and systolic blood pressure greater than 90th percen-
tile; a trend was identified for an association with varicella within
the previous year. Genetic or acquired conditions causing throm-
bophilia were rare causes of stroke in their series. In 79 children
with unilateral ischemic stroke, Braun et al. (52) found that arte-
riopathy, diagnosed by MR arteriography (MRA), CTA, or catheter
angiography, was most commonly stable after the initial inflamma-
tory period (which lasted for months). Outcome was much better
in patients in whom the arteriopathy stabilized or improved than in
FIG. 4-7. Severe macrocystic encephalomalacia. Coronal T1-weighted
image shows large cystic spaces of the cerebral cortex and white matter with those in whom it progressed (52). They postulate that this subset of
sparing of only a small region of periventricular white matter. Large cysts arteriopathies (those that eventually stabilize) are inflammatory in
are separated by septae. nature (indeed, many are seen within a year of varicella infections)
and propose the term transient cerebral arteriopathy to describe this
condition (52). In east Asian populations, the incidence of child-
hood brain infarction is similar to that in western populations, but
infarction, neonates and young infants typically manifest seizures, the underlying causes differ (31,53); congenital heart disease, CNS
decreased level of consciousness, hypotonia, irritability, lethargy, or infection, vascular diseases (particularly moyamoya), and hemato-
poor feeding (38); in this regard, it is important to remember that up logic disease (leukemia, thalassemia) are the most common predis-
to 5% of encephalopathic neonates, who were presumed on clinical posing causes (31,53), although metabolic causes are seen in up to
grounds to have a diffuse brain injury, may be symptomatic from 20% (31).
acute focal infarction (39). However, when neonatal infarctions are Long-term prognosis of children with focal infarctions and no
clinically cryptic (29,40) or if the infarct occurs in utero (41–45), underlying disorder varies with the location of the infarction and the
presentation may be one of early hand preference in infancy (infants age of the patient at the time of the injury (54). Poor motor outcome
do not usually show hand preference prior to the age of 1 year) or is associated with basal ganglia involvement and with periventricular
congenital hemiplegia; such infarcts are currently referred to as PPIS venous infarction (32). Seizures, poor cognition, and delayed develop-
(32). Indeed, it is estimated that approximately 30% of patients with ment are associated with cortical involvement (32,52). Patients with
congenital hemiplegia are the result of PPIS (38). Older infants and larger infarctions or infarctions involving eloquent regions of cortex
children who suffer strokes present in a manner similar to adults, obviously have larger residual deficits than those with smaller infarcts
with an abrupt onset of neurological deficit (31,34–36). Because the and those involving less eloquent regions (28,55). Among children
clinical presentation of stroke in neonates and infants can be quite with infarcts of similar size or location, younger patients typically
subtle, imaging is often the most revealing component of the diag- have smaller clinical deficits; in neonates, hemiparesis typically does
nostic work-up. not ensue unless the cortex, basal ganglia, and internal capsule are all
As many as 50% of focal brain infarctions in the pediatric popu- affected, whereas later in childhood, hemiparesis may ensue even if
lation are “idiopathic”; that is, the cause of the stroke is never found only one or two of those sites are affected (54). Compared with adults,
(46,47). In our experience and that of others (40), this number is prognosis is generally good unless epilepsy develops, because children
even higher in neonatal infarctions. Many strokes are associated with negative workups are unlikely to have another stroke and because
with coagulopathies or metabolic disorders. A study by Ganesan et the pediatric brain retains significant plasticity. Thus, in the absence of
al. (48) indicates that as many as 40% of pediatric stroke patients epilepsy, many functions normally performed by the injured regions of
in western populations are anemic; 21% either had elevated total brain will be taken over by other regions that have been spared (56).
plasma homocysteine or were homozygous for the methylene tetra- When epilepsy develops, however, cognitive recovery is impaired (56).
hydrofolate reductase mutation. Another study showed that 63% of Outcome in East Asian populations is similar to that in western series,
children with stroke had at least one prothrombotic risk factor and with death in 15% to 20%, persistent neurological deficits in 30% to
28% had at least two risk factors; plasminogen activator inhibitor, 40%, and normal outcome in 35% to 50% (31,53). Follow-up of a large
which forms a complex with tissue-type plasminogen activator to number of children with neonatal or infantile stroke from a study in
inhibit fibrinolysis, was the most common abnormality, seen in 27% Marseille showed no residual deficit in 28%, mild to moderate deficit
(49). In a multicenter case-control study of term newborns with in 48%, and need for significant long-term care (severe deficit) in only
symptomatic ischemic stroke, 62 of 91 stroke patients (68%) had at 24% (57). However, it appears that children (but not infants [54]) have

Barkovich_Chap04.indd 246 5/25/2011 11:38:26 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 247

FIG. 4-8. Encephalomalacia secondary to moderate hypoten-


sion. A. Contrast-enhanced CT image in the acute phase shows
low attenuation in the frontal and temporo-occipital (solid
arrows) white matter and the caudate heads (open arrows). B. In
the subacute phase (1 week after injury), the injured regions show
contrast enhancement. C. In the chronic phase (2 months after
injury), the cerebral cortex is shrunken away from the calvarium,
resulting in an increased amount of extraparenchymal fluid. The
ventricles have enlarged secondary to atrophy of white matter
and the caudate heads. Abnormal lucency (arrows) representing
encephalomalacia is present in the frontal and temporo-occipital
white matter.

a higher incidence of persistent dystonia after basal ganglia infarction Choice of Radiologic Study in Pediatric Stroke
(28,58) and a higher (25%–50%) incidence of epilepsy after ischemic The choice of radiologic study depends on the age of the patient. As
stroke (59) than adults. neonates generally present with seizures or nonspecific symptoms,
As alluded to earlier, disorders of coagulation may be identified ultrasound is the first study performed at most institutions. When
in neonatal strokes (48–51,60–66), including deficiencies of proteins properly performed with high frequency probes and when the studies
C and S (60–66), G20210A prothrombin mutations (66), and factor V are performed after day 4, ultrasound detects up to 87% of neonatal
Leiden (51,61,62), as well as the presence of anticardiolipin antibodies strokes involving the basal ganglia and large supratentorial vascu-
(63,67–69). Other causes are listed in Tables 4-2 and 4-3. lar territories (40). However, ultrasound is not very sensitive in the

Barkovich_Chap04.indd 247 5/25/2011 11:38:27 AM


248 Pediatric Neuroimaging

TABLE 4-1 Risk Factors for PPIS


Maternal factors/conditions Twin-to-twin transfusion syndrome
Thrombotic disorders Fetal/neonatal polycythemia
Infertility and infertility treatment Congenital heart disease
Preeclampsia Neonatal hypoglycemia (in preterm infants)
Prolonged rupture of membrane (>24 h) Persistent fetal circulation and extracorporeal membrane
Chorioamnionitis oxygenation therapy
Maternal autoimmune conditions and autoantibodies (platelet Intrauterine growth restriction
alloantigen-1) Fetal/neonatal infections and meningitis
Antiphospholipid syndrome Nonspecific factors
Fetal/neonatal disorders Ethnicity and race (higher incidence in black infants compared
Mutations in COL4A1 (gene for procollagen IVa1) with non-Hispanic white infants)
Inherited thrombophilia Infant gender (higher incidence in boys)

TABLE 4-2 Some Causes of Pediatric Stroke


Cardiac causes Infection (viral [80,83–86], bacterial meningitis [34], aspergillus
Cyanotic congenital heart disease [35,47] if immunosuppressed [87])
Cardiomyopathy [70] Coagulopathies (deficiencies of protein C or protein S [60–64],
Vascular dissection [70] factor V Leiden [61,62], methylene tetrahydrofolate reductase
Mitral valve prolapse [70] [88,89], lipoprotein (a) [88,89], or presence of antiphospho-
Cardiac tumors [71] lipid antibodies [63,67–69])
Thrombosis Maternal drug abuse (90,91)
Polycythemia [34] Migraine (47,92)
Trauma [72] Metabolic disorders (see Table 4-3)
Vasculopathies (sickle cell disease [73], moyamoya disease [74,75], Vascular malformations (35,72,93)
Kawasaki disease [72], PHACE syndrome [76], COL4A1 muta- CNS tumors by compression of vessels (35,72,93)
tions [45,77,78], postinfectious/inflammatory [52,79,80], Vasculitis (94–96)
fibromuscular disease [72], neurofibromatosis Type 1 [72],
radiation-induced [72], spontaneous dissection [81,82])

TABLE 4-3 Metabolic Causes of Stroke in Childhood


Organic acidurias Lysosomal storage disorders
Hyperhomocysteinemia Fabry disease
Propionic aciduria Cystinosis
Methylmalonic aciduria Urea cycle defects
Isovaleric acidemia Ornithine carbamyl transferase deficiency
Glutaric aciduria Type I Carbamyl phosphate synthetase deficiency
Glutaric aciduria Ty pe II Other
3-Methylcrotonyl-CoA carboxylase deficiency Progeria
3-Hydroxy-3-methylglutaryl-coenzyme A lyase deficiency Sulfite oxidase deficiency
Mitochondrial disorders Hyperlipoproteinemia
Mitochondrial myopathies Phosphoglycerate kinase deficiency
Leigh disease Congenital disorders of glycosylation
MELAS syndrome L-carnitine deficiency
Cytochrome oxidase deficiency Disorders of cholesterol and triglyceride metabolism
From (47,97–103) (see also Chapter 3).

Barkovich_Chap04.indd 248 5/25/2011 11:38:28 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 249

detection of small white matter infarctions, small cortical infarctions of marked hyperechogenicity within the echogenic zone should
over the cerebral convexities, or those in the posterior fossa. There- suggest hemorrhage. Cystic degeneration develops over 2 to 4 weeks
fore, we typically perform MRI with MRA or MR venography with associated ex vacuo enlargement of the ipsilateral ventricle
(MRV) to supplement the sonogram, as MRI will find both ischemic (114). Infarcts of the deep cerebral nuclei (Fig. 4-9) and larger corti-
and hemorrhagic lesions throughout the brain and allows excellent cal infarctions, such as those secondary to occlusion of the internal
assessment of both the anterior and posterior cerebral circulations. carotid artery or the main trunk of the middle cerebral artery (MCA)
Indeed, even if ultrasound finds an infarction, MRI often detects (Fig. 4-10), are more easily detected than smaller infarctions that are
additional parenchymal lesions and MRA/MRV can detect vascular restricted to the cerebral cortex or white matter (114). Posterior fossa
thromboses as well as many medium and large vessel vasculopathies infarctions are difficult to detect unless quite large; imaging through
(38, 104). Moreover, diffusion-weighted imaging increases the the posterolateral fontanelle improves sensitivity. The use of color
sensitivity of detecting infarction, and it allows detection of early Doppler and power Doppler sonography, which show changes in
Wallerian degeneration, manifested as reduced diffusion only a few regional cerebral blood flow after infarction (115), improve the sen-
days after the injury; when seen along the corticospinal tracts, this sitivity of sonography.
finding is highly predictive of subsequent hemiplegia (105). Finally, On CT, cerebral infarction in the neonate has a similar appearance
we generally avoid CT in infants (and children) because of the ion- to that in the older child or adult. When the infarction involves the
izing radiation and because venous thrombosis may be difficult to cerebral cortex, CT shows a well-defined, often wedge-shaped, region
detect on routine CT images (because of the high attenuation of of hypoattenuation that initially seems restricted to gray matter but
blood in scans of neonates and infants). Overall, MRI should be after the first 24 hours is seen in both the gray and white matter in
the study of choice in neonates and infants with suspected cerebral an arterial distribution (Fig. 4-11). Some caution must be used, how-
infarction. ever, as certain areas in the posterior temporal and occipital cortex
In older children, MRI with MRA/MRV is the first study of choice, normally have low attenuation on CT of infants and are difficult to
if available. As stated in the prior paragraph, we try to avoid CT in assess. Areas of hemorrhage are unequivocally detected as regions
infants and children. (Nevertheless, CT venography is very accurate that are hyperdense compared to normal parenchyma on noncontrast
in the detection of venous sinus thrombosis and should be performed scans. The administration of iodinated contrast is rarely necessary to
if an infant or child is suspected of having venous thrombosis, and establish the diagnosis. Contrast administration will result in enhance-
MRI is inconclusive, difficult to perform, or contraindicated [106].) ment of the infarcted region of cortex starting about 5 days after the
Catheter angiography is performed if the cause of the infarction is injury; enhancement will slowly disappear starting 4 to 6 weeks after
not diagnosed by laboratory or imaging studies, as it is more sen- the injury.
sitive than CT or MRI to the presence of vasculopathy in medium On MRI, the best way to identify infarctions in the first few
and small vessels (26). It is important to keep in mind, however, that days is by the use of diffusion imaging (Figs. 4-9 to 4-11), which
even high-resolution catheter angiography is often normal in small shows reduced diffusivity within hours of the infarction. The acute
vessel vasculitis, as in systemic lupus erythematosus. At some insti- infarct is seen as hyperintense signal on diffusion-weighted images
tutions, perfusion imaging is routinely performed in infants and (DWIs) and as hypointense signal on calculated diffusivity (also
children suspected of ischemic brain injury, whereas in others, it is called apparent diffusion coefficient, or ADC) images; these abnor-
reserved for patients with known vasculopathy, such as moyamoya malities are easily recognized and are identical to the appearance of
disease (see Chapter 12), usually when revascularization procedures acute infarction in adults. Diffusivity decreases within hours (116)
such as bypass or synangiosis are being considered. When perfusion and remains reduced for about 6 days before pseudonormalization
is assessed, susceptibility-weighted perfusion MRI (see Chapter 1) is occurs (117); diffusivity increases above normal around day 12 (118).
the study of choice. Note that none of these numbers are absolute; they vary somewhat
from individual to individual. Evaluation of white matter tracts dur-
Imaging Appearances of Arterial Infarctions ing the acute phase can show early evidence of Wallerian degenera-
in Children tion (manifested as reduced diffusivity, Fig. 4-10D) (105,119), which
Although the appearance of the infarct on imaging studies is indepen- may be predictive of persistent neurologic impairment (105,120).
dent of the cause of the infarct, some types of infarcts have predilec- Therefore, if available, diffusion images should be obtained in chil-
tions for certain regions of the brain. Knowledge of these predilections dren with an acute onset of unexplained neurologic deficits or infants
may aid in narrowing the differential diagnosis of the cause of infarc- with unexplained seizures. If the deficit is more than a 24 hours old,
tion. For example, posterior circulation infarctions are unusual in chil- the use of diffusion imaging is less critical as routine CT or MRI can
dren; an infarct in this location should raise suspicion for a traumatic identify the infarct; nonetheless, reduced diffusion in a vascular dis-
injury to the vertebrobasilar circulation (27,107,108) or, less likely, tribution increases confidence in the diagnosis. In contrast to CT,
vasospasm secondary to migraine (109), or even MELAS (if not in a the spin echo and FLAIR MR appearance of focal bland (nonhemor-
strict vascular distribution, see Chapter 3 [110,111]). Thalamic infarcts rhagic) infarction in a neonate is very different than in older children
typically occur in the setting of meningitis (infectious vasculitis), con- and can be quite subtle (Figs. 4-9 to 4-11). In newborns, the acutely
genital heart disease, migraine, or trauma (112). edematous, infarcted cortex becomes isointense with the underly-
Although cranial ultrasound is less sensitive than CT or MRI in ing unmyelinated white matter on routine spin-echo sequences. We
the detection of infarcts in neonates and infants (113), ultrasound look for infarcts by trying to identify the entire circumference of the
is usually the first brain imaging study performed, and knowledge hypointense cerebral cortex on T2-weighted images. If a segment of
of the findings is important. Cerebral infarction appears as an ill- the cortex appears to be “missing,” it probably represents focal infarc-
defined region of hyperechogenicity (Figs. 4-9 and 4-10) that slowly tion (the “missing cortex sign,” showed nicely in Figs. 4-10, 4-11). The
develops for several days after the event. Differentiation of hemor- internal capsules should also be carefully examined, as high signal
rhagic from bland infarction may be difficult; however, focal areas intensity of the posterior limb of the internal capsule on T2-weighted

Barkovich_Chap04.indd 249 5/25/2011 11:38:28 AM


250 Pediatric Neuroimaging

FIG. 4-9. Basal ganglia infarction in a neonate with congenital heart disease.
A. Transfontanelle sonogram shows increased echogenicity (arrows) in the left
corpus striatum. B. Axial T2-weighted image shows hyperintensity (arrows) in
the left caudate head and anterior left putamen. C. Axial diffusion-weighted
image (b = 700 s/mm2) shows hyperintensity, indicated reduced diffusion, in
the infarction.

images may be associated with the development of contralateral as is the MR appearance of hemorrhagic infarcts in children of any
hemiplegia (105). It should be noted that FLAIR images can be mis- age. If intravenous paramagnetic contrast is administered (this is not
leading in neonates/infants and will often not allow identification of the usually necessary), it results in an enhancement pattern (Fig. 4-12)
infarction (Fig. 4-11D). In the subacute phase (4–6 days after birth), and temporal evolution similar to that described in the preceding
infarcted gray matter may show high signal intensity on T1-weighted paragraph for CT.
images and low signal intensity on T2-weighted images (Fig. 4-12), Infarction of the deep gray matter nuclei in neonates and young
probably because of petechial hemorrhage, release of myelin lipids, infants is seen on sonography as subtle hyperechogenicity. Although
astroglial reaction (causing reduced amount of free water), or cal- it has been reported that deep infarcts do not become detectable by
cification. The MR appearance of infarctions in older children and sonography until the end of 1 or 2 weeks when the hyperechogenic-
adolescents is identical to the appearance of similar injuries in adults, ity becomes prominent (114), high-resolution sonograms will detect

Barkovich_Chap04.indd 250 5/25/2011 11:38:28 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 251

FIG. 4-10. MCA infarction. A. Sonogram in the coronal plane shows mild hyperecho-
genicity (white arrows) in the white matter in the MCA distribution. Note the slightly
increased echogenicity in the left basal ganglia and compression of the left frontal horn
(small black arrow), consistent with early infarction. B. Axial T2-weighted image shows
the infarct involving the entire MCA cortex (white arrows) that is hyperintense com-
pared with the remainder of the gray matter, and isointense with underlying white mat-
ter. The left caudate (black arrows) is also abnormally hyperintense. C. Axial diffusivity
map (b = 700 s/mm2) shows reduced diffusivity (hypointensity, white arrows) in the
infarcted MCA territory. Note the involvement of the basal ganglia, confirming what
was seen on the T2-weighted image (B). D. Axial diffusivity map (b=700 s/mm2) at the
level of the pons shows reduced diffusivity (black arrow) in the left pons, as a result of
Wallerian degeneration in the corticospinal tract. E. Collapsed image from TOF MR
arteriogram shows lack of flow-related enhancement in the middle portion of the left
MCA (arrow), a thrombus that is the cause of the infarct.

Barkovich_Chap04.indd 251 5/25/2011 11:38:29 AM


252 Pediatric Neuroimaging

FIG. 4-11. Missing cortex sign, utility of CT and FLAIR in cerebral infarctions of neonates. A. Axial CT scan shows hypoden-
sity (black arrowheads) in the left parietal lobe. B. Axial T2-weighted image shows absence of the normal cortical hypointensity
in the infracted area. A region of “missing cortex” suggests cortical injury in a neonate or infant. C. Calculated ADC image
shows reduced diffusion (hypointensity, white arrows) in the infarcted area. D. Coronal FLAIR image through the infarct shows
the subtlety of infarct detection with FLAIR imaging in neonates and infants. We do not use FLAIR in children during the first
year of life.

subtle changes (Figs. 4-9A and 4-10A) in the first few days after injury. The MR appearance of infarcted basal ganglia is variable, depending
Basal nuclei infarctions can be very subtle on CT, as well, particularly on the MR sequences used, the stage of the infarction, and the amount of
if the infarction is bilateral and associated with cerebral edema. This hemorrhage present. The infarction is readily identified as a hyperintense
appearance is discussed and illustrated later in the chapter in the sec- region in a vascular distribution on DWIs (Fig. 4-9C) or as a hypoin-
tion on “profound hypotension.” The point to be stressed is that isch- tense region on a calculated diffusivity (ADC) image (Fig. 4-13B). On
emic injury to the basal ganglia may be overlooked on ultrasound or CT spin-echo imaging, the acutely infarcted basal ganglia will appear nor-
(of neonates or older children, Fig. 4-13A) unless the radiologist specifically mal for the first 8 to 12 hours (Fig. 4-9B) unless hemorrhage is present
assesses the deep gray matter echogenicity or attenuation, respectively. (unusual in the acute phase). In subacute basal ganglia infarction, some

Barkovich_Chap04.indd 252 5/25/2011 11:38:30 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 253

FIG. 4-12. MCA infarction secondary to embolus.


A. Axial T1-weighted image shows hyperintensity in
the right lentiform nucleus (arrows) and poor differ-
entiation of the cerebral cortex from the underlying
white matter in the right MCA distribution. B. Axial
T2-weighted image shows hyperintensity of the cortex
in underlying white matter in the right MCA distribu-
tion. C. MR angiogram shows an embolus (arrow) in
the supraclinoid segment of the right internal carotid
artery. D. Axial T2-weighted image shows abnormal
hypointensity in the right lentiform nucleus and cere-
bral cortex in the MCA distribution. E. Postcontrast
T1-weighted image from follow-up scan performed
1 week after event. Marked enhancement is seen in
the injured regions of brain. (This case courtesy of
Dr. Chip Truwit, Minneapolis, MN.)

Barkovich_Chap04.indd 253 5/25/2011 11:38:31 AM


254 Pediatric Neuroimaging

FIG. 4-13. Infarct of putamen due to vasculitis. A. Axial noncontrast CT shows low attenuation of the left putamen
(white arrows). Compare to normal gray matter signal of right putamen.B. Axial Dav map shows reduced diffusivity (low inten-
sity, white arrows) in the affected putamen. C. Axial collapsed view of 3D TOF MR angiogram shows narrowing of the left MCA
(arrows). D. Axial postcontrast T1-weighted image shows enhancement (arrows) in the walls of the left MCA. Enhancement of
the wall of the blood vessel strongly suggests inflammatory vasculopathy. (This case courtesy of Dr. Susan Blaser, Toronto, ON.)

hemorrhage, calcium, or myelin degradation will typically be present in brain tissue can be saved by intervention (by looking at the extent of
the infarcted nuclei, resulting in a heterogeneous signal on both T1- and perfusion-diffusion mismatch). There is not yet any evidence that per-
T2-weighted images (Figs. 4-12). As with cortical infarctions, FLAIR is fusion imaging is useful in the assessment of infarctions in children.
often falsely negative in basal nuclear infarctions in the neonate. Moreover, both bolus administration and ASL are difficult in young
Evidence in adults suggests that perfusion imaging, either with infants and neonates. Therefore, perfusion imaging is not universally
a bolus of intravenous contrast or using arterial spin labeling (ASL, used in pediatric strokes at this time; it is routinely used at Children’s
see Chapter 1) may be of some value in determining how much Hospital of Philadelphia, but not at UCSF.

Barkovich_Chap04.indd 254 5/25/2011 11:38:32 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 255

Close scrutiny of the cerebral blood vessels is important when Parasagittal injuries, often hemorrhagic, are seen most commonly in
examining an MR scan of an infant or child who has suffered a cere- association with superior sagittal sinus thrombosis (Figs. 4-14 and
bral infarction. Therefore, some type of vascular imaging study should 4-15), temporal lobe hematomas are usually associated with transverse
be obtained in all infants and children with unexplained ischemic sinus thrombosis (and consequent vein of Labbé thrombosis, Fig. 4-16),
infarction (Figs. 4-10, 4-12, and 4-13). Although vascular injury or and thalamic hemorrhage is usually associated with vein of Galen/
inflammation can sometimes be detected on routine anatomic imag- straight sinus thrombosis (Fig. 4-17). Occluded venous sinuses are not
ing by the detection of vessel wall thickening or enhancement (see Fig. commonly seen in association with frontal polar hematomas; however,
4-13D) (104), vascular injury is better detected by MRA, which gives all frontal hematomas and parasagittal frontal infarcts should lead to
an excellent depiction of the intracranial carotid, vertebral, and basilar evaluation of the patency of the superior sagittal sinus (Fig. 4-18).
arteries and their proximal branches (Figs. 4-10E and 4-13C). Indeed, It is important to remember two facts when venous thrombosis is
between 64% and 74% of arterial defects in pediatric ischemic infarc- identified in neonates and children. The first is that all patients with
tions are located in the supraclinoid carotid artery or the proximal spontaneous venous thrombosis should be evaluated for disorders
(M1 segment) MCA (26). Therefore, if infarction is suspected clini- of coagulation, such as partial deficiencies of clotting factors (fac-
cally or on the basis of the anatomic images, MR angiography should tor V Leiden (61,62) or proteins C and S (60–64), high coagulation
be performed to carefully evaluate the blood vessels for evidence of factor VIII (121), G20210A prothrombin mutations (66), thermo-
dissection, thrombosis, stenosis, or dysplasia (the techniques for this labile methylenetetrahydrofolate reductase polymorphism (88,89),
are described in Chapter 1). In particular, dissection appears to be far increased lipoprotein A (88,89), and the presence of anticardiolipin
more common than previously suspected in childhood stroke (25,26). antibodies (63,67,68). The second is that venous thrombosis often
The appearance of dissection in children is not different from that in resolves in neonates without aggressive therapy and without neuro-
adults. It is suggested on MRI by the presence of a crescent of meth- logic residua (125–127). The best current estimates are that 77% of
emoglobin (bright on fat-suppressed T1-weighted images) in the wall affected neonates and 40% to 50% of affected infants and children are
of the lumen of affected vessels and on MRA by the presence of an inti- neurologically normal after a mean of 2.1 years (106,121). The role of
mal irregularity, a double lumen, a pseudoaneurysm, a tapering arterial intervention, such as intravascular thrombolysis, is not clear at this
occlusion, or an elongated area of tight, irregular, and tapering stenosis time; caution is advised in this regard, as nearly 70% of pediatric venous
or occlusion (26). Catheter angiography is no longer necessary unless infarcts are hemorrhagic (106).
subtle abnormalities of branch vessels or vasculitis of small-to-middle Duplex Doppler sonography is very useful for the diagnosis of
sized arteries is suspected; however, these locations are unusual ones sinovenous thrombosis in neonates and young infants. Echogenic clot
for arterial abnormalities in children (26). is seen in the affected sinus, and Doppler analysis shows absence of, or
As of this writing, there is no evidence to support the routine use alterations in, flow. In older infants and children, MRI is the imaging
of proton MR spectroscopy (MRS) in children with acute localized modality of choice. Routine T1- and T2-weighted spin-echo images
infarction. MRS is more useful in the evaluation of patients with dif- should be obtained (and FLAIR in children older than 1 year), in addi-
fuse ischemic injury, to be discussed later in this chapter. tion to diffusion-weighted images, gradient-recalled echo images, and
MRV. If the venous thrombosis is acute (<7 days old), it will exhibit
Infarction Secondary to Venous Occlusion marked hypointensity with apparent expansion of the affected sinus
Venous infarcts secondary to venous thrombosis are fairly common, (due to susceptibility effects) on gradient-recalled echo images with
both prenatally and in childhood (106). At present, best estimates are long echo time (TE = 25–30 milliseconds) (128). Often, the sagittal
that the incidence of venous thrombosis is 0.67 cases per 100,000 chil- T1-weighted images are diagnostic by themselves, as they show sub-
dren per year (106). Of these, up to 40% suffer venous infarction, of acute clot (high signal intensity) in the sagittal, straight, or transverse
which about 70% are hemorrhagic (106). In addition, a significant sinuses (Fig. 4-14); this appearance is diagnostic. CT may also be
number of cases of congenital hemiplegia appear to result from pre- used; indeed, CT venography is the most sensitive study for the detec-
natal periventricular venous infarctions (41,42). Venous thrombosis tion and diagnosis of venous thrombosis (129), showing thrombi as
should be suspected in any child who, in the absence of trauma or hypodense areas within the affected venous structure. However, subtle
infection, has an unexplained hemorrhage or a brain injury that does brain injury is more difficult to detect on CT, especially in neonates
not fit an arterial vascular distribution (in particular, anterior tempo- and young infants. On CT, venous infarcts are usually poorly delim-
ral lobe hemorrhage, frontal or parietal involvement, and absence of ited, hypodense or mixed attenuation areas involving the subcortical
caudate involvement favor venous infarction [121,122]). If family his- white matter and producing a slight mass effect (Fig. 4-15A). The low
tory is present, consider hemorrhage due to Collagen IV A1 (COL4A1) attenuation is probably due to localized cerebral edema, whereas high
mutation (77), which looks identical to hemorrhagic venous infarc- attenuation areas usually represent hemorrhage. Following contrast
tion on MRI (45,78). Hemorrhage due to COL4A1 mutations can administration, linear or round gyral enhancement frequently over-
range from mild to very severe and, in the most severe fetal cases, lies the hypodensity. The thrombosed vein may be seen overlying the
can even cause hydranencephaly (Dr. Linda de Vries, personal com- infarction as a curvilinear region of high attenuation. On MRI, early
munication). Whatever the cause, affected infants commonly present venous infarcts may be identified by visualization of prolonged T1 and
with seizures and diffuse neurologic signs, whereas older children T2 relaxation times in characteristic regions (most commonly fron-
more commonly manifest alteration of consciousness, headache, or toparietal parasagittal [Figs. 4-14 and 4-15] and temporal [Fig. 4-16]).
focal neurologic signs (106). Risk factors are age-dependent: peri- Another early imaging sign is visualization of thrombus in the deep
natal complications in neonates, head and neck infections (otitis medullary veins (Fig. 4-17); this usually indicates thrombosis of both
media, mastoiditis, or sinusitis) in preschool children, and trauma or the superficial and deep venous systems. Of note, diffusion images may
chronic diseases such as connective tissue disorders in older children show reduced, normal, increased, or a mixture of diffusivity in areas of
(106,121,123). venous infarction (130–132). This probably results mainly from the
As will be discussed in a later section of this chapter, venous fact that venous sinus occlusion initially reduces venous outflow, with
thrombosis is a common cause of spontaneous cerebral hemorrhage consequent interstitial (“vasogenic”) edema, which causes increased
in neonates, both intraparenchymal (30) and, rarely, epidural (124). diffusion. If adequate collateral venous outflow is not established,

Barkovich_Chap04.indd 255 5/25/2011 11:38:33 AM


256 Pediatric Neuroimaging

FIG. 4-14. Venous infarction secondary to superior sagittal sinus thrombosis. A. Axial noncontrast CT scan shows
mixed high and low attenuation in the left frontal lobe. Note the high attenuation (arrows) in the superior sagittal sinus.
B. Sagittal SE 550/16 image shows hyperintensity (large arrows) in the superior sagittal sinus, indicating methemoglobin and
subacute thrombus. C and D. Axial T1- and T2-weighted images show a mixture of hemorrhage (bright in C, dark in D) and
edema (dark in C, bright in D) characteristic of venous infarction.

frank venous infarction will occur. The net diffusion characteristics as our first choice for making the diagnosis in infants because of the
of the region are roughly additive; thus, the diffusion characteristics lack of ionizing radiation; MRV with contrast is useful to better evalu-
will depend upon the degree of increased water motion resulting from ate areas where flow-related enhancement is diminished due to rapid
interstitial edema and the degree of decreased water motion secondary flow or flow is parallel to the imaging plane on two-dimensional (2D)
to the infarct. The frequent presence of blood (Fig. 4-18), the para- time-of-flight (TOF) sequences. Venous sinus thrombosis is discussed
magnetic effects of which makes diffusivity values unreliable, may also further in the section on this chapter on “Venous Thrombosis/Venous
contribute to this heterogeneity of diffusion characteristics. Many (up Infarction” and in Chapter 11, in the section on “Complications of
to 70%) venous infarcts are hemorrhagic; hemorrhagic infarcts have Meningitis.”
an imaging appearance that varies from large subcortical hematomas
(Fig. 4-18) to petechial hemorrhages within edematous brain paren-
Diffuse Ischemic or Inflammatory Brain Injury
chyma (Figs. 4-14 and 4-16) (106,133). The hemorrhages are generally
subcortical and often multifocal with irregular margins. They are occa- Diffuse brain injury has many causes in neonates and children, includ-
sionally linear in nature, indicating hematoma in and around the vein; ing metabolic disease, complications of prematurity, complications of
this appearance is quite specific. the birth process, and infection. This section will cover disorders that
Finally, although CT venography is the most definitive method are generally considered to be ischemic or inflammatory (white matter
for establishing the diagnosis of venous thrombosis (129), we use MRI injury of prematurity might be either or both). A number of terms are

Barkovich_Chap04.indd 256 5/25/2011 11:38:33 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 257

FIG. 4-15. Extensive sinus thrombosis in a child with seizures. A. Axial noncontrast CT scan shows areas of low attenuation (white
arrows) in the right frontal and left parietal parasagittal white matter. B. Coronal FLAIR image shows abnormal cortical hyperintensity
(white arrows), representing ischemic injury in the parietal cerebral convexities. C. Axial diffusivity map shows low signal intensity
(white arrows) representing reduced diffusivity in the parasagittal regions of the hemispheres. D. Anteroposterior maximum intensity
projection of 2D TOF MR venogram shows near complete absence of flow related enhancement in the major dural venous sinuses. Only
the right transverse (white arrow) and sigmoid sinuses and the right jugular vein (arrowheads) are patent. Sagittal sinuses, straight sinus,
vein of Galen, and left transverse/sigmoid sinuses are occluded.

Barkovich_Chap04.indd 257 5/25/2011 11:38:34 AM


258 Pediatric Neuroimaging

FIG. 4-16. Temporal lobe hemorrhagic venous infarct sec-


ondary to transverse sinus thrombosis. A. Axial noncontrast
CT shows hypoattenuation in the anterior right temporal lobe
(white arrowheads) and hyperattenuation (white arrow) in the
right sigmoid sinus, suggesting venous infarction secondary to
sigmoid sinus thrombosis. B. Axial T2-weighted image shows
areas of hypointensity (white arrows), suggesting hemorrhage,
in the infarction. C. Postcontrast axial T1-weighted image
shows slow flow and filling defects in the right transverse sinus
(white arrows), confirming thrombosis. Compare with normal
signal void in left transverse sinus (black arrows).

used to describe diffuse ischemic brain injury in the neonate, including Such factors should be actively investigated unless obvious causes such
perinatal asphyxia, neonatal encephalopathy, hypoxic-ischemic enceph- as umbilical cord or uterine rupture, large placental abruption, or car-
alopathy, and asphyxia neonatorum. The causes of this type of injury diocirculatory arrest are found.
have been widely debated. Although some authors have postulated that The physiology of brain injury resulting from hypoxia and isch-
obstetric factors are the most important (134,135), many others have emia is rather complex and not completely understood. Asphyxia,
questioned this assumption (136) and have suggested the importance defined as impaired exchange of oxygen and carbon dioxide, results in
of prenatal and perinatal infection (137–141), placental pathology diminished oxygen content in the blood (hypoxia), increased carbon
(142), or underlying metabolic disorders (143), among other factors. dioxide in the blood (hypercarbia), acidosis, and decreased systemic

Barkovich_Chap04.indd 258 5/25/2011 11:38:36 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 259

FIG. 4-17. Total deep and superficial venous sinus thrombosis. A. Sagittal T1-weighted image shows hyperintensity consistent with subacute blood
(methemoglobin) in the superior sagittal sinus (large white arrows) and vein of Galen/straight sinus (small white arrows). B and C. Axial T2-weighted
images show hypointensity extending radially from the ventricular walls (small arrows) suggestive of thrombus in the deep medullary veins. D. Phase-
contrast MR venogram shows absence of moving protons (no flow) in the superior sagittal sinus and straight sinus.

blood pressure. The hypercarbia and hypoxia cause a loss of the normal and in germinal matrix hemorrhage, or white matter injury in preterm
vascular autoregulation in the brain of the term neonate (144,145), infants (147,149–151). The newborn brain is quite resistant to hypoxia
resulting in so-called pressure-passive flow. (In the noncompromised in the setting of normal cerebral blood flow because glucose and other
term neonate, blood vessels of the brain constrict when blood pres- energy substrates, notably ketone bodies, minimize or prevent brain
sure increases and dilate when blood pressure decreases. This process, damage (152). Thus, most hypoxic-ischemic brain injury in neonates is a
known as autoregulation, maintains constant cerebral blood flow. Loss result of cerebral hypoperfusion.
of autoregulation results in pressure passive flow. In preterm neonates, The duration of the hypoperfusion is a critical factor in determin-
pressure passive flow is present even in the absence of asphyxia or ing whether brain damage results from an episode of hypoperfusion
other disease [145].) The combination of decreased blood pressure (17). Neonatologists and obstetricians have found that babies suffering
and pressure-passive flow results in decreased perfusion of the brain short periods of hypotension suffer no long-term effects. In support
(146,147) and may result in ischemic injury in term infants (probably of this, our anecdotal experience is that neonates and children who
mediated by excitotoxicity (148), discussed in subsequent sections), have been quickly resuscitated after respiratory or cardiac arrests show

Barkovich_Chap04.indd 259 5/25/2011 11:38:36 AM


260 Pediatric Neuroimaging

FIG. 4-18. Obtunded neonate with blown pupil. A. Noncontrast CT shows large left frontal hemorrhage (white arrows) with
left to right subfalcine herniation. Coronal sutures are wide (white arrowheads) and there is a suggestion of subdural blood
(black arrowheads) over the left convexity. B. Axial T2-weighted image again shows large left frontal hematoma and confirms
subdural hemorrhage (white arrows). C. DWI shows the complexity of hemorrhagic venous infarcts on diffusion imaging. Sus-
ceptibility effect of acute or subacute blood can mask diffusion characteristics of water molecules. D. Collapsed view of 2D TOF
MR venogram shows no flow related enhancement of sagittal or straight sinuses.

no abnormalities on their subsequent imaging studies. Animal models our anecdotal experience suggests that longer arrests, in the range of
support this, as well. In asphyxiated neonatal sheep, selective neuronal 15 to 25 minutes, result in brain injury. The amount of time to brain
necrosis is seen only after 10 minutes of perfusion arrest (153,154), and injury from less severe hypotension is more variable, depending upon
in monkeys damage is detected only after 7 minutes of perfusion arrest the amount of energy substrate reaching the cells (158).
(155), with basal ganglia injury seen after 8 to 12 minutes and severe Although hypoperfusion is the primary cause of neonatal asphyxic
injury of the entire brain after 20 minutes (156,157). Although it is brain injury, hypoxia does have a role. In addition to its effect on auto-
difficult, in most cases, to know the precise time of onset of an arrest, regulation, hypoxia reduces cardiac contractility/cardiac output and

Barkovich_Chap04.indd 260 5/25/2011 11:38:38 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 261

FIG. 4-19. Secondary energy failure. During hypoxia-ischemia,


significant changes occur in the brain on magnetic resonance
studies. 31P spectroscopy shows depletion of phosphocreatine or
NTP (including ATP), proton spectroscopy shows elevated lactate,
and diffusion-weighted imaging shows reduced diffusivity. These
diffusion and spectroscopy findings may normalize in the early
recovery period, only to revert and become abnormal again after
about 24 hours. Secondary energy failure is postulated to result
from structural damage to intracellular molecular structures that
are important to cellular metabolism, particularly high energy
phosphate reserves. Structural damage accumulates during the
“early recovery period” and, as this accumulation takes place,
cellular metabolism deteriorates. Don’t be fooled during latency
period into thinking that there has been no brain damage!

alters capillary permeability. Reperfusion of the weakened capillaries flow, shunting of blood is no longer adequate to save the deep struc-
(particularly those within the germinal matrices of premature infants, tures from damage.
which are structurally weaker than the capillaries in the remainder of In profound hypotension, the location of injury is initially to
the premature brain [159]) can cause rupture of cerebral blood vessels the deep cerebral nuclei (thalami and basal ganglia), posterior brain
and result in intracranial or intraventricular hemorrhage (151,160). stem, and the most active regions of the cerebral cortex (the senso-
rimotor region). Damage to the remainder of the cortex and white
Secondary Energy Failure matter occurs only later in the course of the hypotensive episode
A useful concept in understanding imaging results after hypoxic- (16,166,170,173,174,177–179). The location of injury is probably
ischemic injury is that of secondary energy failure (Fig. 4-19). During related to the state of maturity of the brain (17,148,180,181). Positron
hypoxia-ischemia, significant changes occur in the brain on magnetic emission tomography studies of glucose uptake in the developing brain
resonance studies. 31P spectroscopy shows depletion of phosphocre- (182), which reflect metabolic activity; SPECT studies, which show rela-
atine or NTP (nucleotide triphosphates, including ATP) (161), pro- tive regional perfusion (183,184); and proton spectroscopy (185), which
ton spectroscopy shows elevated lactate (162), and diffusion-weighted reflects biochemical maturation, all show excellent temporal and topo-
imaging shows reduced diffusivity (163,164). These findings then logical correlation with the process of myelination (Fig. 4-21) (186). In
normalize in the early recovery period (6–18 hours), only to revert to addition, the regions affected in profound hypotension are connected
abnormality again around 24 hours after the injury. This reversion to by circuits that use glutamate as their neurotransmitter (187). Imma-
abnormality has been called secondary energy failure (162). It is pos- ture NMDA receptors have heightened excitability and can be opened
tulated to be the result of structural damage to intracellular molecular in hypoxia-ischemia. This, combined with impaired removal of gluta-
structures that are important to cellular metabolism, particularly high- mate by energy-dependent transporters, is an important cause of cell
energy phosphate reserves; the structural damage accumulates during injury in severe hypoxia-ischemia (148). As these areas have the most
the “early recovery period” and, as this accumulation takes place, cel- advanced myelination, highest perfusion, increased synaptic plasticity,
lular metabolism deteriorates (165). and greatest glucose uptake, they would seem to be at highest risk for
injury in the absence of glucose and oxygen (which provide energy to
Patterns of Diffuse Hypoxic-Ischemic Brain Injury the cell). Indeed, the pattern of brain injury detected on MR studies of
A number of different patterns of brain injury can be seen as a result of newborns who have suffered profound hypotension corresponds quite
hypoxic-ischemic episodes in neonates, infants, and children (16,166– closely to the patterns of myelination, perfusion, and glucose uptake at
174). These patterns can be best understood if interpreted as being the the time of injury (16,17,170,180–182). These correlations suggest that
result of three primary factors: (a) severity of hypotension, (b) matu- the most metabolically active and mature regions of the brain, those
rity of the brain at the time of injury, and (c) duration of the event. that have the most advanced myelination, biochemical composition,
perfusion, and glucose uptake, are the regions that suffer damage first
Severity of Hypotension in the setting of a nearly complete cessation of brain perfusion.
When blood flow to the brain is mildly or moderately reduced (mild to
moderate cerebral hypotension with impaired autoregulation), blood Maturity of the Brain
flow is shunted from the anterior to the posterior circulation in order The patterns of injury secondary to mild to moderate hypotension and
to maintain adequate perfusion to the brain stem, cerebellum, and those secondary to profound hypotension both change with the post-
basal ganglia (175). As a result, damage is limited to the cerebral cortex conceptional age of the child.
and the intervascular boundary zones (both cortex and white matter Premature infants who suffer mild to moderate hypotension typi-
[176]) of the cerebral hemispheres (Fig. 4-20). However, when reduc- cally sustain injury to the periventricular and deep white matter with
tion of cerebral blood flow is severe (profound cerebral hypotension), sparing of the subcortical white matter and cerebral cortex. In contrast,
resulting in complete or nearly complete cessation of cerebral blood term infants who suffer similar degrees of hypotension sustain injury

Barkovich_Chap04.indd 261 5/25/2011 11:38:38 AM


262 Pediatric Neuroimaging

FIG. 4-20. Intervascular boundary zones. Axial SE T1-weighted images (A and B) show the intervascular boundary zones between
the ACA distribution and MCA distribution anteriorly and between the PCA distribution and the MCA posteriorly.

in the watershed portions of the cerebral cortex and in the underlying is compromised in prematurely born neonates. White matter damage
subcortical and periventricular white matter. Classically, this change in of prematurity (sometimes called periventricular leukomalacia) is,
injury pattern has been attributed to a changing location of the inter- therefore, a fairly common brain injury in prematurely born children.
vascular boundary zones (“watershed regions”). Van den Bergh (188), Three different types of white matter injury are described by Volpe
Takashima and Tanaka (189), and deReuck (190) postulated that the (147): diffuse injury, which has the mildest clinical manifestations;
ventriculofugal blood vessels in the brain (those coursing outward into focal/multifocal noncavitary injury, which has intermediated clinical
the cerebrum from the intraventricular and periventricular regions) are severity; and focal/multifocal cavitary injury, which has severe clinical
poorly developed during the first two trimesters of gestation. Almost manifestations. All three types can be identified and distinguished by
all of the blood supply to the cerebral white matter and cortex, there- MRI and are described in the next section, on imaging of brain injury
fore, was postulated to come from the ventriculopetal blood vessels in premature neonates. White matter injury is particularly common
coursing inward from the surface of the brain. This explanation, and in those with respiratory distress syndrome, antepartum placental
the existence of ventriculofugal arteries, has been disproved (191–193). abruptions, hypocapnia, twin pregnancy, septicemia, or ischemia dur-
In addition, new information better explains the observed findings. ing delivery (167,168,172,199,200). In contrast to the premature brain,
The cerebral vessels in the premature infant have a relatively limited the term neonate (38–42 postconceptional weeks) and postterm neo-
vasodilatory capacity, resulting in an inability of the brain to accept nate (43–46 postconceptional weeks) have normal GluR2 expression in
increased blood flow; this limitation in vasodilatory capacity exacer- the oligodendrocytes, but reduced GluR2 expression in the neocortex
bates the ischemia. Superimposed upon the reduction of blood flow is (196); thus, the cortex becomes selectively vulnerable to mild-moder-
the fact that the periventricular white matter of a premature infant is ate ischemia as the neonate approaches term. Thus, due to matura-
a site of oligodendrocyte proliferation in preparation for myelination. tion of the brain, and (likely) its vascular system, the pattern of injury
The developing cerebral white matter in this region responds to oxy- begins to change between the 34th and 38th postconceptional weeks, as
gen deprivation by utilizing anaerobic glycolysis, an inefficient mecha- the regions at highest risk for injury extend peripherally to include the
nism of glycogen metabolism that results in depletion of high-energy subcortical white matter and cerebral cortex in the interarterial bound-
phosphates and in localized lactic acidosis. Late oligodendrocyte pro- ary zones; note that white matter involvement frequently still occurs
genitors are very susceptible to lactic acidosis from hypoxia-ischemia and this can be seen on good quality imaging studies (176). These so-
(194), probably because their AMPA glutamate receptors lack GluR2 called watershed areas are almost always involved if mild to moder-
subunits, allowing them to flux high amounts of calcium (toxic to ate hypotension of sufficient duration takes place after 36 weeks (18).
the cells [195,196]); in addition, they do not have proper enzymes to In some patients, the damage extends medially and laterally from the
detoxify nitric oxide and other oxygen free radicals (197). The time at watershed zones to involve larger areas of the cerebrum.
which late oligodendrocyte progenitors are present in the brain coincides Pontosubicular necrosis, in which the subiculum and fascia den-
with the period of vulnerability of periventricular white matter injury in tata of the hippocampus and ventral pons are selectively involved (5) is
the fetal brain (198). Thus, as a result of the state of immaturity of the seen almost exclusively in premature infants, typically in the setting of
premature brain and its vascular supply, the periventricular and deep hypocarbia and hyperoxemia (201–203). Although the precise cause is
white matter areas are the regions at highest risk when autoregulation not known, the fact that pontosubicular necrosis is almost always seen in

Barkovich_Chap04.indd 262 5/25/2011 11:38:39 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 263

FIG. 4-21. 18FDG PET scans showing changing glucose uptake with maturity. A. Highest glucose metabolism at birth is in the
posterior fossa, thalami (arrows), and sensorimotor regions of the cerebrum. B. Proton magnetic resonance spectroscopic image of
a neonate shows that the highest NAA concentration is in the thalami (black arrows), confirming that this is the earliest part of the
cerebrum to mature. Note similarity to (A). C. By age 3 months, the basal ganglia (black arrowheads) have similar metabolism to the
thalami and the visual cortex (white arrows) has much greater activity. D. By age 8 months, the cerebral cortex metabolism is the
highest in the brain, greater than that in the thalami and similar to that of the basal ganglia. This relative activity will be similar at
subsequent ages. (A and D are courtesy of Prof. Harry Chugani, Detroit.)

the setting of periventricular leukomalacia added to the fact that the fascia The pattern of injury from profound hypotension evolves as brain
dentata seems to undergo apoptotic cell death (204) suggests that the regions mature, because the increased metabolic demand combined
hypocarbia and hyperoxemia exacerbates cellular injury from hypoxia- with the stage of maturity of the glutamate receptors changes the pat-
ischemia in the pons and hippocampus in the premature infant. terns of vulnerability of the brain (16). The process of maturity involves

Barkovich_Chap04.indd 263 5/25/2011 11:38:40 AM


264 Pediatric Neuroimaging

changes in neurotransmitters used as well as the maturity of the Although there are many conditions that result in brain injury in
receptors (148,196), in local glucose uptake (182), in relative regional the premature neonate, the two most widely accepted pathogenetic
blood flow (183,184), in levels of metabolites (185), and in myelina- mechanisms are maternal-fetal infection and impaired cerebrovascular
tion (170,181). The thalami and brainstem have the highest metabolic autoregulation with resultant ischemia (220). Definitive proof for either
activity in the early third trimester; from the middle of the third tri- mechanism is lacking and a full discussion of the controversy as to which
mester through 40 postconceptional weeks, the brain stem, thalami, mechanism predominates is beyond the scope of this book. However, cur-
basal ganglia, and perirolandic region have the highest metabolic activ- rent work seems to support the ischemia hypothesis (148,196,220,221). For
ity (16,17,182,205). By the end of the first postnatal month (44 post- all of the reasons discussed in the previous section, ischemic injury tends
conceptional weeks), the visual cortex becomes more metabolically to occur in different regions of the brain in preterm infants than in term
active (Fig. 4-21) (182). After the third or fourth postnatal month, the infants. Autoregulation is often absent in premature neonates (145), and,
remainder of the cerebral cortex and the basal ganglia become increas- because of the relatively high incidence of lung immaturity (with resul-
ingly metabolically active (Fig. 4-21D) and the regions more likely to tant hypoxia), patent ducti arteriosi, and sepsis, mild cerebral hypoperfu-
be damaged shift from the thalami and perirolandic cortex to the basal sion is relatively common. Because of the presence of late oligodendrocyte
ganglia and the entire cerebral cortex (170). progenitors, which are very susceptible to lactic acidosis from hypoxia-
ischemia (194) (probably because their AMPA glutamate receptors lack
Duration of the Injury GluR2 subunits, allowing them to flux high amounts of calcium—toxic
Duration of the event is another important factor in interpreting an to the cells (195,196)), the cerebral white matter is the part of the brain
imaging study of an asphyxiated child. It is not possible to know the most sensitive to ischemic injury in this age group (194,198,221); thus, it
precise duration of an arrest or of a hypotensive injury in most cases, is the most common location for injury (146,147,222–224).
particularly in a neonate who may have been hypotensive prior to deliv- The motor and visual pathways course though regions of white
ery. In addition, there is almost certainly variability among infants in matter that are commonly injured; therefore, motor and visual impair-
the ability to tolerate reduced energy substrates to the brain. In general, ment are the most common neurologic sequelae of periventricular white
however, as discussed earlier in this section, short episodes of hypoten- matter injury, although the incidence of these sequelae are decreasing
sion do not seem to cause brain injury; this has been found in animal (225). Because the lower extremity axons course medially to the upper
models, as well (153–155). Moreover, mild degrees of hypotension are extremity axons, they are more affected by periventricular injury and
usually compensated by autoregulation. However, in the setting of motor function is more severely affected in the lower extremities than
pressure-passive cerebral blood flow or in hypotension too severe for the upper extremities. This condition, called “spastic diplegia,” is a com-
autoregulation to compensate, the longer the hypotension persists the mon syndrome in neurologically impaired premature neonates, with
more likely it is to result in brain damage and the more extensive the an incidence of 5% to 15% (225,226). Visual impairment is common
brain damage is likely to be. among prematurely born children with spastic diplegia, with an inci-
dence as high as 70% (227–229). This correlation probably reflects the
Injury in the Premature Neonate fact that the geniculocalcarine tracts (optic radiations) and the visual
Neurologic Sequelae of Brain Injury in the association pathways course through the posterior periventricular white
Prematurely Born Neonate matter; therefore, the same white matter injuries that cause motor dis-
Neuroimaging assessment of premature infants is becoming increas- abilities may also cause visual impairment (227–229). Indeed, presence
ingly important as the number of premature births increases (11% of of visual impairment correlates well with sonographic evidence of PVL
all live births in the United States in 2000 [206]) and survival rates of (230), with MR evidence of damage to the occipital lobes and cerebel-
very low-birth weight (<1500 g) infants increase (207), but the sur- lum (the cerebellum has an important role in oculomotor function)
vivors remain at great risk for the neurodevelopmental impairments (231), and with changes in fractional anisotropy (FA) on diffusion ten-
(208–212). Recent studies suggest that the rate of cerebral palsy has sor imaging (DTI) (232,233). The visual impairment is characterized by
decreased (to 9% (212)) but neurologic disability is still seen in 40%, low acuity, visual field defects, disorders of conjugate gaze, abnormal eye
with a rate of 50% in those born from 24 to 28 gestational weeks movements, and oculomotor disturbances; visual-spatial perceptions
(211–214). Indeed, among infants born at 25 or fewer weeks, up to are affected as well (231,234–236). Of interest, the ophthalmologic dys-
90% are stillborn or die before hospital discharge. Of survivors, half function of prematurely born neonates differs significantly from that in
have neurodevelopmental disability and, of these, half have severe dis- term neonates who suffer injury to the visual pathways (235).
ability (215). In childhood, children born at weights of less than 1000 g Even premature infants (<33 weeks gestation) with normal neu-
have significantly more functional limitations and dependency needs, rologic outcomes have a high incidence of cognitive impairment when
requiring significantly more services than closely matched controls examined at age 8 years (237,238). Cognitive impairment is identified
(212,213). At the age of 5 years, children born at 33 weeks or less have in nearly 50% of preterm children with no sonographic abnormalities
significantly higher incidence of neuromotor dysfunction including (noncystic white matter injury) (215,225,239–241), although the inci-
learning disabilities (212,216). When they reach adulthood, people dence of severe impairment is less than 5% (237,238). The incidence of
born at very low birthweight (infants weighing <1500 g at birth) or severe cognitive impairment increases significantly if hydrocephalus or
born moderately or severely premature have less chance of finishing loss of cerebral parenchyma can be detected by sonography (238). The
high school, lower mean IQ, lower academic achievement scores, higher cognitive impairment is suspected to be the result of injury to com-
rates of neurosensory impairments, and subnormal height (217,218). missural tracts and intrahemispheric association pathways, especially
Special healthcare resources are needed by 42% of children born at 24 the posterior corpus callosum, which functions to transfer cognitive
to 28 weeks and by 31% of those born at 29 to 32 weeks (217,218). information (238,242–244), with a possible contribution from injuries
Looking at the data in another way, infants who weigh less than 2500 g to the cerebellum (245,246).
account for 11% of all births in the United States, but more than 90%
of neonatal deaths, while those who weigh between 500 and 1500 g Types of Brain Injury in Premature Neonates (and Fetuses)
account for 1% of all live births but more than 60% of all neonatal Several types of brain injuries may occur secondary to hemody-
deaths (219). namic alterations in premature infants, including white matter injury,

Barkovich_Chap04.indd 264 5/25/2011 11:38:42 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 265

TABLE 4-4 Brain Injury in Premature Neonates


Type of Injury Location Pathology
Germinal matrix hemorrhage Walls of lateral ventricles (may extend into Hemorrhage secondary to rupture of thin
ventricle and cause hydrocephalus) walled capillaries in germinal matrix
Cerebellar cortex
White matter injury Deep cerebral white matter (may be multifocal Focal/multifocal necrosis, which forms
or diffuse) astroglial scar.
May cavitate.
Venous infarction Cerebral basal ganglia, deep and periventricular Infarction often hemorrhages.
white matter May liquefy, leading to porencephaly
Cerebellar atrophy Cerebellar vermis and hemispheres Unknown

germinal matrix hemorrhage, intraventricular hemorrhage, periven- higher among preterm neonates with cerebellar hemorrhage compared
tricular hemorrhagic infarction (PVHI), and cerebellar infarction with preterm controls (263). Germinal matrix hemorrhage is unusual
and atrophy (Table 4-4) (147, 247). Of note, all of the types of injury after 34 weeks of gestation (3,222,264). The choroid plexus also fre-
described in prematurely born infants can develop in fetuses of the same quently bleeds in premature infants, often in association with germinal
postconceptional age. In our examinations of fetuses with sonography matrix hemorrhage.
and MRI, we have seen germinal matrix hemorrhage, intraventricu- Periventricular and intraventricular hemorrhage in the premature
lar hemorrhage, white matter injury, and PVHI. Others have reported neonate has been divided into four grades, related to its severity (265).
similar findings in the literature (248–251). Indeed, in the series of de Grade I (Fig. 4-22) refers to germinal matrix hemorrhage with no or
Vries et al., 16% of PVHI was present at birth, with another 3% already minimal intraventricular hemorrhage. Grade II bleeds extend from the
evolved into a porencephalic cyst (252). Takanashi et al. found PVHI subependymal germinal zone into the ventricles (which remain normal
to be a common cause of congenital hemiplegia in term infants, again in size). In Grade III bleeds (Figs. 4-23 and 4-24) intraventricular hem-
suggesting prenatal injury as the cause (41,42). orrhage is associated with ventricular enlargement. This ventricular
enlargement can result from parenchymal injury (ex vacuo ventricular
Periventricular and Intraventricular Hemorrhage As stated earlier, enlargement) or, in some cases, communicating hydrocephalus.
when tissue is hypoperfused and subsequently reperfused, especially
in the presence of increased venous pressure, the weakened capillar- Venous Infarctions Grade IV bleeds of Papile et al. (Fig. 4-25) were
ies may rupture, resulting in hemorrhage (151,253,254). This type of at one time thought to be extensions of germinal matrix bleeds into
hemorrhage is most frequently seen in the ventricular/subventricular the surrounding parenchyma. Currently, they are thought to result
zones (also called the germinal matrix), the area within the ventricular from venous infarction (266) and are termed PVHI (267). Approxi-
wall in which the cells that compose the brain are generated (255,256). mately 15% of all infants with intraventricular hemorrhage will
It is thought that the hemodynamic instability associated with birth is develop PVHI (267), with 80% to 90% developing within the first
related to these germinal matrix hemorrhages, as 40% have their onset 96 hours after delivery (252). Bilaterality of PVHI and more exten-
within 5 hours of birth (257) and 90% within the first 4 days (151). The sive hemispheric involvement by the injury are predictive of poorer
germinal matrix is extremely vascular but the vessels have very thin outcome (268). When PVHI are bilateral, or if there is a family his-
walls and are sensitive to changes in oxygen supply and blood flow. The tory of PVHI, some consideration should be given to a genetic cause.
germinal zones are most active between approximately 8 and 28 weeks Familial porencephaly has been reported in several families, caused by
of gestation; neurons are the primary cells produced in early stages, a microangiopathy resulting from mutations to the gene for COL4A1,
whereas glial cells are produced in later stages of germinal zone activ- located at chromosome 13q34 (78). The mutations compromise the
ity (258). Toward the end of the second trimester, the germinal zones structural integrity of the vascular basement membrane, rendering the
diminish in activity and begin to involute; as involution progresses, the vessels susceptible to disruption, especially at times of increased stress
incidence of subependymal hemorrhage decreases. The last areas of the (78). Hemorrhage from COL4A1 may occur at any age, from fetus to
germinal matrix to involute are the regions around the frontal horns. adulthood (45,78).
One area near the posterior aspect of the caudate heads, the ganglionic Ventricular enlargement in Grade III hemorrhages is often asso-
eminence portion of the germinal zone, seems to hemorrhage most fre- ciated with damage of periventricular tissue; therefore, it is an excel-
quently. Another area seems to cap the anterior tips of the frontal horns lent prognosticator of the short- and long-term neurological outcome.
and can be seen for several weeks after birth (see Chapter 2). Only 26% of patients with Grade III/IV hemorrhages survived in one
The external granular layer of the cerebellum is a germinal zone as large study of 484 infants, whereas 67% of infants with Grade I/II hem-
well (259), and hemorrhages in this germinal zone are also common orrhage survived (269). Patients with mild hemorrhage and normal
(260,261) and increasingly recognized in prematurely born neonates, ventricular size have less than a 10% incidence of long-term neuro-
with a prevalence of about 20% (262). Cerebellar hemorrhage is more logical sequelae (270). Patients with enlarged ventricles associated with
common in infants weighing less than 750 g at birth; other risk fac- intraventricular hemorrhage have approximately a 50% incidence of
tors include emergent cesarean section, patent ductus arteriosus, and neurological sequelae. PVHI (266,271) is associated with significant
low 5-day minimum pH (263). Neonatal mortality and morbidity are permanent brain injury; 50% to 90% of affected patients have serious

Barkovich_Chap04.indd 265 5/25/2011 11:38:42 AM


266 Pediatric Neuroimaging

FIG. 4-22. Papile Grade 1 hemorrhage with cerebellar hem-


orrhage. A. Coronal sonogram from anterior fontanelle shows
echogenic hemorrhage (white arrow) compressing the body of
the left lateral ventricle in a premature neonate with adjusted
age of 29 weeks. No intraventricular extension was seen. B. Axial
T2-weighted MR image shows the hemorrhage as a focus of
marked T2 hypointensity (black arrows) in the wall of the body
of the left lateral ventricle. C. Axial T2-weighted image through
the lower cerebellum shows one large (white arrow) and several
smaller (white arrowheads) foci of hypointensity, representing
cerebellar germinal matrix hemorrhages.

neurological sequelae that are related to the location and extent of the matter injury in premature neonates has resulted in the assumption by
parenchymal injury (223,252,269,272,273). many that all periventricular white matter injury is a result of preterm
hypoxic-ischemic injury, which is clearly not the case. Infections, meta-
White Matter Injury Another type of injury commonly seen in pre- bolic diseases, hydrocephalus, and the presence of complex congenital
term infants is white matter injury of prematurity. This injury is most heart disease, to name a few, can cause injury to periventricular white
commonly called periventricular leukomalacia, or PVL. However, it is matter (see Chapter 3, 8, and Refs. 274,275). To avoid these pitfalls and
important to remember that this “white matter injury” is frequently the confusion generated by them, the term white matter injury of pre-
accompanied by neuronal and axonal disease that affects the thalami, maturity will be used in this chapter, keeping in mind that nearly all
basal ganglia, cerebral cortex, brain stem, and cerebellum (224); there- parts of the brain are, to some extent, involved.
fore, it is better considered an encephalopathy of prematurity (224). Thirty years ago, white matter injury was demonstrated pathologi-
In addition, nearly any part of the white matter (periventricular, deep, cally in 85% of infants with birth weights between 900 and 2200 g who
or subcortical) can be affected, and, in our experience, injury is most survived beyond 6 days (276); with advances in neonatology, that num-
common in the deep white matter of the hemisphere, some distance ber has dropped considerably (277–280). Although sonography stud-
from the ventricular wall. In addition, use of the term PVL for white ies report incidences of PVL to be in the 5% to 10% range (281), this

Barkovich_Chap04.indd 266 5/25/2011 11:38:42 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 267

FIG. 4-23. Papile—Grade 2 hemorrhage. A. Intraventricular hemorrhage is shown as hyperechogenic regions (arrows) in the
enlarged frontal horns of the lateral ventricles. B. Sagittal plane sonogram shows hemorrhage (arrows) layering in the occipital horn
of the lateral ventricle. C. Axial T1-weighted image of a different patient shows subacute hemorrhage, manifested as homogeneous
hyperintensity in the left ganglionic eminence (the site of hemorrhage, large white arrow), and in the trigones and occipital horns
of the lateral ventricles (small white arrows). D. Axial T2-weighted image shows that the subacute hemorrhages are homogeneously
hypointense.

technique is much better at detecting cavitary (“cystic”) injury than with impaired autoregulation (176,267,274,278,286,287), as well as
noncavitary injury. Autopsies and MR studies, which show noncavitary secondary effects on brain development (147,224). As discussed pre-
as well as noncavitary injury, reveal white matter injury in about 50% viously, recent data suggest that the injury may result from effects of
of preterm neonates (280,282–284). Thus, it seems that ultrasound ischemia upon late oligodendrocyte progenitors, which show selective
underestimates the prevalence of this type of injury (147,280,283). In vulnerability to hypoxic ischemic injury due to the immaturity of their
view of the fact that 25% to 50% of prematurely born infants with receptors and cellular metabolism (147,194–197,221). Another poten-
white matter injury have cognitive and learning disabilities (285), this tial cause is injury to the neurons of the subplate, a transient deep layer
is an important factor supporting the use of MRI. The occurrence of of the cortex that acts as a way station for axons that will ultimately
white matter injury is likely related to many different factors includ- connect with permanent cortical layers (288). Moreover, the time at
ing intrauterine infection, postnatal infection, hypoxia, premature which late oligodendrocyte progenitors are present in the brain and
rupture of membranes, maternal chorioamnionitis, and hypotension the time at which the subplate is present coincide with the period of

Barkovich_Chap04.indd 267 5/25/2011 11:38:43 AM


268 Pediatric Neuroimaging

FIG. 4-24. Papile scale—Grade 3 hemorrhage, acute and sub-


acute stages. A. Coronal sonogram from anterior fontanelle
shows bilateral large echogenic masses (white arrows), represent-
ing ganglionic eminence hemorrhages that have extended into
the lateral ventricles. B. Parasagittal sonogram shows the extent
of the intraventricular hemorrhage, extending from the body
of the lateral ventricle (white arrowheads) through the trigone
to the temporal horns (white arrows). C. Coronal SPGR image
shows moderately dilated ventricular system, with subacute
hyperintense clot in the inferior frontal horns (white arrows)
and the inferior temporal horns (white arrowheads). D. Axial
T2-weighted image shows subacute/chronic blood products
lining the ventricular walls with more hyperintense clot (black
arrows) adherent to the choroid plexuses. E. Axial diffusivity
(Dav, ADC) map shows the subacute blood as very hypointense
signal in the ganglionic eminences and choroid plexuses. No
acute parenchymal injury is identified.

Barkovich_Chap04.indd 268 5/25/2011 11:38:44 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 269

FIG. 4-25. Evolution of PVHI. A. Coronal transfontanelle sonogram of a 1-week-old prematurely born neonate (born at 25 week of
gestational age) shows echogenic hemorrhage in the left lateral ventricular germinal matrix (white arrowhead) and in the periventricular
white matter (large white arrows), the latter representing hemorrhagic infarction. The ventricle is compressed. Incidental Grade 1 hem-
orrhage (small white arrow) is seen in the germinal matrix of the right lateral ventricle. B. Axial T2-weighted image from an MR scan
performed several days after (A) shows the hypointense hemorrhage extending from the ventricle into the periventricular white matter
(white arrows). C. Coronal SPGR performed 2 weeks after (B) shows marked ventriculomegaly and evolving hyperintense hemorrhages.
The parenchymal hemorrhage (white arrows) shows some central hypointensities, representing breakdown and resorption of the blood
products. D. Axial T2-weighted image shows the hypointense margins of the hemorrhagic infarction (white arrows) and the central hyper-
intensity, confirming the central breakdown and resorption of hemorrhage and infarcted tissue.

Barkovich_Chap04.indd 269 5/25/2011 11:38:45 AM


270 Pediatric Neuroimaging

FIG. 4-25. (Continued) E. Coronal SPGR obtained at 35 weeks shows some reduction in ventricular size, but the region of the
hemorrhagic infarction has evolved into a porencephalic cavity (white arrows) with smooth walls and a communication with the left
lateral ventricle. F. Axial T2-weighted image obtained at the same time as (E) is slightly degraded by motion, but shows the persistent
enlargement of the lateral ventricles and the left frontal porencephaly (black arrows). Some hemorrhage in the wall of the right lateral
ventricle (white arrow) can still be seen.

vulnerability of the fetal brain for white matter injury of prematurity Inder et al. have developed a grading system for overall white
(198). The amount of injury correlates with duration of ischemia, and matter abnormality in prematurely born infants. They developed a
the location correlates with the location of late oligodendrocyte precur- score based upon 5 white matter parameters: (a) foci of T1 shorten-
sors (221). All three different types of white matter injury described by ing in the white matter, (b) reduced white matter volume, (c) ven-
Volpe (147) (diffuse white matter injury, which has the mildest clinical triculomegaly, (d) thinning of the corpus callosum, and (e) impaired
manifestations; focal/multifocal noncavitary white matter injury, which myelination. The white matter is evaluated and a score from 1 to 3 is
has intermediated clinical severity; and focal/multifocal cavitary white assigned for each variable, with the overall grade determined by add-
matter injury, which has severe clinical manifestations) can be identi- ing the individual scores (291). Using this method, they found that
fied and distinguished by MRI. The diffuse and noncavitary lesions motor and cognitive outcome at age 2 years are strongly associated
have stable appearances on MRI, but cavitary white matter lesions with the severity of white matter injury (292). Sie et al. (293) proposed
evolve, with the injured regions undergoing necrosis followed by liq- a different grading system, based only on white matter injury: Grade
uefaction and then shrinkage of the cavity with resultant focal enlarge- 1—normal; Grade 2—punctate T1 shortening; Grade 3—T1 and T2
ment of the adjacent ventricle (264,289). On pathology studies, the shortening with six lesions or less; Grade 4—more than six lesions with
two most common locations are the posterior periventricular white T1 and T2 shortening; Grade 5—extensive white matter changes with
matter adjacent to the lateral aspect of the trigone of the lateral ven- hemorrhage and/or cavitation; and Grade 6—diffuse signal changes
tricles and the frontal white matter adjacent to the foramina of Monro in periventricular and subcortical white matter with hemorrhage and
(5,199,200,290); however, MRI studies indicate a propensity for the cavitation.
lesions to occur in the peritrigonal region and in the deep (as opposed
to periventricular or subcortical) white matter at the level of the bodies Cerebellar Injury It has recently been discovered that late cerebel-
of the lateral ventricles (280,282,283). The incidence of white matter lar growth is impeded in children born prematurely, particularly those
injury seems to increase with very low gestational age. A prospective with associated brain injuries. Normally, the cerebellar volume increases
ultrasound study showed a 25% incidence in very premature (24 weeks) rapidly during the last trimester of pregnancy, more rapidly than the
neonates as compared with 5% in 26 to 27 week premature infants cerebral or mean brain volumes (294). In prematurely born neonates,
(279). Another study showed an incidence of 9.2% among 802 infants mean cerebellar volume at term equivalent age is significantly smaller
born between 24 and 32 weeks gestational age (278). In contrast, de than the volume of term born infants. This cerebellar growth impair-
Vries et al. found no difference in the incidence of cystic white matter ment is strongly correlated with associated cerebral injury, particularly
injury between a group born at 32 gestational weeks or less (5%) and intraventricular hemorrhage, even in the absence of direct cerebellar
a group born at 33 to 36 weeks (6%) (281). Although percentages with injury (245,246,294,295). As signaling between the overlying leptom-
white matter injury are almost certainly low, as ultrasound is known eninges and the developing cerebellum is crucial to development of
to be insensitive to noncavitary white matter injury (280,283), it seems the cerebellum, it is very possible that the subarachnoid blood some-
clear that cystic white matter injury is most common in very prema- how interferes with this signaling. Cerebellar hypoplasia may also be,
turely born neonates. in part, due to cerebellar hemorrhage, a finding being discovered with

Barkovich_Chap04.indd 270 5/25/2011 11:38:46 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 271

increasing frequency in prematurely born infants, particularly those tibility-weighted MR sequences are the methods of choice to look for
born at weights less than 750 g (261), and in part due to transsynaptic hemorrhage in the neonate. Neonatal intraparenchymal hematomas are
degeneration of cerebellar tracts and neurons resulting from supraten- isointense to slightly hypointense on T1-weighted images and markedly
torial brain injuries (263). hypointense on T2- and T2*-weighted images in the acute stage (first 3
days); they are not seen well on FLAIR images. They gradually develop
Imaging Findings and Clinical Correlations in Premature increased signal intensity on T1-weighted images (while remaining low
Neonates in signal intensity on T2- and T2*-weighted images) over the next 3 to
It is apparent from the above discussion that much of the brain injury 7 days (Figs. 4-23 and 4-25) (299). Between 7 and 14 days, the hema-
in premature infants is manifested in the deep areas of the brain. For toma gradually increases in signal intensity on T2-weighted images
example, germinal matrix hemorrhage occurs mostly at the caudotha- and remains so while slowly turning isointense to CSF on T1-weighted
lamic notch and parenchymal damage in the periventricular and deep images over the next several months (Fig. 4-25) (299).
white matter. In addition, the degree of enlargement of the ventricles In the acute phase after intraventricular hemorrhage, ventricular
themselves has significant prognostic value. These are areas seen dilatation results from blockage of the CSF pathways by hemorrhagic
well by sonography (Figs. 4-22 to 4-25). Moreover, sonography can particulate matter; this acute hydrocephalus often resolves and is of
be performed in the neonatal intensive care unit, using the anterior no prognostic value. When the hemorrhage is severe, however, it can
and posterolateral fontanels as sonographic windows. Maintenance cause an obliterative arachnoiditis that usually occurs in the basilar cis-
of body temperature is vital in these infants and it is difficult to pro- terns. The arachnoidal adhesions block the normal flow of CSF and
vide adequate warmth and monitoring outside of the neonatal ICU necessitate permanent CSF diversion. Superimposed upon this sec-
unless an MR compatible incubator is used (296). Therefore, initial ondary hydrocephalus is the fact that patients with posthemorrhagic
imaging evaluation of these infants by sonography is recommended ventricular enlargement have an increased incidence of periventricular
despite the known superior sensitivity and added information with white matter injury, pontosubicular injury, and olivocerebellar injury
MRI (280,283,292,297). (300). Tissue loss secondary to cavitary white matter injury results in
ventricular enlargement that usually appears towards the end of the
Periventricular and Intraventricular Hemorrhage Sonograms second week. As one does not want to divert CSF in a patient with ex
show germinal matrix hemorrhage as a region of increased echogenic- vacuo ventricular enlargement, it is essential to know the baby’s head size
ity. Images acquired in the coronal plane show a well-defined area of and to look for signs of hydrocephalus (such as dilation of the anterior
increased echogenicity adjacent to the ventricular wall. The most com- recesses of the third ventricle, see Chapter 8) before making a diagnosis
mon location is at the “caudothalamic notch,” inferior to the floor of of communicating hydrocephalus in these patients. Whatever the cause,
the posterior part of the frontal horn (Figs. 4-22 and 4-23). The sagittal the development of this secondary ventricular enlargement implies a
plane is often very useful in differentiating germinal matrix hemor- poorer developmental prognosis for the premature infant (225). Such
rhage from the echogenic choroid plexus because the choroid plexus ventricular dilatation is well demonstrated by transfontanelle sonogra-
does not extend anterior to the foramen of Monro, whereas the caudate phy or MRI (Fig. 4-24); the patient should be scanned approximately
heads and the adjacent ganglionic eminence hemorrhage lie immedi- 1 week after initial documentation of the hemorrhage in order to
ately anterior to the foramen. MRI shows germinal matrix hemorrhage exclude subsequent ventricular enlargement.
as round to ovoid regions that are hypointense on T2-weighted images
(Figs. 4-22 and 4-23). When subacute, they are also hyperintense on Periventricular Hemorrhagic Infarctions PVHIs are ischemic
T1-weighted images (Fig. 4-23). Both ultrasound and MRI also fre- parenchymal injuries with associated hemorrhage that typically occur
quently show hemorrhages of variable size in the cerebellar cortex, in the periventricular white matter adjacent to the lateral ventricle. Size
likely as a result of bleeding within the transient external granular of the infarct varies markedly depending upon the area of drainage of
layer, a germinal zone for the cerebellar cortex (260). Limperopoulos the occluded vein and the amount of collateral drainage by other veins.
et al. found such hemorrhages unilaterally in the cerebellar hemisphere The location depends upon the vein that has been occluded; hemor-
in 71%, isolated to the vermis in 20%, and involving both vermis rhage adjacent to the frontal horn implies occlusion of the transverse
and hemisphere in 9% (263); they were isolated to the cerebellum in caudate vein, adjacent to the ventricular body implies occlusion of the
23%, with the rest being associated with supratentorial lesions (263). terminal vein or striate vein, while hemorrhage adjacent to the trigone
These cerebellar hemorrhages have the same appearance (Fig. 4-22) implies occlusion of the lateral atrial vein (more superior injury) or the
as do supratentorial germinal matrix hemorrhages, but do not result inferior ventricular vein (more inferior injury) (301). PVHIs are seen
in hydrocephalus. When cerebellar hemorrhages are large, significant on sonography as globular, crescentic, or fan-shaped areas of mixed
atrophy may ensue (Fig. 4-26). hyper- and hypoechogenicity (Fig. 4-25A). It is difficult to determine
Intraventricular hemorrhage results in filling of a portion or, some- how much hemorrhage is present based solely on the sonogram. CT
times, all of the ventricular system with echogenic material. When shows large areas of periventricular hypodensity, often with high den-
acute, the intraventricular hemorrhage is extremely echogenic (Fig. sity hemorrhage within it. Acutely, MRI shows regions of hemorrhage
4-24) and may be difficult to differentiate from the normally echogenic (dark on T2-weighted images), often surrounded by nonhemorrhagic
choroid plexus. Power Doppler, which shows vascularity within the venous infarct (hyperintense on T2-weighted images) (252). As the
choroid but not within clot, may be useful to make this differentia- lesion evolves, the blood oxidizes to methemoglobin (Fig. 4-25B)
tion (298). Over the first few weeks after the acute event, the intraven- and is slowly resorbed, resulting in liquefaction of the infarcted areas
tricular clot organizes and becomes well defined and less echogenic. At (Fig. 4-25C and D). Ultimately, the affected brain region liquefies. It
this stage, it appears as a relatively sonolucent mass within the lateral may then shrink, so the adjacent ventricle expands (ex vacuo) into the
ventricle, usually in the body or the atrium. At this time, the clot is less affected region of the hemisphere or the cyst may remain, usually in
echogenic than the choroid plexus, which is situated adjacent to the communication with the adjacent ventricle (Fig. 4-25E and F). Cysts
thalamus. Hemorrhage can also be detected by CT, on which it appears associated with PVHI are unlike the cysts in periventricular leukomal-
hyperdense in the acute phase and becomes isodense at 7 to 10 days acia, which are typically multiple and small. Color Doppler ultra-
after the bleed. Conventional spin-echo and gradient-echo or suscep- sound studies of these hemorrhages show that the vein draining the

Barkovich_Chap04.indd 271 5/25/2011 11:38:47 AM


272 Pediatric Neuroimaging

FIG. 4-26. Cerebellar germinal matrix hemorrhages with volume loss. A and B. Axial T1-weighted images show residual hyperintense
hemorrhage (white arrows) at the periphery of the left cerebellar hemisphere. Note that the affected hemisphere is small. C. Different
patient. Sagittal T1-weighted image shows small cerebellar vermis with foci of T1 shortening (white arrows) and small pons (white arrow-
head). D. Coronal gradient-echo 500/35 image shows marked hypointensity (black arrows) representing substantial hemosiderin in the
periphery of the shrunken cerebellar hemispheres, secondary to extensive germinal matrix hemorrhage.

affected region into the lateral ventricle (typically the thalamostriate sient (305). White matter injury of prematurity should be suspected
vein anteriorly, the lateral atrial vein in the periatrial region, and the on ultrasound studies when increased echogenicity is present in the
inferior ventricular vein in the temporal horn [302]) is nearly always periventricular regions (306); however, edema also causes increased
completely occluded (302,303), adding to the accumulating evidence echogenicity and edema can resolve without any subsequent brain
that these are venous infarctions. DTI studies show increased diffusiv- damage (307). In addition, increased echogenicity can be seen in
ity and delayed development of anisotropy of white matter, in addi- this region in the absence of injury or edema (308), probably due to
tion to delayed loss of anisotropy in cortex within affected regions of specular reflections from the normal bundles of axons. Moreover, nor-
the brain (304). mal scans have been reported in infants subsequently proven to have
white matter injury at autopsy (309,310). Therefore, the appearance of
White Matter Injury of Prematurity When the cerebral white mat- hyperechogenicity in itself is not enough to make a diagnosis of white
ter is investigated by cranial ultrasound, high frequency transducers matter injury. The best early sonographic sign of periventricular white
(up to 10 MHz) should be used for optimum evaluation, and multiple matter injury is the periventricular “flare,” an area that shows loss of
sequential exams should be obtained, as abnormalities may be tran- the normal regularly spaced parenchymal echoes (the normal tissue

Barkovich_Chap04.indd 272 5/25/2011 11:38:47 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 273

architecture); instead, the damaged tissue has an indistinct, “globular” called echolucencies or hypoechoic lesions) from liquefaction of the
appearance (Fig. 4-27A). In addition to the loss of normal tissue archi- injured white matter (Fig. 4-27) (306,314–317). The timing of cavita-
tecture, heterogeneity of the hyperechogenic white matter should be tion varies, depending on the extent and severity of the injury, but it
sought, as heterogeneity (likely representing tissue hemorrhage or typically appears on ultrasound 2 to 4 weeks (usually <3 weeks) after
breakdown) correlates best with poor neurodevelopmental outcome at injury (281,317,318). The size of cavities varies over time, as the injured
2 years (305); the more severely damaged tissue has echogenicity equal tissue undergoes necrosis (cavitation starts), the cavities coalesce (this
to or greater than that of the choroid plexus. If prolonged (>2 weeks) causes enlargement), and then shrinkage/astrogliosis ensues (causing
periventricular flares are seen, the incidence of spastic diplegia or tet- contraction).
raplegia is 50% (311). However, flares have a low sensitivity and posi- White matter injury can be graded by the characteristics of the
tive predictive value for the presence of white matter injury (280) and periventricular white matter on sonography (319). Grade I is defined
for long-term outcome (312,313). Definitive diagnosis of white matter as periventricular and deep white matter areas of increased echogenic-
injury by sonography requires demonstration of heterogeneity (called ity present for 7 days or more (prolonged periventricular flare). Grade
heterogeneous flares [293] and caused by necrosis or hemorrhage) II has areas of increased echogenicity that evolve into small, localized
and the subsequent formation of parenchymal cavities (sometimes frontoparietal cysts (Fig. 4-27C). In Grade III, periventricular areas

FIG. 4-27. Evolution of cystic white matter injury of prematurity (cystic PVL). A
and B. Coronal (A) and parasagittal (B) transfontanelle sonograms at 1 week of age
show periventricular “flares” (white arrows), regions of increased echogenicity with
loss of normal echogenic texture of the white matter near the trigones of the lateral
ventricles. The echoes are less evenly spaces and in some areas some of the echoes
seem to coalesce. C. Coronal transfontanelle sonogram at age 18 days shows areas of
reduced echogenicity (white arrows), representing cavitation and areas of increased
echogenicity (white arrowhead), representing necrosis (probably hemorrhagic) as the
damaged white matter evolves.

Barkovich_Chap04.indd 273 5/25/2011 11:38:48 AM


274 Pediatric Neuroimaging

FIG. 4-27. (Continued) D. Coronal transfontanelle sonogram at age 25 days shows increasing bilateral cavitation (white arrows) of
the damaged white matter. E and F. Coronal (E) and sagittal (F) T1-weighted MR images obtained a few days after (E) shows the
cavitary white matter lesions (white arrows) in the periventricular and deep white matter. Some noncavitary white matter lesions
(white arrowheads in E) can also be identified. G. Axial T2-weighted MR image shows abnormal white matter hyperintensity that
are nearly isointense to the regions of cavitation (black arrowheads), making the cavities difficult to identify. H–J. Sagittal T1 (H),
parasagittal T1 (I), and coronal FLAIR (J) images obtained at age 6 months show changes of chronic white matter injury. At this
point, the cavities have shrunk into small glial scars and the volume of white matter has been markedly reduced. The corpus callo-
sum (white arrows in H) is extremely thin. Cortical sulci reach (black arrows in J) and indent (arrowheads in I) the lateral ventricles
due to loss of white matter. Ventricles have enlarged (ex vacuo) due to the loss of white matter.

Barkovich_Chap04.indd 274 5/25/2011 11:38:48 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 275

FIG. 4-27. (Continued)

of increased echogenicity evolve into extensive periventricular cystic are hyperintense on T1-weighted images and mildly hypointense on
lesions involving occipital and frontoparietal white matter (Fig. 4-27D). T2-weighted images (280,282,283). At most institutions, CT and MRI
Dammann has suggested that Grade I white matter injury can be fur- do not currently play a major role in the early diagnosis of white matter
ther divided into “brief flares” that resolve within 6 days, “intermedi- injury of prematurity because of the difficulty involved in transport-
ate flares” that last for 6 to 13 days before resolution, and “prolonged ing and caring for sick premature neonates. However, the institutions
flares” that resolve after 14 days (320). Sie et al. (293) have suggested that use MRI have found many signal abnormalities in the white mat-
that “heterogeneous flares” (Fig. 4-27A and B) have a worse prognosis ter early in the course of the injury. No sonographic abnormalities are
and that they should be classified as Grade 1b (with Grade 1a being seen that consistently correspond to these MR findings in the majority
applied to homogeneous flares). In general, the long-term outcome of reports (280,283,321); although Leijser et al. have reported hetero-
of affected patients is more dependent on the location and extent of geneous hyperechogenicity in affected regions (305), they found MRI
injury than on the sonographic grade. to be much more reliable than ultrasound in the detection of mildly
Of note, white matter cavitation is much less common than and moderately abnormal (i.e., noncavitated) white matter (322). In
noncavitary white matter injury and seems to be becoming progres- our experience, punctate areas of T1 hyperintensity are seen as early
sively less common (280,283,292). Noncavitary white matter injury is as 3 to 4 days after birth (Fig. 4-28); mild T2 hypointensity can usu-
detected by MRI in about 50% of premature neonates as small foci that ally be seen, as well, in the larger lesions. Reduced diffusivity is seen

FIG. 4-28. Noncavitary white matter injury of prematurity. A. Axial T1-weighted image shows multiple foci of T1
hyperintensity (black arrows) in the deep white matter of both cerebral hemispheres. The lesions, which typically appear
during the first week to 10 days, are of uniform intensity, with no regions of hypointensity to suggest cavitation. B. Axial
DWI shows hyperintensity (black arrows), indicating reduced diffusivity (acute injury) in the two largest lesions.

Barkovich_Chap04.indd 275 5/25/2011 11:38:50 AM


276 Pediatric Neuroimaging

if imaging is performed in the first week (Fig. 4-28). The T1 and T2 the white matter myelinates. As mentioned earlier, regions of cavitation
abnormalities may represent reactive astrogliosis (323), possibly due tend to shrink into small foci of gliosis. Therefore, when cavitation of
to microglial activation (324). They typically remain visible in the the white matter in the premature neonate is mild, mild ex vacuo ven-
white matter for several weeks to months after injury (321), although tricular dilatation will develop along with focal gliosis and focal thin-
some disappear after a few weeks (282,283). They should be differ- ning of the corpus callosum; this constitutes end stage white matter
entiated from hemorrhage, which has much shorter T2 relaxation injury. Extensive white matter injury often cavitates; the MR appear-
time and, therefore, much lower signal intensity on T2-weighted and ance evolves into diffuse white matter gliosis with diffuse callosal
susceptibility-weighted images (see, e.g., Fig. 4-19B). When extensive thinning and moderately to markedly dilated lateral ventricles. The
(involving more than one lobe), these white matter lesions are asso- white matter changes vary in severity; extensive lesions throughout the
ciated with reduced FA, higher radial diffusivity, and higher average white matter (periventricular, deep, and subcortical) result in almost
diffusivity (Dav) in much of the cerebral white matter (325). FA may total white matter loss with severe ventricular dilatation, irregular
be reduced at term equivalent age, even in the absence of white matter ventricular margins, and diffuse callosal thinning and shortening; a
hyperintensities at that time (326) (although it is not known whether shorter corpus callosum is associated with poorer neurodevelopmental
the foci of T1 hyperintensity disappeared or never developed); reduced outcome (328).
FA at term equivalent age is associated with impaired neurodevelop- Correlations have been shown between the severity of white matter
mental outcome at age 2 years (326). injury and clinical outcome at ages 18 (327) and 24 months (231,292).
Larger areas of white matter hyperintensity may undergo necro- Of interest, clinical outcome correlated more with the extent and loca-
sis, resulting in the development of T1 hypointensity either within or tion of injury than the type (cystic, noncystic, hemorrhagic) of injury
adjacent to the areas of T1 hyperintensity, and sometimes T2 hyperin- (231,305,327). Neonates with a few scattered white matter lesions were
tensity within the areas of T2 hypointensity (Fig. 4-29). If the area of normal at 18 months but, as the authors point out, further follow-up
damage is large, small cavities may coalesce into larger ones, resulting in is necessary to determine whether these patients might develop mild
the presence of frank cavities within the white matter (Fig. 4-27E–G). neurodevelopmental deficits later in childhood (327).
The process of cavitation decreases the area of T1 hyperintensity. This The Hammersmith group has described the presence of diffuse
process, combined with evolution of the reactive astrogliosis and sub- high signal intensity, without focal injuries, in the white matter of
sequent increasing T1 hyperintensity caused by myelination, causes prematurely born neonates at term equivalent age, which they call
these lesions to disappear on MRI over the 3 to 4 months after injury. DEHSIs (diffuse excessive high signal intensities, Fig. 4-30) (329).
As the cavities shrink over the subsequent 3 to 4 weeks, the cerebral They believe that this represents a diffuse white matter injury, as
hemispheres show diminished volume of white matter (Fig. 4-27H–J), described in the pathology literature (147), and they have shown
characteristic of what has been called “end stage periventricular that it is associated with increased white matter diffusivity and worse
leukomalacia” (199). outcomes (284). Others, however, have described T2 hyperintense
Sie et al. have looked at the MR evolution of white matter lesions “caps” in the deep white matter anterior to the frontal horns and T2
on sequential MR studies of premature neonates (327). They report hyperintense “arrows” in the deep parietal white matter posterior to
that the small white matter T1 hyperintensities become small areas of the trigones in prematurely born neonates (330) and in fetuses of
gliosis (hyperintense on T2-weighted spin-echo and FLAIR images) equivalent gestational ages (331,332). The precise anatomic substrate
with the same size and location on follow-up studies performed after of these regions has not been definitively determined and, indeed,

FIG. 4-29. Mild cavitation of white matter injury demonstrated by MR. A. Axial T1-weighted image shows several foci of T1 shortening (open white
arrows) in the cerebral white matter. Two of the posterior foci have some hypointensity within them, suggesting cavitation/necrosis. A small subependymal
hemorrhage (solid black arrow) is seen in (A) and (B). B. Axial T2-weighted image shows abnormal hyperintensity in the periventricular and deep white
matter. Very faint T2 hypointensity (open black arrows) is seen within the hyperintense white matter around the ventricular trigones. C. Axial T1-weighted
image shows multiple foci of T1 hyperintensity in the deep cerebral white matter. Two of these foci (white arrows) show central hypointensity, indicative of
cavitation/necrosis.

Barkovich_Chap04.indd 276 5/25/2011 11:38:51 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 277

FIG. 4-30. DEHSI of white matter. A and B. Axial T2-weighted images at two levels show white matter intensity that is
almost the same as the CSF in the lateral ventricles (the subcortical white matter is somewhat spared).

the underlying substrate may differ at different gestational ages may be a harbinger of impaired development, as well. Reduced FA in the
(330–333). Whatever the underlying substrate, all agree that the T2 corticospinal tracts prior to term equivalent age correlates with motor
hyperintensity of white matter is abnormal in some prematures and outcome (338); reduced FA in the optic radiations prior to (232,233)
the use of diffusion imaging should be used for this differentiation. and at term equivalent age (339) correlates with impaired visual func-
As the diffusivity of normal white matter changes both temporally tion. Spectroscopy may also prove useful in early evaluation of white
(different diffusivity at different gestational ages in the same loca- matter injury, as the lactate peak enlarges and NAA peak diminishes in
tion) and topologically (different diffusivity in different regions of affected areas before standard imaging sequences become abnormal.
the developing brain at the same gestational age), comparison with MRI and CT are both useful for the diagnosis of end stage white
normal values for a neonate of equivalent postconceptional age (334) matter injury (“end stage periventricular leukomalacia”) in later infancy
should be performed. and childhood after the fontanels have closed. CT and MRI are also
The value of diffusion imaging and proton spectroscopy has not useful in making a diagnosis in mild cases of white matter injury where
been fully established in the evaluation of the premature brain, but some nonspecific ventriculomegaly is the only detectable abnormality on
results are promising. Diffusion imaging shows reduced diffusion in the sonography. The CT findings of end stage white matter injury are (a)
periventricular white matter in the first few days after injury (Fig. 4-31), irregular outline of the body and trigone of the lateral ventricles; (b)
before ultrasound or conventional MR sequences show any abnormal- reduced quantity of white matter, always at the trigones but in severe
ity (335), but the diffusivity changes normalize quickly (usually within cases involving the whole centrum ovale; and (c) deep, prominent sulci
6 days) (117,119). An important point is that it is crucial to look at cal- that abut or nearly abut the ventricles with little or no interposed white
culated Dav values in all scans of neonates, as the DWIs may be falsely matter (199). MR scans show these same abnormalities (Fig. 4-27 H–J).
negative due to the very long T2 values of the neonatal brain (remember In addition, MRI shows abnormally increased signal intensity in the
that DWIs have contribution from both diffusion and T2). In the pre- periventricular white matter on FLAIR and T2-weighted images, most
mature neonate without brain injury, Dav decreases by 0.021 mm3/s/wk commonly observed in the peritrigonal regions bilaterally (Fig. 4-27J)
(336); therefore, sequential studies can detect abnormal white matter (18,200,340), and delayed myelination (341,342). In general, lower
development even if imaging is not obtained immediately after injury. birth age correlates with more severe myelin deficit (343). The midline,
FA, determined by DTI, is also useful to evaluate white matter injury, sagittal MR image will show thinning of the corpus callosum, most
but requires sequential imaging. It was found that preterm neonates commonly the posterior body and splenium, resulting from degenera-
without white matter injury show progressively increased FA (by 0.008 tion of transcallosal axons (Fig. 4-27H) (18,344,345). MR volumetry
per week) of the white matter on sequential scans, but those with white has shown that the white matter volume is reduced (346) and ventri-
matter injury have stable or decreased anisotropy (297) (also unpub- cles enlarged (347) in end-stage white matter injury. DTI studies have
lished results, Dr. Sonia Bonifacio). The relationship of these early shown increased water motion (348), smaller size of the retrolenticular
changes in anisotropy to long-term outcome has not been completely internal capsule and the posterior thalamic radiations (349,350), and
established; however, the presence of reduced FA later in infancy (at reduced anisotropy in the corticospinal tracts (348,351). These findings
term equivalent age) seems to be associated with impaired neurodevel- also seem to be useful for predicting the severity of neurodevelopmen-
opment (326,337), suggesting that the earlier microstructural changes tal deficits in affected patients (232,326,339,352). These sophisticated

Barkovich_Chap04.indd 277 5/25/2011 11:38:51 AM


278 Pediatric Neuroimaging

FIG. 4-31. Acute white matter injury in a 4-day-old 28-week


gestational age premature infant. A. Axial T1-weighted image
shows slight hyperintensity (arrows) of the deep white matter.
B. Axial T2-weighted image is essentially normal. C. Axial Dav
map shows reduced diffusion (arrows) in the deep white matter,
compatible with acute injury. (These figures courtesy Dr. Joel
Cure.)

techniques are not necessary, however, to make the diagnosis of end One final comment should be made regarding abnormal periven-
stage white matter injury. tricular signal in infants and children. A finding of periventricular
The signal abnormalities of end-stage white matter injury (end hyperintensity and volume loss on FLAIR or T2-weighted images is not
stage periventricular leukomalacia) have a superficial resemblance to specific for white matter injury of prematurity. In fact, periventricular
the normal areas of slow myelination dorsal and superior to the trigo- tissue damage can be caused by many different disorders, such as
nes (see Chapter 2 and Fig. 4-32). The normal regions of unmyelinated ventriculitis (a common sequel of meningitis in infants, see Chapter
white matter, however, are separated from the ventricular wall by a 11), inborn errors of metabolism (see Chapter 3), hydrocephalus (see
thin band of myelinated white matter in the splenium of the corpus Chapter 8), and in utero events (354,355). The diagnosis is best made
callosum and tapetum, whereas the abnormal signal attributable to in conjunction with a thorough past medical history.
injured white matter directly abuts the ventricular wall. The differen-
tiation is best seen on coronal, T2-weighted sequences; axial views may Cerebellar Injury in Premature Neonates Studies have shown
not depict this finding (340). Moreover, loss of volume of the cerebral that a small but significant number of premature neonates (in the
white matter is not present on normal scans (353) and the ventricular range of 8%–20%) suffer either focal or diffuse cerebellar injury
contour in normal patients is smooth, not irregular. (247,262,295,356–358). Focal cerebellar injury is usually due to

Barkovich_Chap04.indd 278 5/25/2011 11:38:52 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 279

FIG. 4-32. Normal immature periventricular white matter. SE 2500/70 (A) and FLAIR (B) images show T2 prolongation in the
periventricular white matter (large arrows) is not accompanied by any loss of white matter volume. A stripe of normally myelinated
white matter (small arrows in B) is present in the immediate periventricular region.

cerebellar hemorrhage. When ultrasound is performed through the frequently involved (16). The remainder of the cerebral cortex is usu-
posterior or the mastoid fontanelle (which improves visualization ally spared. The postulated reasons for this pattern were discussed in
of the cerebellum), evidence of cerebellar hemorrhages is found in an earlier section. Ultrasound acquired in the first day or two may be
up to 9% of premature neonates (260,262,358); detection sensitivity normal; however, by the second or third day, hyperechogenicity may
seems to be increased by use of the mastoid fontanelle (262). Only be detected in the basal ganglia and thalami (Fig. 4-34). CT shows
half of these hemorrhages are associated with supratentorial hemor- hypodensity of the thalami and basal ganglia (often isodense to white
rhage (Fig. 4-22) and nearly all of the smaller ones are clinically silent matter instead of gray matter, Fig. 4-34). MR images acquired in the
(260). MRI shows a higher incidence of hemorrhage (∼20%), seen as first 2 days after injury will show reduced diffusion of dorsal mid-
marked T2 shortening in the periphery of the cerebellum, often mul- brain and thalami (Fig. 4-35A and B), and the thalami will typically
tifocal (Fig. 4-22C) (262). In those patients with moderate to large be hyperintense on the T2-weighted sequence. As in term infants (see
hemorrhages in whom subsequent MR scans were obtained, focal or next section), premature infants who suffer prolonged (>20 minutes)
diffuse (Figs. 4-26 and 4-33) cerebellar atrophy is found (260). There- profound hypotension may suffer injury to the entire brain; in this
fore, it is likely that small cerebellar bleeds, probably originating in the situation, diffusivity is diffusely reduced, and the diffusion images
germinal matrix (external granular layer), are the cause of focal cer- may appear qualitatively normal. Therefore, in the setting of suspected
ebellar injury in a substantial number of premature neonates. Diffuse brain injury, Dav values should be calculated and compared to age-
cerebellar volume loss is found in a group of prematurely born infants matched norms. By the second or third day after injury, T1-weighted
at term equivalent age or later, usually in those of extremely low birth images will show faint, diffuse hyperintensity (Fig. 4-35C) in the
weights. The mechanism of volume loss is unknown (295,356,357), injured regions. T2 hypointensity does not develop until the end of the
although Volpe suggests that it is a form a selective neuronal necrosis first week (5–6 days, Fig. 4-35D). By the middle of the second week,
(147). Other possibilities include diaschisis due to cerebral white mat- the T1 hyperintensity becomes more globular in appearance and more
ter injury or interference with trophic influences from surrounding localized within the dorsal brainstem (if affected), ventrolateral thal-
posterior fossa leptomeninges (245,246,359). ami and posterior putamina (Figs. 4-35 and 4-36). In patients who
are injured prior to about 28 weeks postconceptional ages, damage
Profound Hypotension or Circulatory Arrest in Premature to the basal ganglia seems to result in liquefaction; follow-up scans
Infants A different pattern of brain injury is seen in premature infants show cavities in the location of the lentiform nuclei (Fig. 4-37) (16).
who have suffered profound hypotension or cardiocirculatory arrest; Imaging 1 to 3 weeks after injury may show white matter abnormal-
outcome in these patients is typically very poor (360). In these infants, ity, typically mild, diffuse T2 hyperintensity associated with small foci
injury is predominantly in the deep gray matter nuclei and brainstem of T1 hyperintensity scattered throughout the centrum semiovale.
nuclei (those areas that are most mature and have highest metabolic Imaging studies obtained in the chronic phase, after injured tissue has
rates in the premature and term neonate, Fig. 4-18), although white undergone liquefaction and astrogliosis, will reveal small, shrunken,
matter injury and germinal matrix hemorrhage may develop as well often calcified thalami (Figs. 4-35F, G and 4-37), small brainstem and
(5,16,177,361). More specifically, the dorsal brain stem, anterior cer- cerebellum, small or absent basal ganglia, and reduced cerebral white
ebellar vermis, and thalami are the regions most commonly injured, matter volume (16). The reduction of cerebral white matter is pre-
but the lentiform nuclei (globus pallidus and putamen, particularly sumably the result of loss of the thalamocortical, corticothalamic, and
posterior putamen) and the precentral and postcentral gyri are corticoputaminal axons.

Barkovich_Chap04.indd 279 5/25/2011 11:38:53 AM


280 Pediatric Neuroimaging

FIG. 4-33. Large cerebellar hemorrhage in premature neonate. A. Coronal reformation of SPGR image shows large hem-
orrhage (white arrows) involving nearly the entire right cerebellar hemisphere, the vermis, and extending into the medial
aspect of the left cerebellar hemisphere. B. Axial T2-weighted image shows the subacute hematoma with black hemosiderin
rim (white arrows) and necrotic, hyperintense center. The right cerebellar hemisphere will be extremely small.

FIG. 4-34. Ultrasound and CT of profound hypoten-


sive injury in a 28-week premature infant. A and B.
Sagittal (A) and coronal (B) sonograms at age 4 days
show marked hyperechogenicity (open arrows) in the
thalami and basal ganglia. C and D. Axial noncontrast
CT images at age 5 days show absence of normal gray
matter attenuation in the basal ganglia. Note that some
germinal matrix hemorrhage is present in the walls of
the right lateral ventricle.

Barkovich_Chap04.indd 280 5/25/2011 11:38:53 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 281

FIG. 4-35. MRI of profound hypotensive injury in a 2-day-old, 28 weeks gestational age infant. A–E are from age 5 days. F and G
are from age 3.5 weeks. A and B. Dav maps show reduced diffusivity (hypointensity) in dorsal pons (black arrows in A), ventrolateral
thalami (black arrows in B) and optic radiations (black arrowheads in B). C. Axial reformation of 3D SPGR sequence shows abnormal
hyperintensity in the ventrolateral thalami (black arrows) and posterolateral putamina (black arrowheads). D. Axial T2-weighted
image shows the ventrolateral thalamic nuclei to have very abnormal, sharply-defined hypointensity (black arrows). The putamina
(black arrowheads) and caudate heads are abnormally hyperintense (compare to other gray matter).

Barkovich_Chap04.indd 281 5/25/2011 11:38:54 AM


282 Pediatric Neuroimaging

FIG. 4-35. (Continued) E. Proton MR spectrum (288 millisec-


onds) from basal ganglia shows abnormally low NAA at 2.0 ppm,
slightly elevated lactate (Lac) at 1.33 ppm, and the presence of
propan-1,2-diol (arrowhead) at 1.1 ppm. Propan-1,2-diol is a
base used in Phenobarbital (used for treatment of seizures, see
Chapter 2). F. Axial reformation of 3D SPGR sequence acquired
at age 3.5 weeks shows the thalami (white arrows) are shrunken
and hyperintense, probably representing gliosis and calcifica-
tion. G. Axial T2-weighted image acquired at the same time as
(F) also shows small, calcified thalami (black arrows). Note areas
of abnormal hyperintensity in posterior putamina (black arrow-
heads).

An important concept is that both hypoxia/ischemia and anoxia/ and use of high frequency transducers (363) may ultimately result in
arrest can occur in utero (Table 4-5 and Fig. 4-38A). The pattern of improved detection of parenchymal injury by ultrasound and make
brain damage seen in infants who have suffered in utero injury is, by utilization of other techniques unnecessary. At present, however,
imaging criteria, identical to that seen in postnatal infants of the same sonography is not very sensitive to the detection of nonhemorrhagic,
gestational age (Fig. 4-38B and C) (16,18). noncavitary parenchymal injury (280,283,364) or to brainstem or cer-
ebellar injury. Therefore, if the neurologic status of a child is worse
Imaging Choices in Premature Neonates than can be explained by ultrasound findings, another imaging study is
Premature infants are often hemodynamically unstable and, therefore, usually necessary. In our experience, CT is not significantly more sensi-
transporting them is somewhat risky. Transfontanelle ultrasound can tive than ultrasound for the detection of nonhemorrhagic brain injury
be performed in the neonatal intensive care unit without moving the in premature neonates. The low contrast sensitivity of CT and the high
neonate and is, therefore, the initial imaging study of choice in all pre- water content of the premature brain make white matter injury (the
mature neonates with definite or suspected neurologic impairment. most common brain injury in premature neonates) very difficult to
Techniques such as color Doppler, quantitative sonographic feature identify. Moreover, it is best to avoid the use of ionizing radiation in
analysis (362), examination through posterolateral fontanelles (260), children and in neonates in particular. Therefore, we generally use MRI

Barkovich_Chap04.indd 282 5/25/2011 11:38:55 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 283

FIG. 4-36. Profound hypotensive injury in a


32-week neonate. Imaging performed at age
20 days. A. Axial T1-weighted image shows T1
shortening (arrows) in the dorsal pons. B and
C. Axial T1- and T2-weighted images show T1
> T2 shortening in the midbrain and amygdalae
(arrows). D and E. Axial T1- and T2-weighted
images show T1 shortening and T2 prolonga-
tion in the lateral thalami, globi pallidi, and lat-
eral putamina.

as the neuroimaging study of choice after ultrasound. Standard MRI process, the incidence of moderate to severe neonatal encephalopathy
has much higher contrast resolution and can detect brain injury (man- seems to be falling, with recent estimates of 1 to 3 per 1000 live term
ifest as T1 shortening of affected brain parenchyma) within 2 to 3 days births (369–372). Approximately 0.3 per thousand live term births
of injury. Proton spectroscopy shows metabolic disturbances within (15%–20% of those with encephalopathy) have significant neurologi-
hours of injury (365). Diffusion imaging may miss injuries if performed cal residual (225). Patients may manifest spastic or dyskinetic motor
in the first few hours after injury and will underestimate the extent of impairment (it is estimated that 8% to 15% of cerebral palsy is caused
injury if performed within the first 24 hours (117,119,365,366), but by perinatal hypoxic-ischemic injury [287,373,374]), delayed develop-
is highly sensitive from 2 to 5 days after injury (117,119). In addition, ment (225,375), cognitive impairment (225,376,377), or visual impair-
DTI can be used to assess the size and integrity of white matter path- ment (378); not surprisingly, the clinical outcome seems to be related
ways (232,338,339,367,368). Moreover, MRI can be performed safely to the pattern, as well as the severity, of injury (176,225,375,377,379). It
if chemical blankets or MR compatible incubators (now commer- is, therefore, important to understand both the pathophysiology of the
cially available) are used to keep the neonate warm and nonmagnetic brain damage in these situations and the pathological sequelae. More-
or properly shielded life support and monitoring equipment is used over, a considerable amount of work is being done to find therapies
(see Chapter 1). for asphyxiated neonates (380–382). Therefore, early identification of
those children who have suffered brain injury may soon become vital.
Injury in the Term Neonate
Neonatal encephalopathy in term infants is an important patient care Cortical/Subcortical Injury
issue and carries medical/legal implications, as well. Although some As discussed in the previous section, the regions of the brain most
degree of hypoxia or ischemia is fairly common during the birth susceptible to ischemic damage change as the infant matures. In term

Barkovich_Chap04.indd 283 5/25/2011 11:38:56 AM


284 Pediatric Neuroimaging

FIG. 4-37. Profound in utero hypotensive injury at 28 weeks of gestational age. A and B. Axial CT images 3 weeks after injury show cavitation
(solid arrows) of the thalami with surrounding blood or calcium. The basal ganglia (open arrows) are markedly hypointense. C. Different patient, imaged
approximately 3 weeks after injury. Axial T1-weighted image shows cavitation (arrows) of the basal ganglia.

infants, mild to moderate hypotension results in injury to the cerebral reactive astroglial cells that form scar tissue, although far less scar tissue
white matter and cortex, particularly in the intervascular boundary is formed in the neonate than in the mature brain (2,3,383). Patients
zones (also called “parasagittal zones” or “watershed areas,” Fig. 4-20), with parasagittal injuries tend to present with seizures and/or hypoten-
which lie in the regions between the regions perfused by the anterior sion. Eventually, the neurological exam evolves to proximal extremity
and middle cerebral arteries, and those between the middle and pos- weakness and spasticity (225). Cognitive outcome is variable but seems
terior cerebral arteries, but more extensive injury may occur. Another worse when the frontal watershed zones are involved (375,384); our
change that has occurred in the infant brain since the sixth and seventh data indicate that this cognitive impairment becomes more apparent
month of gestation is the onset of glial response to injury. The brain as the child matures (375,377,385). Disproportionate disturbances in
of the second trimester fetus responds to trauma by liquefaction and development of language or visual-spatial abilities may also be noted
resorption of the damaged tissue without accompanying astrogliosis; (225,376).
the more mature brain of the term infant responds by production of Pathologically, most prolonged mild to moderate hypotensive
events in term infants result in discrete, sometimes multicystic, infarc-
tions in the intervascular boundary zones.
Detection of watershed ischemic injury is difficult by ultra-
TABLE 4-5 Some Causes of Fetal Brain sonography (114) because the affected areas of the brain lie close to
the inner table of the skull near to the midline and angling the trans-
Injury ducer to visualize these regions can be difficult. Using state-of-the-
art techniques with high frequency transducers, the affected areas
Maternal origin
can be seen and will show increased echogenicity of the parasagittal
Maternal shock
white matter, reflecting edema in the subcortical watershed regions
Maternal hypoxia
(Fig. 4-39A) (363). In severe cases, the cortex will also become edem-
Maternal thrombophlebitis
atous; the resulting increase in cortical echogenicity results in blur-
Maternal abdominal trauma
ring of the cortical-white matter junction and effacement of sulci
Maternal hypo/hypertension
with consequent inability to see the extremely echogenic leptom-
Feto-maternal transfusion
eninges. Detection by CT in the acute phase is also difficult because
Fetal origin the frontal and parietooccipital white matter is normally of low
Fetal infection (arteritis, hypotension) attenuation in the newborn (see Chapter 2) and the cortex may be
Fetal arteriopathy (COL4A1 mutations) partially obscured by beam hardening artifacts from the overlying
Hydrops fetalis calvarium (Fig. 4-39B). Beam hardening is less severe with modern
Fetal embolism (placenta, other) CT scanners but the price one pays for good visualization of the
Feto-fetal transfusion cortex is the use of fairly high (radiation) dose techniques. Finally,
even if the parasagittal cortex is edematous, it is not clear that edema
Placental origin augurs permanent damage (307). Evaluation with MRI is, therefore,
Premature placental separation optimal.
Excessive placental infarction If it is possible to obtain MRI during the first 24 to 48 hours of
life, diffusion-weighted imaging and proton spectroscopy are the

Barkovich_Chap04.indd 284 5/25/2011 11:38:57 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 285

FIG. 4-38. MRI of prenatal profound hypoxic-ischemic injury. A.


Axial SSFSE image of 27-week fetus whose mother had suffered an
arrest at 25 weeks gestational age. Thalami are small and abnormal
hypointensity is seen in the lateral thalami (white arrows). Postero-
lateral right putamen is abnormally hyperintense (white arrowhead).
B. Sagittal T1-weighted image 2 days after elective cesarean section
shows abnormal hyperintensity (white arrows) in the dorsal brain-
stem, indicating injury with gliosis and probable calcification in that
region as a result of the prenatal injury. C. Axial T1-weighted image
shows small thalami and abnormal hyperintensity in the ventrolateral
thalami (white arrows) and posterior putamen (small white arrow-
head). The lateral thalamus is abnormally hypointense (large arrow-
head), indicating injury to that structure, as well.

most sensitive MR techniques for the identification of brain injury; injury is isolated (174,176), but it is usually associated with cortical
diffusion-weighted imaging is better for determining the pattern of injury. The reason for the variability in topology of injury is not fully
injury. In mild-moderate hypotensive injury, diffusion imaging shows understood but likely is related to the severity and the duration of the
extensive involvement of the cortex and underlying white matter in hypotension, and possibly accompanying metabolic factors such as
the intervascular boundary zones (watershed zones, Fig. 4-39D and E), hypoglycemia (which is known to exacerbate hypoxic-ischemic injury
but sometimes the area of reduced diffusivity is confined to subcor- [386]). Diffusion-weighted imaging can give false-negative results if
tical white matter and the cortex appears normal, while other times performed in the first few hours after injury (prior to secondary energy
the entirety of the cerebral cortex is involved, well beyond the inter- failure [117,163,164,365,366,387]) and will underestimate the extent of
vascular boundary zones (Fig. 4-40); the basal ganglia and posterior injury if performed in the first 24 hours (119,365). Increased sensitivity
fossa structures are relatively spared (Fig. 4-40). Sometimes (usually in is gained by assessing calculated Dav images or the Dav values themselves
patients who are born before 40 gestational weeks), the white matter (119,388), rather than the DWIs, to compensate for the very long T2

Barkovich_Chap04.indd 285 5/25/2011 11:38:57 AM


286 Pediatric Neuroimaging

FIG. 4-39. Watershed injury in a term neonate at age 2 days. A. Transfontanelle sonogram
using high frequency transducer shows increased white matter echogenicity and blurring
(arrows) of the junction between cortex and white matter. B. Axial noncontrast CT scan
shows subtle cortical and subcortical hypodensity (white arrows) in the intervascular bound-
ary zones (watershed zones). C. Axial T2-weighted image shows hyperintensity of the cortex
(white arrows) in the intervascular boundary zones. The hyperintense cortex becomes nearly
isointense to underlying white matter. D and E. Axial ADC images (b = 700 s/mm2) shows
reduced diffusion (low signal intensity, white arrows) in the intervascular boundary zones;
note that both cortex and subcortical white matter are affected.

Barkovich_Chap04.indd 286 5/25/2011 11:38:58 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 287

FIG. 4-39. (Continued) F. Proton MR spectrum (TE = 288 milliseconds) in the basal ganglia is essentially normal. G. Proton MR spectrum (TE =
288 milliseconds) in the frontal intervascular boundary zone shows the presence of lactate (Lac) at 1.33 ppm, indicating cellular injury and anaerobic
metabolism. Notice the doublet (two peaks) at 1.1 ppm from 1,2-propanediol.

values of the neonatal brain (remember that signal intensity on DWIs affected cortex will be hyperintense compared with adjacent normal
is determined by both T2 and diffusivity factors). In the acute phase, cortex beginning within 24 hours of injury (Figs. 4-39C and 4-40C).
proton MRS shows elevation of lactate and, in severe cases, reduction The first echo (60 ms) of the T2-weighted images is often more sensi-
of NAA (119,365,389,390) in the cerebral cortex (most notably in the tive to edema than the second echo (120 ms) in the first 2 days after
watershed zones) and subcortical white matter, more so than in the injury (391). On T1-weighted images, edematous brain will appear as
deep gray matter (Fig. 4-39F and G). Between 24 hours and 8 days, areas of low signal intensity in the cortex and subjacent white matter
diffusion imaging, the pattern of injury seen on the Dav maps evolves, when compared with normal white matter (392,393); the most obvious
with diffusivity normalizing in some regions and becoming abnormal finding, when present, is loss of gray matter-white matter distinction
in others (119). The reason for this is not certain, but it is likely either (Fig. 4-40B), but this is mainly seen in very severe injury. The multi-
Wallerian degeneration along white matter tracts or spreading excito- planar imaging capacity of MR also aids detection of ischemic brain
toxic injury. Proton MRS remains abnormal, with lactate levels peak- damage; the coronal and sagittal planes are best for assessing high con-
ing at 3 to 5 days after injury, then slowly declining, while NAA levels vexity regions, while the anterior and posterior poles of the cerebrum
start to decline about 3 days after injury (119). On T2-weighted images, are better assessed in the axial and sagittal planes.

FIG. 4-40. Severe cortical-subcortical injury in 4-day-old neonate. A. Coronal transfontanelle sonogram shows diffuse hyper-
echogenicity of the white matter with blurring of the cortical-white matter junction. Note that the deep structures (thalami [T]
and basal ganglia [B]) are relatively spared. B. Coronal T1-weighted SPGR image shows low intensity of the cerebral white matter
with marked blurring of the cortical-white matter junction, compatible with diffuse edema and cortical injury.

Barkovich_Chap04.indd 287 5/25/2011 11:38:59 AM


288 Pediatric Neuroimaging

FIG. 4-40. (Continued) C. Axial T2-weighted image shows


diffuse hyperintensity of the white matter and cortex. The
normal hypointensity of the cortex and its sharp margin
with underlying white matter is gone, replaced by diffuse
blurring of cortex. Basal ganglia (B) and thalami (T) are
relatively spared. D and E. Axial Dav maps show marked
hypointensity (reduced diffusivity) throughout the cere-
brum, most marked in the cortex. Again, note relative spar-
ing of basal ganglia and thalami (in D).

As the injury evolves, spectroscopy and diffusion images undergo signal intensity of the white matter may not be apparent until the end
pseudonormalization (from about 6–10 days) and then begin to show of the first year when T2 hypointensity, secondary to myelination,
evidence of more chronic brain injury, with increased diffusivity on Dav appears (394).
maps and decreased NAA on proton MRS (117,119,388). On anatomic A correlation has been shown between the severity of the cortical
images, cortical thinning and cystic degeneration of the underlying damage and the severity of the subsequent spastic paresis (166,391,395)
white matter is seen in the parasagittal vascular boundary zones. After and cognition (375–377,384) of the patient. The shrunken cortex has a
3 to 4 weeks, the cavitary areas shrink into glial scars (Fig. 4-41). Ex peculiar pattern in which the deep portions of the gyri are more affected
vacuo dilatation of the adjacent lateral ventricles can be seen, particu- than the superficial portions, creating mushroom-shaped gyri known
larly in the trigones and occipital horns. The parasagittal white matter as ulegyria (5). These peculiarly shaped gyri form because of the unique
shows abnormal T2 and FLAIR hyperintensity, although the abnormal vascular supply to the gyri in the infant brain. Takashima and Tanaka

Barkovich_Chap04.indd 288 5/25/2011 11:39:00 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 289

FIG. 4-41. Chronic watershed injury at age 5 months. A. Sagit-


tal T1-weighted image shows an abnormally think corpus cal-
losum (white arrows) secondary to white matter injury in the
cerebral hemispheres. B. Axial T1-weighted image shows abnor-
mal cortical hyperintensity in the affected watershed regions
(white arrows). The hyperintensity is likely a result of astrogliosis
causing increased amount of bound water in this (still) imma-
ture brain. C. Axial T2-weighted image at age 5 months shows
loss of hypointensity of the cortex and abnormal hyperintensity
of underlying white matter (white arrows) at the areas of injury.

(396) have shown that, in the newborn, there is greater perfusion to the cardiocirculatory arrest, as compared to mild or moderate hypoxia/
apices of the gyri than to the cortex at the depths of sulci. Therefore, hypotension (Table 4-6) (5,17,119,156,170,177,379,397). This group
when a hypoxic-ischemic event occurs, the tissue loss is greater in the of patients shows injury primarily in the lateral thalami, posterior
depths of the sulci, resulting in the characteristic “mushroom” shape. putamina, subthalamic nuclei, hippocampi, and corticospinal tracts
MRI can identify ulegyria because of the characteristic gyral pattern on MR studies (170,180,181,388,398), locations corresponding to
and the underlying tissue loss and gliosis (Fig. 4-42). Probably the most those regions of brain most metabolically active (182), with the highest
important reason to identify this entity is to differentiate it from poly- blood flow (184), and highest degree of synapse construction (187,399)
microgyria, which is a malformation of neuronal organization and is at the time of birth. Some patients will, in addition, show injury in the
seen in a number of inherited syndromes. In myelinated brain, polymi- lateral geniculate nucleus and optic radiations. The cortex is relatively
crogyria (see Chapter 5) appears on MRI as an area of thickened cortex spared, other than the perirolandic gyri (170,180,181,379). The most
with irregularity of the cortical-white matter junction and, often, para- severely injured patients in this group have injury to brainstem nuclei
doxically smooth pial surface. It is not associated with perinatal brain (119,379,400); the majority of these brain stem-injured patients likely
injury or with neonatal encephalopathy. die before they are imaged and, therefore, are represented more in
pathologic (5,177,180,181,361,401) than in the radiologic literature.
Profound Hypotension Patients with the profound hypotension pattern appear to have
As discussed in an earlier section, a different pattern of brain injury different neonatal courses and different subsequent neurologic deficits
is seen in neonates who have suffered profound hypotension or than those with the moderate hypoxia/hypotension pattern. Patients

Barkovich_Chap04.indd 289 5/25/2011 11:39:00 AM


290 Pediatric Neuroimaging

FIG. 4-42. Ulegyria. A. Coronal T1-weighted image shows shrunken left parietal cortex with hypointensity of the underlying white
matter (white arrows), and an enlarged ipsilateral ventricle. The overlying gyri are shrunken, with the deeper portions of the cortex
being more severely involved than the superficial portions. B. Coronal T2-weighted image shows abnormal hyperintensity in the
parietooccipital white matter and of the depths of the sulci (black arrows), reflecting tissue damage. Note that the superficial aspects
of the cortex are less involved, giving the gyri a mushroom-shaped appearance.

with the profound pattern of injury have lower 1 minute Apgar abnormal movement until as late as 7 to 14 years (225,408–410). More-
scores, usually 3 or below (397,402), or a postnatal cardiocirculatory over, nearly half of the patients with late onset of choreoathetosis have
arrest (379,403). Clinical manifestations depend upon the severity of a history of normal neurologic development until the extrapyramidal
the injury. Patients with very severe brain injury often die in infancy signs and symptoms develop (408–410) (of interest, such children
(379,397,402) or develop choreoathetosis in addition to quadripare- seem to have injury to the subthalamic nuclei [398]). Finally, many
sis, severe seizure disorders, microcephaly, and mental retardation children who appear normal at age 1 or 2 years go on to have cognitive
(375,397,403–406). Patients with moderate injury almost always have delay (375) and learning difficulty when they reach school age (407).
spastic and dyskinetic motor impairment (398,402,403). Patients with Therefore, even mild imaging abnormalities may have significant con-
mild injury may initially develop normally after mild neonatal enceph- sequences on long-term outcome.
alopathy or may be mildly delayed (173,179,400,402,407). It is impor- On pathologic studies, the patients are found to have moderate
tant to remember that many children who develop choreoathetosis to severe destruction in the hippocampi, putamina, inferior colliculi,
may not manifest extrapyramidal signs until after the first year of life central gray matter of the midbrain, other brainstem nuclei, and the
(225,403). The large majority develops choreoathetosis between the ventro-lateral nuclei of the thalami (156,361,401,406). Positron emis-
ages of 1 and 4 years (408,409); however, some patients do not develop sion tomographic studies of patients with athetoid cerebral palsy show

TABLE 4-6 Patterns of Injury in Diffuse Hypoxic-Ischemic Injury


Age of Child Mild to Moderate Hypotension Profound Hypotension
Premature Neonate (up to 32 Periventricular/deep white matter injury Thalamic, basal ganglia, and brainstem injury
postconceptional weeks
Term neonate (∼34–56 Cortex and white matter injury (white matter exclu- Dorsal brainstem, anterior cerebellar vermis,
postconceptional weeks) sively in mild injury; parasagittal watershed cortex thalamus, basal ganglia, corticospinal tracts,
and white matter in moderate injury; entire cortex and perirolandic cortex injury. In severe
and underlying white matter in severe injury) injury, all supratentorial structures affected.
Older child (more than 4–6 Parasagittal watershed injury (cortex and white Basal ganglia and diffuse cortical injury;
postnatal months) matter) perirolandic and thalamic sparing

Barkovich_Chap04.indd 290 5/25/2011 11:39:01 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 291

TABLE 4-7 Findings and Effective Times for Imaging Techniques in Perinatal
Hypoxic-Ischemic Injury
Technique Findings Timing Comments
Ultrasound Increased echogenicity 2–10 d Poor assessment of deep structures and of cerebral convexities
X-ray CT Low attenuation 1–7 d Temporal lobes difficult to assess. Ionizing radiation.
Anatomic MRI T2 hyperintensity 24 h
T1 hyperintensity 2–3 d–mo
T2 hypointensity 6–7 d–mo
Tissue loss (atrophy) 10–12 d
Proton MRS Increased lactate 1–15 d Relatively mild elevation until secondary energy failure (>24 h).
Decreased NAA Day 3 or later
DWI Reduced diffusivity 1–5 d Minimal during first 24 h. Maximizes at 3–4 d. Pattern evolves over 8–10 d.
Pseudonormalization 5–10 d
Increased diffusivity >10 d

CT, x-ray computed tomography; DWI, diffusion-weighted imaging of the patient; MRI, magnetic resonance imaging; MRS, proton magnetic resonance spectroscopy.

normal cortical activity, but diminished metabolism in the thalami and evolve. If no intervention (which might necessitate earlier imaging) is
lentiform nuclei (405), consistent with the MR and pathologic findings. planned, the optimal time for early imaging is 3 to 4 days after the injury
(Figs. 4-43 and 4-44). Sonograms will show hyperechogenicity of the
Timing of Appearance of Imaging Findings The time of detection affected structures, CT will show reduced attenuation of affected struc-
of imaging abnormalities varies with the technique utilized (Table 4-7) tures, anatomic MRI will show T1 and T2 hyperintensity, diffusion-
and the severity of the injury; the most severe injuries can be detected weighted MRI will show reduced diffusion, and proton MRS will show
during the first 16 to 24 hours after injury, whereas the milder inju- elevated lactate and diminished NAA peaks.
ries are not visible on imaging studies (other than diffusion-weighted In the first 2 to 3 days after profound hypotension, ultrasound may
MRI) for several days. The imaging findings evolve continuously for show hyperechogenicity in the thalami, globi pallidi, putamina, periven-
many weeks, so it is important to either image the neonates at a con- tricular white matter, and perirolandic cortex (Figs. 4-43 to 4-45). This
sistent time after the injury or be aware of how the imaging findings hyperechogenicity can easily be overlooked if the sonographer is not

FIG. 4-43. Profound neonatal hypotension of moderate severity; evolution at ages 17 hours, 4, and 8 days. A. Axial T1-weighted
image shows a nearly normal appearance. Note that the posterior limbs of the internal capsules (white arrows) are bright at this
age despite the injury. B. Axial T2-weighted image at age 17 hours shows slightly increased signal intensity of white matter but is
otherwise normal.

Barkovich_Chap04.indd 291 5/25/2011 11:39:02 AM


292 Pediatric Neuroimaging

FIG. 4-43. (Continued) C. Axial Dav map at age 17 hours shows


minimal reduced diffusivity in the ventrolateral thalami (white
arrows). D. Proton MRS (TE = 288 milliseconds) at age 17
hours from basal ganglia region shows normal height of NAA
peak. Minimal elevation of lactate (Lac) is seen at this stage of
injury. E and F. Parasagittal and coronal sonograms performed
at age 3 days show increased echogenicity (arrows) in the thalami.
G. Axial CT image performed at age 4 days shows hypodensity in
the basal ganglia and thalami.

Barkovich_Chap04.indd 292 5/25/2011 11:39:02 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 293

FIG. 4-43. (Continued) H. Axial T1-weighted image at age 4


days shows absence of the normal spot of hyperintensity in the
posterior limbs of the internal capsules. The lentiform nuclei
and the ventral thalami show faint, diffuse hyperintensity.
I. Axial T2-weighted image at age 4 days shows diffusely hyper-
intense white matter (nearly as bright as CSF) and slightly bright
posterior thalami. The hypointensity in the ventrolateral thal-
ami (white arrowheads) is normal. J. Axial Dav map at age 4 days
shows hypointensity (reduced diffusivity, white arrowheads) in
the ventral thalami and posterior putamina. K. Proton MRS (TE
= 288 milliseconds) from basal ganglia region at age 4 days shows
decreased height of NAA peak and increased height of lactate
(Lac) compared with initial spectrum (D). L. Axial T1-weighted
image on day 8 after birth shows that the hyperintensity in
the basal ganglia and thalami is more localized in the ventro-
lateral thalami (white arrowheads), posterolateral putamina
(white arrows) and medial putamina (black arrowheads).

Barkovich_Chap04.indd 293 5/25/2011 11:39:03 AM


294 Pediatric Neuroimaging

FIG. 4-43. (Continued) M. Axial T2-weighted image at age


8 days shows persistently hyperintense thalami and basal gan-
glia, but no focal abnormalities. N. Axial Dav map at age 8 days
shows that the reduced diffusivity (hypointensity) has shifted
largely to the posterior putamina (white arrows). The diffusiv-
ity in the ventrolateral thalami (white arrowheads) has nearly
completely disappeared. O. Proton MRS (TE = 288 millisec-
onds) from basal ganglia region at age 4 days shows continued
decreased height of NAA. Lactate (Lac) is decreasing and will
be gone by the end of the second week.

actively searching for it (363,411); the finding of thalamic hyperecho- surrounding white matter (170); this finding can be easily overlooked,
genicity portends a poor neurologic outcome (173,412). With very as can they hypointensity of the cerebral cortex (particularly perirolan-
severe injury, increased echogenicity is seen diffusely throughout the dic) and superior cerebellar vermis. It is, therefore, essential to specifi-
cortex, white matter, and basal ganglia (Fig. 4-44). Initially, it will be cally assess gray matter structures (cortex, thalami and basal ganglia) to
more apparent in the white matter and the differentiation between be sure that the attenuation values are similar to other gray matter struc-
hyperechoic white matter and hypoechoic cortex becomes more pro- tures. The presence of low attenuation in the thalami on postnatal days
nounced than usual. However, over the first few days after the event, the 2 to 4 is highly predictive of poor neurological outcome (397). In very
cortex becomes increasingly edematous and hyperechogenic, resulting severe brain injury, the CT shows diffuse hypodensity on the first day
in a loss of cortical-white matter differentiation (Fig. 4-44A, B and of life, involving basal ganglia, white matter, and cortex (Fig. 4-46A);
4-45A). Transcranial Doppler shows decreased resistive index in the the cerebellum is often spared.
first days after injury, probably because of impaired autoregulation. Anatomic MRI will typically be normal if performed on the day
(Normal resistive indices [RI, peak systolic minus end diastolic veloc- of birth. Proton spectroscopy may show elevation of lactate within a
ity divided by peak systolic velocity] of the major intracranial vessels few hours of birth (365,389,390), although it is important to realize
are between 0.71 and 0.75 with a standard deviation between 0.07 and that the lactate level may nearly normalize from approximately 6 to
0.08 [413].) Fluctuations in RI have also been reported, likely reflecting 18 hours before rising again due to secondary energy failure. In some
impaired autoregulation (363). patients with severe injury, reduced NAA may be seen within the first
CT shows hypoattenuation of the thalami and basal ganglia 24 hours of life (365,389,390), although it usually takes 48 to 72 hours.
(Fig. 4-43C), with these gray matter structures become isodense with Diffusion imaging will typically show reduced diffusion in the lateral

Barkovich_Chap04.indd 294 5/25/2011 11:39:04 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 295

FIG. 4-44. Very severe profound neonatal hypotensive injury: acute (3 days), subacute (15 days) and chronic (8 months) imaging.
A and B. Coronal (A) and parasagittal (B) transfontanelle sonograms at age 3 days show increased echogenicity in the basal gan-
glia and white matter, as well as blurring of the cortical-white matter junctions. C. T1-weighted image at age 3 days shows diffuse
hyperintensity of the basal ganglia (white arrows) and thalami and diffuse hypointensity of the white matter. The normal hyperin-
tensity of the posterior limb of the internal capsule is not seen. D. T2-weighted image at age 3 days shows slight hyperintensity of
the posterior thalami and the cerebral white matter. The hypointensity normally seen in the posterior limb of the internal capsule
is absent, confirming severe, diffuse edema.

Barkovich_Chap04.indd 295 5/25/2011 11:39:05 AM


296 Pediatric Neuroimaging

FIG. 4-44. (Continued) E and F. Dav maps at age 3 days show


extensive hypointensity (reduced diffusivity) in the basal ganglia
(B), thalami (T), cerebral cortex (C), and subcortical white mat-
ter (W). The extent of reduced diffusivity suggests a severe injury.
G. Proton MRS (288 milliseconds) at age 3 days shows reduced
NAA and markedly elevated lactate (Lac). Both of these findings
suggest severe injury. H. Axial T1-weighted image at age 15 days
shows markedly abnormal hyperintensity (white arrows) of the
basal ganglia, thalami, and posterior perisylvian cortex and central
cavitation (C) within the putamina and thalami. The white matter
is abnormally hypointense. I. Axial T2-weighted image at age 15
days confirms the cavitations (C) of the deep gray nuclei and the
abnormal edema (hyperintensity) of the white matter.

Barkovich_Chap04.indd 296 5/25/2011 11:39:05 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 297

FIG. 4-44. (Continued) J and K. Axial T2-weighted images at age 8 months show that the basal ganglia cavities have largely shrunk
and the basal ganglia are consequently small. The white matter volume has markedly diminished and is abnormally hyperintense,
and the cortex is very thin (black arrows) in multiple areas. This is the end result of very severe profound neonatal hypotension.

FIG. 4-45. Profound neonatal hypotension of moderate sever-


ity, evolution from 2 days to 2 years. A. Coronal sonogram at
age 3 days shows mildly increased echogenicity of thalami (T),
lentiform nuclei (B) and deep white matter. Some blurring at
the cortical-white matter junction (white arrowheads) is seen.
B. Axial T1-weighted image at age 2 days shows diffusely
increased signal intensity in the basal ganglia (white arrows).
C. Axial T2-weighted image at age 2 days shows diffuse hyper-
intensity of the thalami, white matter (almost isointense with
CSF), and cortex, highly suggestive of diffuse edema.

Barkovich_Chap04.indd 297 5/25/2011 11:39:06 AM


298 Pediatric Neuroimaging

FIG. 4-45. (Continued) D–F. Axial Dav maps at age 2 days show hypoin-
tensity (reduced diffusivity) in the dorsal midbrain (white arrows, D),
basal ganglia (white arrowheads, E), ventrolateral thalami (white arrows,
E), and corticospinal tracts (black arrows, F). These are the most com-
mon location of early findings on diffusivity maps. G. Proton MRS
acquired from the basal ganglia/thalami at age 2 days shows a small
NAA peak and elevated lactate (Lac). H. Axial T2-weighted image at age
12 months shows T2 prolongation (white arrows) and sulcal enlargement
in the cerebellar vermis.

Barkovich_Chap04.indd 298 5/25/2011 11:39:07 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 299

FIG. 4-45. (Continued) I. Axial T2-weighted image at age 12 months shows small
basal ganglia and hyperintensity in the thalami (open arrows) and putamina (solid
arrows). J. Axial T2-weighted image at age 12 months at the level of the frontopari-
etal convexity shows abnormal hyperintensity (arrows) along the perirolandic gyri.
K. Axial T2-weighted image at age 2 years shows T2 hyperintensity and volume loss
in the lateral thalami (white arrowheads) and posterior putamina (white arrows). L.
Axial T2-weighted image at age 2 years shows T2 hyperintensity and volume loss in the
perirolandic white matter and cortex (white arrows).

thalami within the first 24 hours of life (Fig. 4-43C) (119,365,366). abnormalities will be at their maximum; both diffusion-weighted/Dav
(Remember that sensitivity is improved by using calculated average dif- images and T1-weighted images will show abnormal signal intensity
fusivity (Dav) images or Dav values instead of unprocessed DWIs, which in the lateral thalami and posterior putamina (Fig. 4-43H and J) and
underestimate injury because of the very long T2 relaxation values of along the corticospinal tracts up to the perirolandic cortex (119). If the
neonatal brain.) However, diffusion imaging may yield false negative brainstem is injured, reduced diffusivity is seen in the dorsal midbrain
results in the early hours of life (163,365,366) and will nearly always or pons (Fig. 4-45D). In severely injured patients, Dav values may drop
underestimate the amount of damage if performed in the first 24 hours to less than 50% of normal in the thalami, basal ganglia, and dorsal
of life (before secondary energy failure) (117,119,365). Dav values of brain stem during this time period, with values as low as 0.4 × 10−3
less than 0.8 × 10−3 mm2/s in the thalami or posterior limb of the inter- mm2/s (119). Reduction of Dav by more than 25% to 35% is associ-
nal capsule and less than 1.1 × 10−3 mm2/s in the white matter at ages ated with poor long-term outcome (119,337,388,414,415) (for normal
1 to 4 days seem to be associated with permanent injury (388,414). It neonatal Dav values see Bartha et al. [334]). Extensive cortical injury,
should be noted that metabolite ratios (on MRS) and Dav values (on if seen (Figs. 4-44E, F and 4-46D, E) portends a very poor neurologic
DTI) change continuously over the first week of life (see Fig. 4-43) and outcome (379,395). The second echo T2-weighted images may be
pseudonormalize in neonates by the fifth or sixth day (117,119). By day normal or show symmetric absence of the normal spot of hypointen-
3 after injury, T1- and T2-weighted images become definitely abnor- sity in the posterior limb of the internal capsule (Fig. 4-44D). When
mal, although findings are subtle (119,169). T2-weighted sequence severely injured, the basal ganglia will show T2 hyperintensity, becom-
show that the basal nuclei, particularly the posterior thalami, are isoin- ing isointense with white matter on T2-weighted images (Fig. 4-43M
tense with white matter (instead of normal hypointensity); this hyper- and 4-46C). On proton MRS, NAA is reduced by up to 40% by day
intensity is best seen on the first (60 milliseconds) echo but will be seen 3 to 4, while the size of the lactate peak maximizes during this time
with longer echoes, as well (Fig. 4-43I). During days 3 to 5, diffusion period (119,415). In very severe brain injury, the T1-weighted images

Barkovich_Chap04.indd 299 5/25/2011 11:39:08 AM


300 Pediatric Neuroimaging

FIG. 4-46. Value of physiological imaging in two patients. A–F. Extremely severe profound hypotensive injury; images
obtained 1 day after birth. A. Axial noncontrast CT shows extensive brain swelling with hypoattenuation of the basal gan-
glia, thalami, white matter, and cortex. The ventricles are compressed and the cranial sutures are widened (“split”). B. Axial
T1-weighted image shows compressed lateral ventricles and diffuse hypointensity of the cortex, white matter, and basal gan-
glia. The ventrolateral thalami (white arrows) stand out as relatively hyperintense, possibly because they are more cellular
and myelinated than the other portions of the cerebrum. C. Axial T1-weighted image shows diffuse hyperintensity, with near
isointensity of gray and white matter due to massive edema. Ventrolateral thalami stand out as hypointense, probably for the
same reason as their T1 hyperintensity in (B). D and E. Axial Dav maps show marked hypointensity (reduced diffusion) of
nearly all parts of the brain with the exception of the frontal white matter (F) and cerebellum (C). F. Proton MRS (TE = 288
milliseconds) from basal ganglia/thalami shows enormous lactate peaks (Lac). Note that the NAA peak is already lowered.
G–J. Perfusion imaging of acute, severe profound hypotensive injury associated with meconium aspiration. G and H. Axial
Dav maps show markedly reduced diffusivity in the deep gray nuclei and perirolandic cortices. I. Relative cerebral blood flow
map from arterial spin labeled perfusion imaging demonstrates hyperperfusion of damaged deep gray nuclei and perirolan-
dic cerebral hemispheres. J. Axial T2-weighted image shows marked enlargement of the deep perforating arteries in the basal
ganglia and thalami.

Barkovich_Chap04.indd 300 5/25/2011 11:39:09 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 301

FIG. 4-46. (Continued)

are nearly uniformly hypointense and T2-weighted images nearly shrunken and show T2 hyperintensity compared to normal myelinated
uniformly hyperintense due to the diffuse edema, while the Dav maps brain tissue (Fig. 4-45H–J). In mild-moderately affected patients,
are nearly uniformly hypointense due to the extent and severity of abnormalities are most commonly seen (>90%) in the ventrolat-
injury; proton MRS shows an enormous lactate peak and small NAA, eral thalami, posterior putamina, and corticospinal tracts from the
even on the first day after birth (Fig. 4-46). Limited experience with internal capsule up through the perirolandic cortex (Fig. 4-45H–L);
perfusion imaging in the subacute phase shows hyperperfusion of in 50% to 60% of patients, the anterior cerebellar vermis is affected
injured regions (Fig. 4-46G–J). (Fig. 4-45H) (416,417). Involvement of the subthalamic nuclei may
By 7 to 10 days after the injury, ultrasound shows better definition be seen in children with dyskinesia (398). In more severely affected
of the hyperechogenic areas and some increase in ventricular size as patients, more extensive involvement is seen until, in the most severely
the edema resolves. CT scans also show resolution of edema and may affected patients, multicystic encephalomalacia may involve most of
begin to show high signal, probably representing hemorrhage or calci- the brain (Fig. 4-47). The T2 prolongation may not become evident
fication, in the affected gray matter nuclei (170,178). On MRI, Dav has in the cerebral white matter until myelination is well under way (Fig.
nearly normalized in the thalami, basal ganglia, and corticospinal tracts 4-45 H–J). It may be seen at ages as young as 8 months (394) or not
(Fig. 4-43N), although it may be reduced in locations that were initially until 13 to 14 months, depending on the location of the injury and the
normal (119); the new areas of reduced diffusion may represent Walle- degree of associated myelination delay. MRI is much more sensitive
rian degeneration or spread of excitotoxic injury. On proton MRS, the than CT to the damage in the deep cerebral nuclei and corticospinal
size of the lactate doublet is decreasing during this time and the NAA tracts (170,418) and to the myelination delay that almost invariably
and choline peaks have decreased (Fig. 4-43O) (119). On anatomic accompanies diffuse ischemic brain injury (18,341).
images, the high signal seen in the deep gray matter on T1-weighted
images becomes heterogeneous, with small areas of marked focal Effects of Brain Cooling
hyperintensity in the globi pallidi, the ventrolateral thalami, and in the In the past few years, brain cooling (to 34°C for 3 days) has begun to be
posterior putamina (Fig. 4-43L). Heterogeneous high and low signal is used as an immediate treatment for neonatal hypoxic-ischemic brain
seen in the basal nuclei on the T2-weighted images, probably the result injury. Few definitive results have been published. Our experience is
of astrogliosis and mineralization (323). The extent of injury is vari- that cooling seems to reduce the amount of injury in mild or moderate
able, being limited to the lateral thalami and posterior putamina in the brain injury detected by MRI if the MR scan is performed within 2 days
least severely injured patients, and progressively involving the periro- of the time the cooling has ended. It appears to have little or no effect
landic cortex, hippocampi, superior cerebellar vermis, the remainder of on severely brain-injured babies. The pattern of injury also appears to
the deep cerebral nuclei, the central mesencephalon, and the remainder be affected, as we have seen less basal ganglia injury and more white
of the cerebral cortex as the injury becomes progressively more severe. matter injury than we were accustomed to seeing in noncooled neo-
In the most severely injured patients, the entire cerebral cortex, all the nates with similar clinical pictures at birth. These results remain very
deep cerebral nuclei, the cerebellar vermis, and many brainstem nuclei preliminary at the time of publication of this book.
are injured (166,169,170,395). The extent of involvement is most likely
related to the duration and severity of the hypotensive incident, possi- Notes on MRS in Asphyxiated Term Infants
bly superimposed upon genetic predisposition. The T1 hyperintensity Proton MRS is extremely valuable in the assessment of encephalo-
and T2 hypointensity slowly fade (Fig. 4-44) as atrophy of the injured pathic neonates. Although early work focused upon phosphorus (31P)
tissues develops and myelination progresses. MRS (161,419–422), the limited availability and long acquisition time
In the chronic phase, weeks to months after the injury, the affected of 31P MRS on most clinical scanners and the improvements of much
areas of brain may undergo cavitation (Fig. 4-44H–K); or they become faster proton MRS (117,365,366,384,389,390,423–427) has resulted in

Barkovich_Chap04.indd 301 5/25/2011 11:39:10 AM


302 Pediatric Neuroimaging

FIG. 4-47. Severe cystic encephalomalacia of most of the brain in a 4-month-old infant who suffered extremely
severe profound hypotension at birth. Basal ganglia, white matter, and cortex are severely damaged. A. Axial
T1 weighted image at the level of the basal ganglia shows diffuse volume loss. The basal ganglia are shrunken
and hyperintense. The white matter shows marked volume loss and multiple cysts of varying size. The cortex is
thinned, especially centrally. B. Axial T2 weighted image at a higher level shows decreased volume of abnormal,
hyperintense white matter, with multiple cysts of variable size.

proton MRS becoming established as a key sequence in the evaluation outcome at age 12 months (390). Amess et al. (427) found that thal-
of these neonates. Proton MRS is particularly useful in the assessment amic Lac/NAA more than 2 standard deviations above the mean
of neonates in the first hours after birth, when the imaging studies neonatal value predicted adverse outcome at age 12 months with a
are normal or subtly abnormal and diffusion studies are unreliable specificity of 93% and a positive predictive value of 92%. In their data,
(117,119,365,366,390). Lactate elevation, resulting from anaerobic Lac/NAA was more predictive than Lac/Cr or Lac/Cho. Barkovich et
glycolysis secondary to mitochondrial dysfunction, is detected on long al. found that the basal ganglia Lac/Cho ratio was most predictive of
echo proton MRS by identification of the characteristic lactate doublet 12-month outcome (384). Penrice et al. found that normal control
(two sharp peaks) centered at 1.33 ppm (Figs. 4-43 to 4-46). Elevated neonates have Lac/NAA ratios less than 0.3 in the thalami, but asphyxi-
lactate can be detected by proton MRS as early as 4 to 8 hours after ated neonates generally have values above 0.4 and seriously injured
birth (365), although lactate levels remain relatively low until the onset infants above 0.5; they found that elevated Lac/NAA ratios were asso-
of secondary energy failure, about 24 hours after the injury (119). ciated with poor long-term prognosis (429). Others have also found
It appears that the most important metabolite values in the assess- correlations between brain lactate levels in the first few days of life and
ment of brain injury are NAA and lactate. Cheong et al. looked at abso- neurologic outcome (424,426). In general, the presence of a detectable
lute concentrations of metabolites and found increased lactate and lactate peak in the first few days after birth is a sign of significant brain
decreased NAA, choline, and creatine after neonatal hypoxic-ischemic injury and is predictive of subsequent neurologic impairment; higher
injury (428). Of these metabolites, the decreased NAA was the most lactate predicts worse prognosis. (CAUTION: lactate can be present in
prognostically accurate in distinguishing normal/mildly injured neo- the CSF of normal neonates (430,431). Therefore, make certain that your
nates from severely injured neonates (428). However, calculation of voxel does not contain CSF when acquiring your spectra!) Reduced NAA
absolute metabolite concentrations is impractical in the acute clinical and creatine in the subsequent few days to weeks is also evidence of
setting, so the ratios of peak areas (which depend upon both concen- significant brain damage and emphasizes the poor prognosis of the
tration and T2 relaxation time) are more commonly used. We have affected child (384,425,432). It is important to recognize, however, that
found Lac/NAA to be the most useful ratio. In our experience, initial the presence of lactate is normal in the immature white matter ([433]
lactate elevation (in the first few hours after injury) usually dimin- and Chapter 2) and that the quantity of NAA in the brain varies with
ishes and remains at low levels until 18 to 24 hours when it begins to maturity (434,435). Moreover, different regions of the brain mature at
increase; this delayed elevation is thought to relate to delayed energy different rates (see Chapter 2) (185). Therefore, it is crucial to know the
failure (429). Hanrahan et al. (389) found that lactate to creatine ratios postconceptional age of the infant and the region of the brain from which
(Lac/Cr) correlated well with a reduction of high energy phosphates as the spectrum was obtained before interpreting the proton spectra.
detected by 31P MRS and with stormy neonatal courses. They found that Some authors have investigated the importance of glutamate levels,
Lac/Cr greater than 1 in the first 18 hours of life had a high predictive as determined by in vivo proton MRS, in perinatal asphyxia (436–438).
value (87%) of either neonatal death or abnormal neurodevelopmental Both Groenendaal et al. and Zhu et al. studied glutamate levels in the

Barkovich_Chap04.indd 302 5/25/2011 11:39:10 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 303

first 2 days after birth using short echo time (~30 milliseconds) proton hemorrhage, of whatever cause, results in dilation of the fourth ven-
MRS at 1.5 T (they are not elevated at the end of the first week [437]). tricle, the outcome is usually very poor (444).
Both the alpha glutamate peak at 3.75 ppm and the beta and gamma It seems that venous thrombosis is the most common cause of neo-
glutamate peaks (at 2.0–2.5 ppm) were elevated, although the elevation natal thalamic (straight sinus thrombosis, Fig. 4-48), choroid plexus
of alpha glutamate did not reach significance (probably because of the (straight sinus thrombosis), parasagittal (superior sagittal sinus throm-
minor contribution of alpha glutamate to total glutamate signal). In a bosis, Fig. 4-49), and temporal or occipital lobe (vein of Labbé, trans-
larger series, Zhu et al. (438) report elevation of the alpha glutamate verse sinus, or sigmoid sinus thrombosis, Fig. 4-50) hemorrhage (30),
peak, as well, and found correlations between the Glu/Cr ratios and possibly as a result of injury to the immature neonatal venous system
outcome. due to changing positions of the cranial bones during delivery (445).
Another important detail in the evaluation of encephalopathic Venous infarctions have already been discussed in an earlier section of
neonates by spectroscopy is to carefully determine the chemical shift at this chapter on localized infarctions. When checking for coagulopathy,
which the lactate doublet is located. The methyl protons in lactate reso- it is a good idea to look for COL4A1 mutations, which can result in
nate at 1.33 ppm. Propane-1,2-diol (propylene glycol), a solvent that is hemorrhage due to congenital vascular wall weakness (77,78). Lobar
used as the injection vehicle for administration of phenobarbital and hemorrhages can be seen in many locations in the neonatal brain.
diphenhydantoin, crosses the blood–brain barrier and accumulates in Lobar hemorrhages are presumed to have many causes; however, in
the brain. The methyl protons of propan-1,2-diol resonate as a doublet most neonates, the cause is never identified (446). In general, neonates
at 1.15 ppm (see Chapter 2 and Fig. 4-39), immediately upfield from with lobar hemorrhages of unknown cause have a good prognosis; a
lactate. It is critical to carefully assess the chemical shift of the peaks so as poorer outcome is associated with lobar hemorrhages in the setting of
not to mistake the propan-1,2-diol doublet for that of lactate; the prog- low Apgars, perinatal hypoxia-ischemia, or respiratory distress (446).
nostic implications of lactate accumulation in the brain are important, Another disorder to consider when large parenchymal hemor-
whereas the presence of propan-1,2-diol is not. rhages are seen in fetuses or neonates is neonatal alloimmune thrombo-
cytopenia. In this disorder, fetal platelets that carry paternally derived
Parenchymal and Intraventricular Hemorrhage in the antigens cross the placenta and enter the maternal circulation where
Term Neonate they induce the production of maternal antibodies. When these mater-
In contrast to premature infants, in whom germinal matrix hemor- nal antibodies cross the placenta and enter the fetal circulation, fetal
rhage fairly commonly extends into the lateral ventricles, intraventric- platelets are destroyed and their function impaired (443,447). Affected
ular hemorrhage is uncommon in term infants. Documented sources infants typically present with widespread petechiae or purpura that
of intraventricular hemorrhage in this age group include the choroid develop within a few hours of birth. Intracranial hemorrhage is seen
plexus, residual germinal matrix, genetic disorders, vascular malfor- in up to 30% of cases; as many as half of these occur before birth
mation, tumor, extension of hemorrhagic cerebral infarction (usually (448,449). The diagnosis is made by demonstrating maternal alloan-
thalamic), and coagulopathy (30,78,160,439–442). The most impor- tibodies directed against a paternal platelet antigen. The rate of recur-
tant reasons for differentiating the various sources of neonatal hemor- rence in subsequent pregnancies is quite high (448). Imaging acutely
rhages are that (a) choroid plexus and thalamic hemorrhages are most shows large parenchymal hemorrhages or choroid plexus hemorrhages.
commonly seen in stressed infants; (b) patients with thalamic hemor- Subdural blood may be present, as well. The regions of hemorrhage
rhages have a high incidence of neurologic sequelae, such as spastic subsequently undergo liquefaction and, if imaged in the late subacute
paraparesis, hydrocephalus, and seizures (439); and (c) patients with or chronic phase, large cyst-like regions of porencephaly or macrocys-
coagulation disorders, such as alloimmune thrombocytopenia, have tic encephalomalacia are present, usually with ex vacuo enlargement of
a good prognosis if properly treated (441–443). When intracranial the adjacent ventricle (447).

FIG. 4-48. Thalamic hemorrhage in a neonate. A. Axial noncontrast CT shows left thalamic hemorrhage (arrows) with intraventricular extension.
B. Sagittal SE 550/11 image shows subacute hemorrhage in the lateral, third, and fourth ventricles. High signal intensity (open white arrows), presumably
thrombus, is present in the proximal straight sinus. C. Phase-contrast MR venogram (Venc = 20 cm/s) shows flow (high intensity, solid white arrows) in the
vein of Galen and the distal straight sinus, but occlusion of the proximal straight sinus at the site of the thrombus in B.

Barkovich_Chap04.indd 303 5/25/2011 11:39:10 AM


304 Pediatric Neuroimaging

FIG. 4-49. Hemorrhagic left frontal infarct secondary to


sagittal sinus thrombosis. A. Sagittal T1-weighted image
shows complex hematoma in left frontal lobe. Note hyper-
intense methemoglobin (black arrows) and isointense
deoxyhemoglobin (black arrowheads) within the hema-
toma. B. Axial T2-weighted (SE 3000/120) image shows
the markedly hypointense hematoma (black arrows). Note
the enlarged superior sagittal sinus (black arrowheads), sug-
gesting thrombosis. C. Sagittal maximum intensity projec-
tion from 2D TOF venogram shows absence of flow related
enhancement along the course of the superior sagittal sinus
(black arrows).

FIG. 4-50. Acute occipital lobe hemorrhage in a neo-


nate. A. Axial T2-weighted image (SE 3000/120) shows
large acute hemorrhage (black arrows) in the right
occipital lobe. B. Maximum intensity projection from
2D TOF venogram shows lack of flow related enhance-
ment in the right transverse sinus (white arrows), con-
sistent with acute thrombosis.

Barkovich_Chap04.indd 304 5/25/2011 11:39:11 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 305

The imaging appearance of intraventricular blood was described injured regions, whereas MRI shows T2 prolongation of affected gray
in the section on premature infants. and white matter (seen best on first echo of T2-weighted sequence); both
typically show diminished ventricular and sulcal size. After 24 to 48 hours,
Timing of Studies diffusivity becomes markedly reduced, NAA decreases, and lactate mark-
The subject of optimal timing of imaging studies after neonatal hypoxic- edly increases in injured areas, correlating with secondary energy failure;
ischemic brain injury warrants special mention. In catastrophic injury, these findings portend poor neurological outcome (388). At this time,
severe abnormality is seen immediately by nearly any imaging modality. standard imaging studies cannot determine whether brain damage and
For less than catastrophic injury, however, timing is best understood in neurological sequelae will be severe or minimal unless other associated
the context of secondary energy failure, discussed at the beginning of the features such as focal hemorrhages are present. Therefore, the best time
section on Diffuse Ischemic and Inflammatory Brain Injury. As discussed to image patients is at 3 to 4 days after injury; edema (seen on anatomic
in that section, brain metabolism transiently normalizes for 18 to 24 hours MRI, CT, and sonography), reduced diffusivity, and metabolite abnormali-
after an ischemic injury (162,165). At 18 to 24 hours, it is thought that suf- ties (reduced NAA, elevated lactate and glutamate) are present and near
ficient structural damage to membranes and molecules accumulates such maximum at these times. The worst possible time to image is 1 week after
that the electron transport chain in the mitochondria ceases to work and injury. At 1 week, diffusivity has pseudonormalized, lactate has nearly dis-
normal metabolic processes start breaking down (162,165). As a result of appeared, and edema has resolved, but significant tissue loss is not yet
this 18 to 24 hour window of somewhat normal cellular function after the apparent; therefore, an imaging study at 1 week may look nearly normal,
injury, spectroscopy and diffusion images can be falsely negative during the even if significant damage has occurred. If interpreted as normal, the cli-
first hours after injury in asphyxiated neonates and, even when positive, will nician and parents may be led to falsely believe that the infant has suf-
generally underestimate the extent of brain injury (117,119,162,366). This fered no significant brain damage. If follow-up scans are to be performed
clinical experience correlates with findings in animal experiments, indicat- to assess the extent of brain injury, therefore, they should be obtained a
ing that diffusivity and spectroscopy may be transiently abnormal (mild minimum of 3 weeks (best 4 months [450]) after the injury. The repeat
injury), biphasic (moderate injury—abnormal during hypoxia-ischemia, scan (preferably an MR, which is considerably more sensitive to detection
followed by transient normalization during the “latency period,” followed of white matter and deep gray matter damage) will show a return to near-
by permanently abnormal), or severely and permanently abnormal (early normal in those patients who will have relatively mild deficits, whereas
cell necrosis) (164,387). The animal experiments indicate that transiently cystic or hemorrhagic parenchymal degeneration and progressive atro-
reduced diffusion is associated with selective neuronal death, whereas phy indicate a more guarded prognosis.
both the biphasic and permanently abnormal situations are associated
with widespread cell death in affected areas (164,387); energy produc- Injury in Older Infants and Children
tion in animals with biphasic diffusion metrics follows a similar bipha- When older children are encephalopathic after hypoxia, ischemia, or
sic pattern (165). Humans are not imaged during hypoxia-ischemia. In circulatory arrest (e.g., after drowning), findings are generally similar
catastrophic injury, reduced diffusivity, low NAA and high lactate are seen to those in adults. If the injury is caused by profound hypotension, low
almost immediately; these finding persist and outcome is terrible. In less attenuation is seen in the basal ganglia on CT (Fig. 4-51) and reduced
severely affected patients, imaging during the first 24 hours shows find- diffusivity with T2 hyperintensity is seen in basal ganglia and cortex
ings of the “latency period,” with nearly normal diffusivity and metabo- on MRI (Fig. 4-51 and 4-52). If injury is caused by mild to moder-
lite levels (119). By 12 to 24 hours, CT shows diffuse low attenuation in ate hypotension, watershed injury is seen (Fig. 4-53). MRI is the ideal

FIG. 4-51. Profound hypotensive injury in a 12-month-old infant. A. CT image obtained 36 hours after arrest
shows edema with low attenuation in the basal ganglia and cerebral cortex. B. Axial T2-weighted (SE 2500/80) MR
image 10 days later shows abnormal hyperintensity of the basal ganglia, posterior thalami, and cerebral white matter
diffusely.

Barkovich_Chap04.indd 305 5/25/2011 11:39:12 AM


306 Pediatric Neuroimaging

FIG. 4-52. Drowning injury in a 4-year-old boy. A. Axial SE 600/9 image shows blurring of the cortical-white matter junction in
the occipital lobes. B. Axial SE 2500/70 image, somewhat degraded by motion, shows hyperintensity of the frontal and parietooc-
cipital cortex. C and D. Axial DWIs (b = 1000 s/mm2) shows reduced water motion in the corpus striatum (caudate nucleus and
putamen) and in most of the cerebral cortex and white matter with the exception of the perirolandic regions (white arrows).

study if it can be done quickly and diffusion-weighted imaging is avail- days, the use of this criterion may result in false negative results if the
able. Findings are similar to those in adults. DWIs will show reduced study is performed less than 3 days after injury (451). Standard MRI
diffusivity in the cerebral cortex and corpus striatum within the first findings of asphyxic injury in a child are very subtle in the first 24 to
24 hours of injury (Fig. 4-52), while spectroscopy will show glutamate 72 hours, consisting of subtle swelling with mild T1 and T2 prolonga-
and glutamine elevation if short echo times are used; lactate is not con- tion in the basal ganglia and insular cortex and with blurring of gray
sistently seen (451). In severe injury, a “reversal sign” may be seen on the matter-white matter interfaces (Figs. 4-52 and 4-54) (451). On spin-
Dav images, with markedly reduced diffusivity of cerebral white matter echo T2-weighted images, an early finding that may be very helpful is
compared with cortex and basal ganglia, by the third and fourth day the appearance of a curvilinear stripe of T2 prolongation (Fig. 4-55) at
after injury (Fig. 4-54) (452). If diffusion imaging is abnormal, MRS can the cortical-white matter junction, particularly at the depths of sulci in
be used for prognostication (426). The presence of lactate or reduced the watershed region (18,451,453), which corresponds to blurring of the
(by 25% or more) NAA, Cr, or NAA/Cr correlates strongly with poor cortical-white matter junction on T1-weighted images. This sign, which
outcome (451). However, as NAA levels remain normal for several may represent cortical laminar necrosis, indicates significant cortical

Barkovich_Chap04.indd 306 5/25/2011 11:39:12 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 307

damage. Of note, Christophe et al. (453) found these signs to be both an accumulation of venous and capillary blood in the white matter
sensitive and specific, with a positive predictive value of poor outcome secondary to impaired venous drainage. The prognosis of children
of 82% when the imaging was performed in the first 3 days after injury. with this sign is dismal (456,457).
If MRI is not quickly available or if adequate monitoring/support Note that the pattern of brain injury from hypoxic/ischemic injury
cannot be performed on the MR scanner, CT can be performed, as the in older infants and children differs from that seen in neonates. This
unstable child is easily monitored within the CT scanner. Initial CT difference seems to be related to the physiological and biochemical
images (<24 hours) will show subtle hypoattenuation of the basal gan- changes that occur in the brain with maturation; there are different
glia and insular cortex; sometimes, the cisterns around the midbrain patterns of regional activity at different ages in the maturing brain
are effaced. Subsequent scans (24 to 72 hours) will show diffuse cere- (see Fig. 4-21) (182–185). The changes are most apparent in injury
bral edema, with decreased gray matter-white matter differentiation, from profound hypotension; older infants, children, and adults show
effacement of sulci and cisterns, and decreased attenuation of the basal involvement of the basal ganglia, with relative sparing of the thalami,
ganglia and cortex (Fig. 4-51A) (454,455). Hemorrhage may become and significant cortical involvement with relative sparing of the periro-
apparent in the basal ganglia or cortex by 4 to 6 days after the injury. landic regions (Figs. 4-52 and 4-55) (170,453). In chronic stages, severe
An ominous finding is the so-called CT reversal sign in which the cere- atrophy of the entire cerebrum may be seen (Fig. 4-55C and D). In
bral hemispheric white matter becomes hyperdense compared to cor- injury from mild to moderate hypotension, the differences from the
tex (Fig. 4-54A) (456,457). This reversal sign is believed to result from pattern of injury seen in neonates are minor (18).

FIG. 4-53. Watershed injury in 4–month-old


infant. A and B. Axial T2-weighted (SE 3000/120)
image 5 days after injury shows abnormal hyper-
intensity (white arrows) of the cerebral cortex and
subcortical white matter in the intervascular bound-
ary zones. C. Dav map shows hypointensity (reduced
diffusivity, white arrows) in intervascular boundary
zones.

Barkovich_Chap04.indd 307 5/25/2011 11:39:13 AM


308 Pediatric Neuroimaging

FIG. 4-54. Reversal signs. A. CT image shows the cerebral


white matter in the left hemisphere and much of the right hemi-
sphere is hyperintense compared to the gray matter of the cere-
bral cortex. B and C. MR reversal sign in a 2-year-old scanned
after a drowning accident. Axial T2-weighted image (B) shows
faint hyperintensity of white matter and blurring of the cortical-
white matter junction. Axial Dav map (C) shows hypointensity
(reduced diffusivity) in the white matter with, paradoxically,
relatively normal gray matter signal intensity.

each embryo develops its own chorion and placenta; they are no more
CNS INJURY IN MULTIPLE PREGNANCIES alike than any other two siblings born to the same parents.
Ischemic brain injury in twin gestation is an important subject and deserv-
ing of some discussion. Twin pregnancies represent 1.2% of spontaneously
conceived pregnancies (458). Multiple pregnancies can be caused by either
Ischemic Brain Injury in Multiple Pregnancies
of two mechanisms. Monozygotic (identical) twins result from splitting of The incidence of prenatal mortality is high in twins; up to 80% of twin
a single zygote into two or more embryos. These twins may share the same pregnancies diagnosed by routine sonography in the first trimester
amniotic sac and chorion (monoamniotic monochorionic), may share are singletons by the time of birth (459,460); the surviving fetus has
the placenta but have his or her own amniotic sac (diamniotic monocho- one chance in five of neurological impairment (461). The incidence of
rionic), or may each have their own amniotic sac and placenta (diamni- brain injury in twin pregnancies that continue to delivery is also rela-
otic dichorionic; these fetuses are in the same situation as dizygotic twins). tively high, being estimated at 30% in monochorionic twins (about ten
When more than one ovum is present and each is fertilized by a different times more than in age-matched controls); by contrast, the incidence
sperm, dizygotic (fraternal or nonidentical) twins result. In this situation, in dichorionic twins is about 3% (5,147,462–466).

Barkovich_Chap04.indd 308 5/25/2011 11:39:13 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 309

FIG. 4-55. Profound childhood hypotensive injury in a 2-year-old. Acute and chronic phases. A. Axial proton density image
(SE 2500/30) shows hyperintensity of the putamina and caudates compared with the thalami. The cortex-white matter junc-
tion is indistinct. B. Axial T2-weighted (SE 2500/70) image shows a stripe of hyperintensity (arrows) between the cortex and
underlying white matter. This can be a useful sign in evaluating asphyxiated children in the first few days after injury. C and D.
Follow-up images at age 3 years. Marked diffuse atrophy of the cerebrum is present.

Part of the neurologic morbidity is related to the high prevalence “twin-twin transfusion” and the spectrum of disorders is referred to as
of premature birth in twin gestations, but comparisons between single- the “twin-twin transfusion syndrome (TTTS).” If TTTS is untreated,
tons and twins matched for gestational age show an increased risk for the prognosis is very poor with overall mortality in the range of 80%
death and developmental impairment in twins (467,468). Although to 100% (470). As a result of the abnormal placental anastomoses, one
other factors (disseminated intravascular coagulation, hypotension, twin (called the donor) loses blood to the other (called the recipient);
or thromboembolism due to death of cotwin) are often factors (147), the donor becomes hypovolemic with associated oliguria, oligohy-
most ischemic brain injury seems to result from the presence of placen- dramnios, and growth restriction while the recipient becomes hyperv-
tal vascular anastomoses (artery to vein is the most important type), olemic with polyuria, polyhydramnios, and cardiac dysfunction (458).
which are nearly always present in monochorionic placentas (469). In Although laser therapy has been used in an attempt to close the anas-
most cases, the anastomoses are balanced, but in 10% to 15%, they are tomoses, they are difficult to cure and the treatment is associated with
unbalanced; the process in unbalanced anastomoses has been termed significant fetal morbidity (471).

Barkovich_Chap04.indd 309 5/25/2011 11:39:14 AM


310 Pediatric Neuroimaging

The type of ischemic brain injury is dependent upon the age of encephalomalacia, and cavitary white matter injury (Fig. 4-56). If the
the fetuses at the time of the transfusion. Injuries reported from events diagnosis is made after birth, the appearance of these injuries in twins
during the second half of pregnancy include intraventricular hemor- is identical to that of singletons with similar injuries at similar postcon-
rhage, localized cerebral infarctions (single or multiple), porencephaly, ceptional ages; all have been discussed and illustrated in earlier sections
hydranencephaly, multicystic encephalomalacia, periventricular leu- of this chapter.
komalacia, and polymicrogyria (5,147,458,462–465). Mechanisms of
injury that have been elucidated include (a) disseminated intravascular
coagulation caused by transfer of thromboplastin material from the
NEONATAL HYPOGLYCEMIA
dead twin, (b) embolism from the dead fetus or placenta, (c) severe Neonatal hypoglycemia is probably under recognized, since common
hypotension and cerebral ischemia resulting from transfusion of blood symptoms, such as stupor, jitteriness, and seizures, may be lacking or
from the surviving fetus to the dead fetus, (d) placental circulatory inconspicuous in many affected neonates; thus, hypoglycemia may
stasis resulting in impaired umbilical cord blood flow or embolism, remain undetected until seizures develop (473). The definition of hypo-
and (5) venous hypertension (147,458,462,463). Mechanism (4) is glycemia in infants varies with the maturational state of the infant, as
currently believed to be the most common (469). Norman et al. (472) less mature infants tolerate lower glucose levels than more mature ones
make the point that monoamniotic twins are particularly prone to and mature infants tolerate lower levels than older children and adults.
umbilical cord entanglement. Currently accepted levels defining significant hypoglycemia in the new-
Although the diagnosis of TTTS is made by sonography, which born are below 30 to 35 mg/dL in the first 24 hours and below 40 to
will show the oligo- or polyhydramnios, the small or distended urinary 45 mg/dL after 24 hours in term infants (473–475). Volpe, and others,
bladder, and the presence of cardiomegaly, fetal MRI best detects brain make the point that absolute values are not useful, as other factors, such
injury (458). In particular, one should look for hemorrhage, infarc- as rate of cerebral blood flow, cerebral utilization of glucose, duration
tions, polymicrogyria, porencephaly, hydranencephaly, multicystic of hypoglycemia, and association with exacerbating factors (hypoxia,

FIG. 4-56. Fetal MRI manifestations of injuries from twin-


twin transfusion syndrome. A. Sagittal SSFSE image shows
periventricular hemorrhage (white arrows) in 22 week fetus
due to presumed hypotension/ischemia. B. Coronal SSFSE
image shows parenchymal loss (black arrows) in 23-week
fetus from infarction in right frontal lobe. Postnatal scan
showed polymicrogyria. C. SSFSE image shows normal brain
of normal cotwin (N on left of image) and injury resulting
in schizencephaly (white arrows) and periventricular nodular
heterotopia (black arrowheads) affected cotwin.

Barkovich_Chap04.indd 310 5/25/2011 11:39:15 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 311

FIG. 4-56. (Continued) D. SSFSE shows twin demise of smaller fetus (black arrows) with nearly complete resorption of brain tissue.
Larger cotwin is normal. E. SSFSE image shows hydranencephaly of surviving cotwin (this case courtesy Dr. Joel Cure).

hyperbilirubinemia) all impact on whether or not brain injury will UCSF, the most common cause of neuroimaging-apparent brain dam-
occur (473,476). Indeed, up to 8% of low-risk infants suffer an episode age secondary to neonatal hypoglycemia is hyperinsulinism secondary
of hypoglycemia using the blood glucose level criteria, typically at 3 to to congenital tumors or hyperplasia of pancreatic beta cells. As Inder
4 hours after delivery (474). Reasons for tolerance of low glucose levels points out, three principles are important in relation to hypoglycemic
by infants include (a) the ability of the newborn brain cells to utilize brain injury in the newborn. First, prolonged and severe, rather than
lactate as an energy source (477,478), (b) low energy requirements of transient or minor, hypoglycemia seems to be required. Second, the
the immature neurons resulting from the low level of neuronal activity injury involves primarily neurons of the upper cortical layers (layers 2
(473), (c) the relatively minor effect of hypoglycemia upon cardio- and 3) in the parietal and occipital cortex, as well as the hippocampi,
vascular function of the newborn (479), and (d) marked increase in caudates, putamina, brain stem, and cerebellum. Finally, mild hypogly-
cerebral blood flow provided by even moderate hypoglycemia (473). cemia combined with mild hypoxia causes injury, whereas either of the
It should be noted that the presence of hypoglycemia exacerbates two conditions in isolation does not (386).
hypoxic-ischemic injury, but that this condition has characteristics of Imaging studies of patients who have suffered damage from peri-
hypoxia-ischemia and not hypoglycemic brain injury (386,473). natal hypoglycemia reflect the pathological findings (480–482) of dif-
Volpe (473) describes four major causes of neonatal hypoglyce- fuse brain damage, with the most severe injury localized primarily to
mia. Transitive-Adaptive hypoglycemia is the term he uses to describe the parietal and occipital cortex of the brain and the underlying white
a heterogeneous, relatively common condition in which the onset is matter (19,20,483); hippocampi, corpus striatum, and cerebellum are
very early after birth, the duration is brief, the degree is mild, and the involved to a lesser extent. In the acute phase, reduced diffusivity is seen
response to glucose is prompt. Included in this group are infants of dia- along with edema of the cerebral cortex and underlying white matter
betic mothers, infants with erythroblastosis, and preterm infants who (Fig. 4-57), including the callosal splenium; T1 hypointensity and T2
suffer hypoxia-ischemia. Infants in this group are rarely symptomatic. hyperintensity in the affected cortex results in lack of distinction between
Secondary-associated hypoglycemia describes infants with hypoglyce- the cortex and subcortical white matter on T1- and T2-weighted images
mia secondary to associated illnesses, such as hypoxia-ischemia, intrac- (Fig. 4-57). MRI shows short T1 hyperintensity and T2 hyperintensity,
ranial hemorrhage, sepsis, and congenital anomalies. Affected neonates probably secondary to necrosis, in the cortex in the subacute phase.
typically present at the end of the first day of life, duration of hypogly- As in ischemic injury, imaging in the subacute phase shows hyperin-
cemia is brief, degree is fairly mild, and response to glucose therapy is tensity and some damage to affected cortex, while diffusion imaging
prompt. About half of infants in this category exhibit neurologic symp- shows reduced diffusivity in the white matter beneath the cortex, rather
toms. Classical transient neonatal hypoglycemia includes principally than in cortex itself (Fig. 4-58) In the chronic phase (Fig. 4-57D and E),
small for gestational age term infants who have suffered intrauterine the cortex and underlying cerebral white matter initially show cystic
malnutrition; it is significantly less common than the first two groups. encephalomalacia and, ultimately, become shrunken and atrophic.
Most affected infants are symptomatic, typically presenting toward the The only reported proton MRS studies were performed with a short
end of the first day of life. The degree of hypoglycemia is relatively echo time of 30 milliseconds. Large peaks were seen from 0.9 to 1.6
severe, duration may be prolonged, and large amounts of glucose ppm (484). Although the authors suggested that lactate was present, no
are required as therapy. Severe recurrent hypoglycemia is the final and intermediate or long echo spectrum was performed to unequivocally
least common category; most infants in this group have primary dis- demonstrate the presence of lactate. It is likely that the macromolecular
orders of glucose homeostasis. Causes include Beckwith-Wiedemann peaks were the result of massive cellular necrosis with release of macro-
syndrome, beta cell nesidioblastosis, beta cell hyperplasia, endocrine molecular lipids. Although it is not unreasonable to suppose that some
deficiencies, and inborn errors of metabolism. In our experience at lactate was present, verification will require further MRS studies.

Barkovich_Chap04.indd 311 5/25/2011 11:39:16 AM


312 Pediatric Neuroimaging

FIG. 4-57. Neonatal hypoglycemia, acute and chronic phases. A. Axial CT


scan shows parietal edema (arrowheads). Note that the low attenuation of
white matter seems to continue to the inner table of the skull. B. Axial T2
weighted image shows abnormal high signal intensity in the cerebral cortex,
primarily in the parietal and occipital lobes (arrows). C. Axial ADC image
(b = 700 s/mm2) shows reduced water motion (hypointensity, arrows) in the
posterior cortex. D and E. Follow-up sagittal and axial 3 weeks after injury
shows marked necrosis and atrophy (arrows) in the parietal and occipital
cortex.

Barkovich_Chap04.indd 312 5/25/2011 11:39:16 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 313

FIG. 4-58. Neonatal hypoglycemia, subacute phase. A.


Sagittal T1-weighted image shows abnormal hyperintensity
(black arrows) of the parietal and occipital cortex. B. Coronal
T2-weighted image shows abnormal hyperintensity (small white
arrows) of the parietooccipital cortex and underlying white
matter. Note that the more anterior parietal cortex (large black
arrows), superiorly, has normal signal intensity. C. Axial Dav map
shows hypointensity (reduced diffusivity, white arrows) in the
occipital white matter.

BILIRUBIN ENCEPHALOPATHY becomes apparent during the first postnatal year. Extrapyramidal signs
(especially athetosis), abnormalities of vertical and horizontal gaze,
(KERNICTERUS) and hearing deficits typically develop after the age of 1 year (406,485).
Although the relationship between serum levels of unconjugated bili- It is not clear precisely how the bilirubin gets into the brain. It
rubin and brain damage is complicated, it is generally accepted that appears that bilirubin can be transported across an intact blood–brain
sustained or pronounced neonatal hyperbilirubinemia results in brain barrier; it also appears, however, that disruption of the blood–brain
damage (485,486). Hyperbilirubinemia may be the result of a number barrier, whether by hyperosmolarity, hypercarbia, hypoxia-ischemia,
of predisposing factors. Red blood cell hemolysis (secondary to blood or acidosis, facilitates the transport (485). A group of transporter mol-
group incompatibility, intrinsic defects of the red cell membrane or of ecules in neurons, glia, and capillary endothelial cells usually export the
hemoglobin, or degradation of blood from hematoma formation) is bilirubin from brain cells to the extracellular space and then across cap-
the most common cause. Other causes include polycythemia, inherited illary walls into the blood. Disturbance in the action of these transport-
or acquired defects of bilirubin conjugation, disorders of gastrointesti- ers, either due to genetic causes or due to depletion of energy sources,
nal transit, prematurity, and hormonal disturbances (485,486). may play an additional role in neuronal injury by bilirubin (485). In
Neonates with bilirubin encephalopathy manifest stupor and any event, the presence of high cellular bilirubin in neurons results in
hypotonia in the first few days of life; seizures occur in a minority injury, most prominent in certain locations of the brain. Pathologic
of affected patients. By the middle of the first week, hypertonia, with studies of children with chronic bilirubin encephalopathy have dem-
backward arching of the neck or back, develops. Delayed development onstrated damage to the globus pallidus, subthalamic nucleus, and

Barkovich_Chap04.indd 313 5/25/2011 11:39:17 AM


314 Pediatric Neuroimaging

the CA2 and CA3 regions of the hippocampus (5,406). Of interest, inconsistently show hyperechogenicity of the globi pallidi (Fig. 4-59A)
sick, acidotic, and edematous preterm infants with low serum albu- (488,489). On MRI, neonates with acute bilirubin encephalopathy
min levels can develop pallidal injury and hearing loss from bilirubin show inconsistent T1 shortening and, rarely, T2 prolongation in the
encephalopathy, even in the setting of clinically acceptable serum bili- affected regions (Fig. 4-59B and C) (488,490,491). Proton MRS with
rubin levels, possibly because they have a less mature blood–brain bar- TE of 144 milliseconds in the acute phase shows elevated myo-inositol/
rier or because their low albumin level results in higher free bilirubin Cr and Glx/Cr but no lactate (492). In the chronic phase, T2 prolonga-
levels (485,487,488). tion and atrophy can be seen in the globi pallidi, subthalamic nuclei,
No specific CT or ultrasound findings have been described in bili- hippocampi, and cerebral white matter (Fig. 4-59D), while proton
rubin encephalopathy. In our experience, transfontanelle sonograms MRS shows reduced NAA (493).

FIG. 4-59. Kernicterus. Acute and chronic stages. A. Coronal transfontanelle sonogram in a 2-week-old neonate with bilirubin
encephalopathy shows hyperechogenicity (white arrows) in the globi pallidi. B. Axial T1-weighted image in a 9-day-old infant with
bilirubin encephalopathy shows abnormal hyperintensity (black arrows) in the globi pallidi. C. Axial T2-weighted image in same
infant as (B) shows hyperintense white matter but normal appearing globi pallidi. It is not know why globi pallidi often appear nor-
mal acutely after injury. D. Coronal T2-weighted image in a 17-month-old infant shows evidence of kernicterus injury; abnormal
hyperintensity of the globi pallidi (white arrowheads), hippocampi (small white arrows), and right subthalamic nucleus (large white
arrow).

Barkovich_Chap04.indd 314 5/25/2011 11:39:18 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 315

ischemic, Fig. 4-60B–D), and abnormally elevated lactate are seen in the
BRAIN INJURY ASSOCIATED WITH brains of approximately 25% of children with congenital heart diseases
CONGENITAL HEART DISEASE before they undergo reparative surgery. Additional injuries (infarctions
Neurological deficits have been described in a large proportion of neo- in ~20%, white matter injury in ~40%, and parenchymal hemorrhage
nates with complex congenital heart disease (494–497). It is not clear, in ~10%) seem to be acquired between the preoperative MR studies and
however, what proportion of these deficits are the result of the underlying postoperative studies obtained within 2 weeks of the surgery (494,498).
disease as compared to resulting from the surgical correction of the dis- The presence of predominantly white matter injury (Fig. 4-60B–D) is
ease (33,498). Ultrasound studies have suggested the presence of cerebral somewhat surprising as, outside of the spectrum of children with con-
parenchymal disease in affected children prior to surgery (499) and this genital heart disease, isolated white matter injury is usually seen in pre-
has recently been confirmed by MRI (33,498). Recently, MRI and MRS mature infants and not in term infants. The white matter injury may be
studies at UCSF (498) and other institutions (494) have shown that cere- due to chronic hypoxia (163, 176) or, perhaps, to other factors, associ-
bral infarctions (Fig. 4-60A), cerebral white matter injury (presumably ated with heart disease, that delay brain maturation (498).

FIG. 4-60. Brain injuries associated with congenital heart disease include infarction,
ischemic cerebral white matter injury and, in the chronic phase, atrophy. A. Axial DWI
shows hyperintensity (white arrows) indicating an acute infarction in the left caudate
head and anterior putamen. B. Axial Dav map prior to heart surgery in a child with
transposition of the great vessels shows hypointensity (black arrows) behind the ventric-
ular trigones, indicating reduced diffusivity and acute injury. C. Axial inversion recov-
ery image shows abnormal T1 hyperintensity (black arrows) in the same regions that
had reduced diffusivity in (B). D. Sagittal T1-weighted image better shows the extent of
white matter injury (black arrows). The white matter injury is likely the result of chronic
mild to moderate hypoxia.

Barkovich_Chap04.indd 315 5/25/2011 11:39:18 AM


316 Pediatric Neuroimaging

choice if available. MRI better demonstrates acute cerebral ischemia


HYPERNATREMIC DEHYDRATION than CT or sonography, as well as more accurately revealing posterior
Young infants and, especially, premature infants are susceptible to fossa pathology such as retrocerebellar subdural hematomas, brain-
hypernatremia resulting from their relatively large surface area com- stem injury, and small cerebellar hemorrhages or infarctions. Although
pared to body weight (and thus increased insensible water loss through it has been argued that oxyhemoglobin, the hemoglobin form seen
the skin) and the inefficiency of water conservation secondary to their in hyperacute hemorrhage, is difficult to detect by MRI, hyperacute
renal immaturity (500). The most common cause of hypernatremia hematomas are seen by MRI as space-occupying masses; their hem-
dehydration is severe diarrhea (500,501). Affected infants typically orrhagic nature can be confirmed by the very low signal intensity on
are extremely lethargic, yet hyperirritable to stimuli. Other signs and T2 gradient-echo or susceptibility-weighted sequences. It is difficult
symptoms include muscle rigidity, hyperreflexia, and, occasionally, sei- to detect acute subarachnoid hemorrhage by MRI, although there is
zures (500,501). Brain damage is common, often with significant neu- some evidence that FLAIR sequences can be more sensitive than CT
rologic sequelae (501). [506,507]); however, the identification of subarachnoid hemorrhage is
Neurologic injury in hypernatremic dehydration results from the rarely critical in the neonate. Similarly, the difficulty of distinguishing
elevation of sodium concentration in the extracellular spaces. Fluid the signal void of intracranial air (pneumocephalus) from that of over-
moves from intracellular to extracellular space, resulting in cellular lying cortical bone is not a problem in the neonate, as pneumocephalus
shrinkage. The brain pulls away from the calvarium, resulting in tear- rarely accompanies birth trauma. Nonetheless, in the acutely ill neonate,
ing of bridging veins. In addition, the hyperosmolality within the blood the overriding factors are speed and safety, often making CT the second
may cause capillary enlargement; superimposed upon shrinkage of the imaging study of choice after ultrasound. When CT is obtained in the
vascular endothelial cells, this may predispose the affected patient to acutely ill newborn, the patient should be scanned with 3 mm slice
capillary rupture and intraparenchymal hemorrhage (502). Finally, thickness without the use of IV contrast. At this stage, the major goal is
dural venous sinus thrombosis can develop, with associated venous to rule out large, space-occupying hematomas; small subdural hema-
infarctions (503). tomas (particularly at the falcotentorial junction) are common after
Neuroimaging studies in hypernatremic dehydration reflect the vaginal deliveries and are of no clinical significance (508,509). Acute
pathologic findings of interstitial edema and focal hemorrhages. On interhemispheric and posterior fossa subdural hematomas are seen
sonography, the interstitial edema is seen as hyperechogenicity with on transfontanelle sonography as mildly echogenic fluid collections,
normal echo texture. Hyperacute hemorrhages are seen as focal, well- and on CT as hyperdense, extra-axial collections. Convexity subdural
demarcated regions of hypoechogenicity with a finer echogenic struc- hematomas are sometimes difficult to see on sonography because of
ture than normal brain tissue; as the hemorrhages coagulate, they the difficulty in angling the transducer adequately. Cerebral edema and
become hyperechoic. On CT and MRI, the interstitial edema is seen infarcts appear as areas of increased echogenicity in the hemispheric
as decreased attenuation and prolonged T1 and T2 relaxation times, white matter on sonography and as areas of hypoattenuation on CT.
respectively, in the cerebral and cerebellar white matter. Reduced diffu- After the patient has stabilized, or if injury is not adequately revealed
sion is seen acutely in the lateral thalami and globi pallidi (504). Focal, by CT or ultrasound in the setting of strong clinical suspicion of injury,
well-defined hemorrhages are seen in the cerebrum and cerebellum in magnetic resonance is the preferred imaging modality for assessing the
the cortex and the cortical-white matter junctions (505). full extent of damage to the brain. MRI has been proven to be more
sensitive than CT in the detection of small extra-axial fluid collections
and white matter injuries (510–516). Brainstem injuries, important
CNS TRAUMA IN INFANCY AND for determining prognosis, are better detected by MRI (514). In order
CHILDHOOD to detect hemorrhage with maximal sensitivity, T2 gradient-echo or
Pediatric trauma can be divided into two distinct groups. Birth trauma susceptibility-weighted images should be obtained in all trauma or
is composed of those injuries that occur during and as a result of the suspected trauma cases. Diffusion imaging can also be useful to detect
birth process. This form of trauma is unique in its pathological and acute injury in the watery neonatal brain; ADC images should always
radiographic manifestations because of the immaturity of the CNS and be calculated to detect subtle injuries to the parenchyma.
of the surrounding calvarium and spinal canal. Birth trauma will there-
fore be dealt with in some detail in this section. Trauma that occurs Spinal Cord Injury
later in life (postnatal trauma) results in pathological and radiological Various anatomical differences increase the susceptibility of infants to
exams that are similar to those in the adult. Since most neuropathol- spinal cord injury. The interspinous ligaments, posterior joint capsule,
ogy and neuroimaging textbooks discuss the adult type of trauma, only and cartilaginous end plates are elastic and can be redundant, making
those aspects unique to children, such as nonaccidental trauma, will be the pediatric spine more deformable than the more rigid adult spine.
discussed in detail. Hypoxic-ischemic injury is sometimes considered Moreover, the planes of the facet joints are horizontally oriented (in
a form of birth trauma. However, as hypoxic-ischemic injury was dis- contrast to the more vertically oriented adult facet joint), creating
cussed earlier in this chapter, it will not be duplicated here. more mobility and less stability. Thus, infants are more susceptible to
hyperextension injury, which is most common in the cervical region
(517–521). Ligamentous laxity also makes the pediatric spinal cord
Birth Trauma
more susceptible to distraction injuries, as the neonatal spinal column
Ultrasound is the initial imaging modality of choice for the evalua- can be stretched 2 inches without structural disruption, whereas the
tion of birth trauma because the scans can be obtained in a rapid fash- spinal cord ruptures after being stretched 0.25 inches (522); as a result,
ion without moving the patient; as a result, the patient can be closely the neonatal spinal cord may rupture but the spinal column remains
monitored during the exam. If the neonate has neurologic signs that intact (523). Distraction injuries are most common in breech deliver-
are unexplained by the ultrasound findings, further imaging should ies and tend to occur in the lower cervical and thoracic regions (Figs.
be obtained. Although CT is adequate for assessment of injury, it has 4-61 and 4-62) (522,524–526). In cephalic delivery, injury to the upper
the disadvantage of radiation exposure. Therefore, MRI is the study of or midportion of the spinal cord is more common (5,523,527,528)

Barkovich_Chap04.indd 316 5/25/2011 11:39:19 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 317

although injury at any level is possible and the involvement of multiple When injury to the spinal canal or spinal cord is suspected, MRI
levels is not uncommon. Spinal cord injuries that have been attributed is the imaging modality of choice in the acute, subacute, or chronic
to birth injury include contusions, infarctions, lacerations, transections stage (531,532), although in experienced hands, sonography may be
(Fig. 4-61), dural disruption (Fig. 4-62), and subdural and epidural useful (533,534). Sagittal, T1-weighted images should be obtained with
hematomas (Figs. 4-63 and 4-64) (522,524,529). no greater than 3 mm slice thickness; these images will show subacute
The clinical presentation depends upon the level and extent of blood and any structural deformity of the spinal cord or canal (Fig.
injury. Severe injuries at the cranio-vertebral junction cause death 4-63 to 4-66). Sagittal, T2-weighted images should also be obtained;
immediately. Incomplete injuries may allow survival. Mildly or mod- fast/turbo spin-echo images are optimal because they are fast and
erately injured patients may have low Apgar scores, respiratory distress, will usually show blood (Fig. 4-66). It is a good idea to acquire a T2
or hypotonia. Prognosis is related to the severity and level of the injury; gradient-echo sequence to look for subtle hemorrhage, the presence
high cervical cord injuries carry the worst prognosis because of the of which suggests a guarded prognosis. The T2-weighted images will
effect on diaphragmatic motion and, consequently, ventilation (530). help to identify areas of edema or ischemia, which will have long T2
Differential diagnosis includes amyotonia congenita (Oppenheim dis- relaxation times, and areas of hemorrhage, which will have short
ease) or infantile spinal muscular atrophy (Werdnig-Hoffman disease). T2 relaxation times (Fig. 4-66). The worst prognosis for subsequent

FIG. 4-61. Birth trauma secondary to difficult breech delivery; sequential scans. A. Sagittal T2-weighted image shows abnor-
mal hyperintensity (white arrow) of the spinal cord at the C-2 level and retropharyngeal edema (white arrowheads). B. Axial
T2-weighted image more clearly shows hyperintense edema (arrows) in the spinal cord. C. Sagittal T1-weighted image several
days later shows narrowing of the cord at C-2 and hypointensity developing ventrally (arrows). D. Sagittal T2-weighted image
at the same time as (C) shows central hypointensity (black arrow), indicating hemorrhage within the spinal cord.

Barkovich_Chap04.indd 317 5/25/2011 11:39:19 AM


318 Pediatric Neuroimaging

FIG. 4-62. Birth trauma with spinal injury. A. Sagittal T2-weighted imaging shows the fracture-dislocation (large white arrow) through the C-5 vertebrae,
with spinal cord compression and complete ligamentous disruption. B. Axial T2-weighted imaging shows the abnormal T2 prolongation in the spinal cord and
the marked subcutaneous and deep soft tissue injury. C. Axial T2 gradient-echo image shows hypointense intramedullary hemorrhage (low intensity, arrow).

spinal cord function is in patients who have intramedullary hemor- cord damage (Figs. 4-63 and 4-64). Sonography will show injury as
rhage; the next most severe is in those with edema extending over more hyperechogenicity and swelling of the cord in the acute and subacute
than one segment. The best prognosis is in those patients without hem- phases; regions of hemorrhage are more echogenic than regions of
orrhage or edema and in those with edema involving one segment or edema (533,534). In the chronic phase, atrophy will be detected by
less (535–539). MRI performed in the chronic phase, several months focal narrowing or, in severe cases, complete interruption of the spinal
after the event, can be helpful to determine the full extent of spinal cord. The importance of sonographic findings with respect to long-
term outcome has not been established.

Nerve Root and Brachial Plexus Injuries


Injuries to the nerve roots as they exit the spinal cord are relatively com-
mon, occurring in 0.3 to 3.6 of every 1000 live births (540); although
these are generally categorized as brachial plexus injuries, the injury most
commonly occurs to the roots that form the trunks of the plexus (541).
Injuries range in severity from mild stretching of the nerve, resulting in
transient neurologic dysfunction, to complete avulsion from the spinal
cord with resultant permanent deficit. The precise nature of the injury
depends upon the precise direction of the traction; most result from
traction of the shoulder while delivering the head of an infant in the
breech presentation or from turning the head away from the shoulder
in a difficult cephalic presentation of a large infant (542,543). Shoulder
dystocia and birth weight of greater than 3500 g are present in more
than half of cases (542) (cesarean deliveries have a lower rate of brachial
plexus injuries than do vaginal deliveries [544]). Clinically, Erb palsy
(adducted and internally rotated shoulder, extended, pronated elbow,
and flexed wrist resulting from injury to C-5, C-6, and C-7 roots) is
the most common presentation, with Klumpke palsy (extended wrist
and fingers, often associated with ipsilateral Horner syndrome resulting
from injury to C-8 and T1) considerably less common (530,541). The
majority of children with birth-related brachial plexus injury improve.
Although older studies suggest that up to 10% will go on to surgery
(545), more recent work indicates lower spontaneous improvement
rates and 10% to 15% permanent disability rate (546,547). At UCSF
and CHOP, these infants are monitored for 6 months; if no improve-
ment is seen or if improvement plateaus, surgery is performed.
FIG. 4-63. Birth trauma. Spinal and intracranial extraparenchymal hema- The brachial plexus is most sensitively assessed by MR neurog-
tomas. Sagittal T1-weighted image shows hyperintense extraparenchymal raphy, looking for disruption of the individual roots and trunks of
blood in the dorsal spine (open white arrows), posterior fossa subdural
the plexus (540,548,549). Coronal and axial STIR sequences, precon-
space (small solid black arrows) and interhemispheric subdural space (open
curved black arrows). (This case courtesy Dr. Chip Truwit, Minneapolis, trast T1-weighted spin-echo images, and postcontrast fat-suppressed
MN.) T1-weighted spin-echo sequences are acquired (see Chapter 1 for

Barkovich_Chap04.indd 318 5/25/2011 11:39:20 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 319

FIG. 4-64. Spinal epidural hematoma from birth


trauma. A. Sagittal T1-weighted image shows abnormal
heterogeneous hyperintensity within the spinal suba-
rachnoid space. B. Sagittal T2-weighted image shows
an irregular layer of hypointensity (black arrows) in the
dorsal aspect of the spinal canal, strongly suggestive of
acute hemorrhage. Note the presence of edema (white
arrows) in the spinal cord. C and D. Axial T2-weighted
images show the hypointense dorsal subdural hematoma
(open black arrows) compressing the edematous spinal
cord (solid black arrows). (This figure courtesy Dr. Erik
Gaensler, San Francisco, CA.)

exact technique). Abnormalities seen include pseudomeningoceles, image well the exiting nerve (550). Identification of a posttraumatic
enhanced, thickened nerves suggesting neuroma or scar, and low-lying, neuroma has a high sensitivity and specificity in determining lateral-
“sagging” nerve roots suggesting avulsion without pseudomeningocele ity of injury but cannot reveal the exact area (i.e., trunk or division)
formation (540) (Fig. 4-65). Avulsion of the roots may be seen on neu- involved (550). Thus, MRI, which reliably shows the brachial plexus
rography as abruptly terminating, thickened roots within a pseudo- and pseudomeningocele sac, in combination with MR neurography
meningocele. Avulsed roots may also be seen on fat-suppressed RARE and/or CT myelography, which shows the presence or absence of the
images (Fig. 4-66) or by CT myelography, which demonstrate empty individual nerve roots, is the study of choice in affected patients.
(without a nerve root) small pseudomeningoceles at the site of the
avulsions. However, conventional myelography has essentially no role
Head Trauma
in the modern radiologic evaluation of these patients. Thin section T2-
or T2*-weighted MR images, obtained in the coronal or axial planes, Extracranial Birth Trauma
will show the pseudomeningocele as an ovoid mass of hyperintensity Caput succedaneum is one of the three major varieties of extracranial
extending from the surface of the spinal cord to or through the affected hemorrhage, the other two being subgaleal hemorrhage and cephalo-
neural foramen; as little CSF motion will be present in the pseudo- hematoma. Caput succedaneum refers to a hemorrhage and edema
meningocele, thin section (2–3 mm) RARE sequences demonstrate within the skin that is observed very commonly after vaginal delivery.
the pathology well (Fig. 4-66). Identifying a pseudomeningocele with This edema is soft, superficial, pitting in nature, and crosses suture
MRI or MR myelography has been reported to have a low sensitiv- lines. The lesion steadily resolves over the first few days of life. Radiog-
ity but a high specificity for nerve root avulsion although it cannot raphy is neither diagnostic nor necessary (551).

Barkovich_Chap04.indd 319 5/25/2011 11:39:21 AM


320 Pediatric Neuroimaging

FIG. 4-65. Neurogram of brachial plexus injury due to shoulder dystocia.


A and B. Coronal STIR images show edematous right C-7 and T-1 nerve
roots (bright and enlarged roots, white arrows). C. Axial STIR image shows
the edematous roots/trunks (arrows) as they course through the neck.

FIG. 4-66. Nerve root avulsion in a neonate. A. Coronal fat-suppressed T2-weighted image shows a pseudomeningocele
(arrows) at the right lateral spinal canal. B. Axial fat-suppressed postcontrast T1-weighted image shows the hypointense,
nonenhancing pseudomeningocele (black arrow) extending toward the right C-7 to T-1 neural foramen.

Barkovich_Chap04.indd 320 5/25/2011 11:39:21 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 321

Subgaleal hemorrhage refers to hemorrhage subjacent to the aponeu- present. Resolution occurs in a few weeks to months (551). When
rosis covering the scalp beneath the occipitofrontalis muscle. Subgaleal seen on CT or MRI, cephalohematomas appear as crescent-shaped
hematomas present as firm, fluctuant masses that increase in size after lesions adjacent to the outer table of the skull; they are hyperdense
birth; they sometimes dissect into the subcutaneous tissue of the neck. on CT and show T1 and T2 shortening on MRI when imaged in the
Although patients can become symptomatic from these lesions second- subacute stage (Fig. 4-67). The hematoma does not cross the cranial
ary to blood loss, the lesion usually resolves over 2 to 3 weeks; imaging sutures. A few may eventually calcify; these gradually disappear over
of subgaleal hematomas is almost never necessary (551). many months of skull growth and remodeling. Occasionally, infants
The term cephalohematoma refers to a traumatic subperiosteal are imaged at age 3 to 4 months when the parents or pediatrician
hemorrhage; because the blood is beneath the outer layer of perios- discover a firm mass (the calcified hematoma) on the skull. A CT at
teum, it is confined by the cranial sutures (Fig. 4-67). Cephalohemato- this stage will show both an inner rim (calvarium) and an outer rim
mas occur in approximately 1% of live births; the incidence increases (calcified elevated periosteum) of calcification (Fig. 4-67D). The MR
markedly with the use of forceps and even more significantly with appearance of these lesions is usually that of subacute blood with high
vacuum-assisted deliveries (552). Cephalohematomas usually increase signal intensity on T1- and long T2-weighted images that does not
in size after birth and present as a firm, tense mass; they are rarely cross a suture; when imaged more acutely, hypointensity may be seen
of clinical significance, unless a complicating intracranial lesion is on T2-weighted images.

FIG. 4-67. Cephalohematoma. A. Axial CT scan shows soft tissue density overlying the skull between the lambdoid suture and
the coronal suture. Cephalohematomas do not cross sutures. Note that the outer wall of the cephalohematoma is periosteum,
which may calcify (arrows). B and C. Sagittal T1-weighted and axial T2-weighted MR images show the appearance of the acute
cephalohematoma. D. Axial CT at age 2 months show that the outer periosteal layer of the cephalohematoma has calcified.
Parents may bring these children to medical attention because of the “lump” on their head.

Barkovich_Chap04.indd 321 5/25/2011 11:39:22 AM


322 Pediatric Neuroimaging

Skull Fractures The most common skull fractures are linear and are most often
The importance of skull fracture in the setting of head trauma is localized to the parietal or frontal bones. When the bone is not dis-
unclear. While older studies indicate very little prognostic value for placed, the fracture heals spontaneously and no treatment is indicated.
fractures predicting neurological damage that results from trauma In infants, skull fractures may heal in less than 6 months. In older chil-
(553,554) and some authors reporting that skull fractures may actually dren, fractures generally heal within a year while in adults a healing
diminish injury to the underlying brain (by dispersing the force from time of 2 to 3 years is common. As fractures heal, the fracture line on
the trauma) (555), more recent, larger studies indicate that the pres- radiographs becomes less and less distinct until, eventually, the fracture
ence of a skull fracture increases the likelihood that the child has suf- may be difficult to differentiate from a vascular groove (557).
fered a clinically significant traumatic brain injury (556). Certainly, the An important difference between infants and older children and
absence of a skull fracture by no means excludes brain damage. Docu- adults is the fact that the middle meningeal artery is not embedded
mentation of fractures may be useful in the overall documentation of in the calvarium of the infant. Therefore, one need not be overly con-
trauma, however (especially nonaccidental trauma in older infants; cerned about a developing epidural hematoma when an infant sustains
see section later in this Chapter). When documentation is desired, ref- a linear fracture of the squamous temporal bone (57).
ormations in the coronal plane or 3D reformations using bone algo- Depressed skull fractures may result from pressure of the head
rithms may be valuable to detect fractures in the horizontal plane, can against the pelvis during the birth process or may be induced by appli-
be difficult to detect on routine CT scans. Alternatively, fractures can be cation of forceps (558,559). Whereas plain films may adequately visu-
confirmed by obtaining skull x-rays or by close examination of the lat- alize linear fractures of the cranial vault, CT more accurately defines
eral “scout” view from the CT scan; remember that the “scout” view may the amount of displacement in a depressed fracture. Most importantly,
be extremely valuable for the identification of skull fractures and should CT identifies space-occupying hematomas and assesses damage to
carefully studied in all head trauma cases. the underlying brain (Fig. 4-68), whereas plain radiography does not.

FIG. 4-68. Fracture and parenchymal injury secondary to


birth trauma, forceps delivery. A. Axial noncontrast CT scan
shows left parietal fracture (black arrows) and underlying
subdural hematoma (S) and small parenchymal hematoma
(white arrow). A large amount of scalp swelling and hemor-
rhage are also present. B. Axial proton density image shows
left medial parietal lobe contusion (black arrow) and periven-
tricular injury. C. Axial DWI shows reduced diffusion, mani-
fest as hyperintensity in the affected cortex.

Barkovich_Chap04.indd 322 5/25/2011 11:39:23 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 323

Depressed skull fractures are most often elevated, treated surgically or neonates. Subarachnoid hemorrhage is common in the neonate. When
noninvasively (560). seen in premature neonates, it is almost always the result of germi-
Occasionally, skull fractures are associated with tears of the under- nal matrix hemorrhage that ruptures into the ventricular system. In
lying dura. When dural tears occur, meninges and brain tissue can her- term neonates, it is almost always found in association with subdural
niate into the diastatic fracture site. The interposition of the meninges hemorrhage, but the pathogenesis is unknown; clinical consequences
between the diastatic bones prevents the osteoblasts from migrating are rare unless the hemorrhage is extensive, in which case it may lead
across the fracture site and inhibits healing of the fracture. Moreover, to hydrocephalus (551). Intraparenchymal hemorrhage is less common
the constant CSF pulsations upon the fracture cause enlargement of the than extraparenchymal hemorrhage and is typically associated with
fracture and extracranial extension of meninges. This situation has been venous injury or venous thrombosis.
called a “growing fracture” or a leptomeningeal cyst (557,561,562). Lep- Subdural hemorrhage is common in vaginally delivered neonates,
tomeningeal cysts are seen in 0.6% of all fractures, with 90% occurring and is probably caused by lacerations of subdural veins near the fal-
in patients less than 3 years old (561,562). On imaging studies leptom- cotentorial junction that result from molding of the head. MRI has
eningeal cysts have distinct bony margins at the fracture site (512,557), greatly increased the knowledge concerning the frequency and sever-
simulating a lytic calvarial lesion if the associated abnormalities of the ity of subdural hemorrhage in newborns. Whereas it was previously
subarachnoid space and underlying brain are not appreciated (563). believed that posterior fossa subdural hematomas in infants were
Intracranial tissue or CSF may be seen extending between the edges of rare and often life threatening, we now know that small falcine and
the bone at the fracture site (Figs. 4-69 and 4-70); this finding may be posterior fossa subdural hematomas are common in the postnatal
useful to make the diagnosis early in the course of the injury, before the period (Fig. 4-71), reported in 26% to 63% of uncomplicated vaginal
fracture begins to progressively enlarge (564). Encephalomalacia of the deliveries (508,509,568). They are usually small (<3 mm) and are rarely
underlying brain is usually present, in our experience, appearing as an of clinical importance (508,509). Indeed, most resolve by the end of the
area of low attenuation on CT (Fig. 4-70) and as an area of prolonged first postnatal month and nearly all resolve by 3 months. There are no
T1 and T2 relaxation time on MRI (Fig. 4-70). developmental sequelae unless they are associated with parenchymal
injury (i.e., venous infarction due to venous thrombosis) or result in
Traumatic Intracranial Hemorrhage in the Newborn sufficient compression to impair CSF flow or brainstem function; in
Mechanical trauma to the infant’s brain during delivery is some- the latter cases, they are surgical emergencies (569,570). There are four
times seen in association with prolonged or precipitous delivery, major categories of neonatal subdural hemorrhage: (i) tentorial lacera-
vaginal breech delivery, instrumented delivery, use of forceps or ven- tion, (ii) occipital osteodiastasis, (iii) falx laceration, and (iv) rupture
touse extraction (565). Most typically, the result is subdural hemor- of bridging superficial cerebral veins.
rhage; epidural, subarachnoid, and intraventricular hemorrhage may
also occur, but are almost always in conjunction with subdural blood Tentorial Laceration and Occipital Osteodiastasis
(551,566). Epidural hematoma is quite rare and usually is associated These two injuries are discussed together because both result in pos-
with a large cephalohematoma and a subjacent skull fracture (567). terior fossa subdural hemorrhage. Large tears of the tentorium result
Patients typically present with increasing intracranial pressure/bulg- in rupture of the vein of Galen, straight sinus, or transverse sinus;
ing anterior fontanel (551). Intraventricular hemorrhage is common the resulting subdural hemorrhage tends to be massive. The rapidly
in the premature neonate, in which it is almost always the result of expanding subdural collection can cause compression of the brain-
germinal matrix hemorrhage that ruptures into the ventricular system, stem and result in death (569). Lesser degrees of infratentorial sub-
as was discussed in the section on brain injury in prematurely born dural hemorrhage may result from smaller tentorial tears or rupture of

FIG. 4-69. Leptomeningeal cyst. A. Plain film shows a well marginated lucency (arrows) with a sclerotic border at the site of a
previous skull fracture. B. Axial CT image shows brain herniating through a dural defect and causing erosive changes in the bone at
the fracture site. Encephalomalacia (arrows) is present in the underlying brain.

Barkovich_Chap04.indd 323 5/25/2011 11:39:23 AM


324 Pediatric Neuroimaging

FIG. 4-70. Leptomeningeal cyst. A. Axial SE T1-weighted image shows encephalomalacia (curved black arrows) in the left temporal lobe with a well-
demarcated defect (curved white arrows) in the adjacent bone. B. Axial T2-weighted image better demonstrates CSF extending into the fracture site (arrows).
C. Bone window of a CT image shows the heaped-up bone edges (arrows) at the fracture site. (This case courtesy Dr. Chip Truwit, Minneapolis, MN.)

FIG. 4-71. Small posterior subdural hematomas in neo-


nates. A. Sagittal T1-weighted image shows an incidental
small retrocerebellar subdural hematoma (white arrow).
B and C. Sagittal (B) and axial (C) T1-weighted images
of a different patient show slightly more extensive benign
hemorrhage, shown as hyperintense subdural blood pos-
terior to the cerebellum (curved arrows) and along the
tentorium cerebelli (small solid arrows) and posterior
falx cerebri (open arrow).

Barkovich_Chap04.indd 324 5/25/2011 11:39:24 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 325

smaller infratentorial veins without tentorial tears (Fig. 4-71). As stated this appearance results from the presence of deoxyhemoglobin in the
above, these small subdural collections are being recognized more fre- red blood cells (575–578). RARE sequences are less sensitive to the
quently as a result of the increased sensitivity of MRI and are usually presence of blood products but they will usually be hypointense (Fig.
of no clinical consequence. The falx cerebri or supratentorial veins may 4-72G). As the hematoma evolves, it develops high signal intensity that
be torn at the same time, resulting in simultaneous supratentorial and starts in the periphery on the T1-weighted spin-echo sequences and
infratentorial subdural hematomas (Fig. 4-71) (508,570,571). progresses centripetally toward the center of the hematoma. The high
Occipital osteodiastasis consists of traumatic separation of the signal intensity, which appears first on T1-weighted spin-echo images
squamous portion of the occipital bone and the exoccipital portion of and later on T2-weighted spin-echo images, results from the conver-
the occipital bone (at a site called the posterior occipital or supraoc- sion of deoxyhemoglobin to methemoglobin. The hematoma remains
cipital-exoccipital synchondrosis) during birth. This can be seen on largely hypointense on DWI and ADC images at this stage (578). A
lateral radiographs or on CT with bone windows (572). In severe cases, fluid-fluid level may be seen at this phase of hematoma evolution, as
the dura and occipital sinuses are torn; the result is cerebellar lacera- the intact red blood cells remain in a dependent position (intermediate
tion and a massive posterior fossa subdural hemorrhage (541,573). signal on T1-weighted images, dark on T2-weighted images) with free
The posterior fossa subdural hematoma lies under the tentorium and methemoglobin in the more superior supernatant liquid (bright on
extends laterally between the dura and arachnoid overlying the cerebel- both T1- and T2-weighted images) (579). Finally, as the blood break-
lar hemisphere. down products are reabsorbed, signal intensity on the T1-weighted
On sonography, the hematoma is seen as mildly to moderately spin-echo images diminishes until eventually the subdural collection
echogenic material lying superior to the cerebellum on the sagittal is isointense with CSF on all sequences. It is important to evaluate the
and coronal images (Fig. 4-72A and B). Both the fluid collection lying size of the ventricles in these patients because acute hydrocephalus may
beneath the tentorium and the hydrocephalus resulting from compres- develop as a result of compression of the fourth ventricle or aqueduct.
sion of the fourth ventricle and aqueduct can be detected (Fig. 4-72). It is important to remember that spine injury may occur in con-
The CT appearance of these hemorrhages in the acute phase is that of junction with brain injury (Fig. 4-73). The possibility of injury to the
a high density thickening of the affected tentorial leaf with the high cervical spine should always be kept in mind when studying patients with
density extending inferiorly, posterior to the cerebellar hemisphere traumatic delivery and either a severe skull base injury or a worse neuro-
(Fig. 4-72C and D). These lesions are usually seen well on coronal and logic exam than can be explained by the amount of intracranial injury.
sagittal views; the high density hematoma is seen immediately beneath
the tentorium (557,574). When some supratentorial veins or the falx Falx Laceration and Superficial Cerebral Vein Rupture
cerebri are also torn, a supratentorial interhemispheric subdural These two injuries are discussed together because both result in
hematoma may also be present. In the anemic infant, the acute sub- supratentorial subdural hematomas. Laceration of the falx cerebri is
dural hematoma may be isodense or even hypodense to brain because much less common than, and usually occurs in conjunction with, lac-
of the relatively low protein content of the subdural collection. The eration of the tentorium. It usually occurs near the junction of the falx
MR appearance of subdural hematomas varies with their chronicity. A with the tentorium, the source of bleeding most commonly being the
hyperacute hematoma is isointense to slightly hyperintense compared inferior sagittal sinus. The cause of both falx and tentorial tears seems
with CSF on T1-weighted images, isointense or mixed iso/hypointense to be excessive vertical molding of the head with frontaloccipital elon-
compared to CSF on T2-weighted images, hyperintense on DWIs, and gation. When laceration of the falx occurs, the subdural hematoma is
hypointense on calculated ADC images. An acute subdural hematoma usually located in the inferior aspect of the interhemispheric fissure
is isointense to brain on T1-weighted spin-echo sequences, hypoin- over the corpus callosum (551).
tense to brain on T2-weighted spin-echo and gradient-echo sequences Rupture of superficial cortical veins that bridge the dura results
(Fig. 4-72 and 4-73), and hypointense on both DWI and ADC images; in hemorrhage over the cerebral convexity. In contradistinction, an

FIG. 4-72. Tentorial and retrocerebellar subdural hematoma in a newborn. A and B. Sagittal (A) and coronal (B) sonograms
show the subdural hematoma as a hyperechogenic region (white arrows) within the tentorial incisura and below the tentorium,
pushing the cerebellum downward.

Barkovich_Chap04.indd 325 5/25/2011 11:39:25 AM


326 Pediatric Neuroimaging

FIG. 4-72. (Continued) C and D. Noncontrast CT image shows


interhemispheric subdural hematoma (arrows). Axial CT image
at the level of the fourth ventricle (C) shows the high attenu-
ation of the subdural hematoma in the left side of the poste-
rior fossa (large arrow) and behind the cerebellar hemispheres
(small arrows). Axial CT at a higher level (D) shows the subdural
hematoma (arrows) along the left leaf of the tentorium cerebelli.
E–G. Sagittal T1 (E), coronal postcontrast T1 (F), and axial
T2-weighted (G) MR images show the hematoma (white arrows)
within the tentorial incisura, pushing down on the cerebellum
and brainstem.

Barkovich_Chap04.indd 326 5/25/2011 11:39:25 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 327

FIG. 4-73. Birth trauma. A and B. Coronal sonograms show slight hyperechogenicity of the thalami, suggesting hypoperfusion injury (later confirmed).
Increased echogenicity (solid white arrows) suggesting hemorrhage is present at the tentorial incisura. Hypoechogenicity (open white arrows) is present
behind the cerebellum. C and D. Axial T2-weighted images show acute retrocerebellar (open black arrows), falcotentorial junction (solid black arrows), and
supratentorial convexity (small white arrows) subdural blood. E and F. Axial T1-weighted images shows abnormal hyperintensity of the globi pallidi and
the perirolandic cortex, as well as a subgaleal fluid collection (open white arrows). G. Sagittal T2-weighted image shows fracture dislocation of the mid-level
cervical spine (large white arrow) with prevertebral edema (small white arrows). Note the hypointense hemorrhage (black arrowheads) at the falco-tentorial
junction.

interhemispheric subdural hematoma results from lacerations of the falx The ultrasound, CT, and MR appearances and evolution of
or falcine veins. In contrast to subdural hematomas occurring later in supratentorial subdural hematomas (Fig. 4-74) are identical to those
infancy, which are more commonly bilateral, convexity subdural hemato- previously described for infratentorial subdurals. As with infratentorial
mas in the newborn are usually unilateral and accompanied by subarach- subdural hematomas, coronal views are often helpful in assessing the
noid blood. An underlying cerebral contusion may be present (580). true size and extent of subdural collections. Whereas interhemispheric

Barkovich_Chap04.indd 327 5/25/2011 11:39:26 AM


328 Pediatric Neuroimaging

FIG. 4-74. Birth trauma. A. Coronal sonogram shows a left sided sub-
dural collection (open white arrows) with mass effect on the left lateral
ventricle. The brain parenchyma looks relatively unaffected. B and C.
Axial noncontrast CT images show subdural hemorrhage along the left
leaf of the tentorium (large white arrow), in the interhemispheric fissure
near the falco-tentorial junction (small white arrow), along the left cere-
bral convexity subdural space, and in the subcutaneous and subgaleal
spaces. Note the severity of the cerebral cortical involvement, which is
not detected by the sonogram (A).

subdural collections can be seen relatively easily on sonography, con- characteristics in children that are similar to those in adults will be
vexity hematomas are more difficult to visualize because of the dif- only briefly discussed in this book. The differences in imaging findings
ficulty involved in angling the transducer to see the convexity region. between children and adults will be emphasized.
Importantly, sonography can be performed in the neonatal ICU and, CT is the initial imaging study of choice for acute head injury, to
therefore, does not necessitate transporting very ill patients. detect herniation, large hematomas, or pneumocephalus that portends
fracture of the skull base; consideration should be given to including
the upper cervical spine (occiput to C-3), an area that is highly suscep-
Postnatal Trauma
tible to injury in young infants (583). After the patient has stabilized,
Although traumatic brain injury in infants and children is very similar magnetic resonance is the preferred imaging modality for assessing the
to that in adults, the causes are very different. Severe accidental head full extent of damage to the brain. MRI has been proven to be more sen-
trauma, common in older children and adults, is rather uncommon sitive than CT in the detection of small extra-axial fluid collections and
in infants less than 2 years old. In fact, nonaccidental injury (child white matter shearing injuries (510–516). Brainstem injuries, which
abuse) is 10 to 15 times more common than accidental head trauma have important prognostic significance, are better detected by MRI
in infants less than 1 year old (581) and nonaccidental trauma is the (514). Moreover, MRI is extremely sensitive to the presence of hemo-
leading cause of subdural hemorrhage at autopsy in children less than siderin from old hemorrhage (575); the presence of old CNS injuries in
1 year of age (582). Thus, when findings of severe head trauma are association with newer ones may be important evidence in the diagnosis
found on imaging studies of infants, the suspicion of nonaccidental of nonaccidental trauma. In order to detect hemorrhage with maximal
trauma should always be raised. The imaging findings of trauma in sensitivity, T2 gradient-echo images or susceptibility-weighted images
adults are described in a number of publications (512); therefore, those (584) should be obtained in all trauma or suspected trauma cases.

Barkovich_Chap04.indd 328 5/25/2011 11:39:27 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 329

Spinal Trauma Although neurological function at presentation is the single best


Vertebral column fractures and spinal cord injuries in children are less predictor of long-term neurological outcome (595,596), children have
common than in other age groups; however, they are not rare and have a significantly greater recovery of function after spinal cord injury than
received increasing attention in the medical literature since the advent do adults (597). MRI is a useful adjunct in the evaluation of spinal
of modern imaging techniques. As discussed in the section on birth trauma, especially in children who have abnormal findings on radio-
trauma, the pediatric spinal column has anatomic characteristics that graphs or CT; unexplained instability after normal studies in the acute
create different patterns of susceptibility to injury among children, as phase; persistent or delayed symptoms; disturbances of consciousness;
compared with adults. These include increased elasticity of ligaments or distracting injuries (598–601). In addition to demonstrating spinal
and soft tissues, open epiphyses, lack of development of ossification cord injury or extramedullary hematomas, MRI may show swelling of
centers, and changes in osseous strength, shape, and size (524,585,586). prevertebral soft tissues, injury (seen as abnormal T2 prolongation) to
For example, epiphysial plates are still open in the vertebral bodies of interspinous soft tissues (Fig. 4-75) or the posterior longitudinal liga-
younger children; they start to fuse at many levels by age 8 years. Facet ment, or traumatic disc herniations, in addition to cord injury (602).
joints are relatively horizontal in infancy, becoming more vertical with These findings may affect management in patients who need surgical
ossification between ages 7 and 10 years. Another factor concerns the stabilization (596,598). In addition, as discussed in the section on neo-
large head size and the relatively poorly developed paraspinous mus- natal spinal injury, MR findings are a sensitive indicator of the severity
cles in infants. This combination, coupled with the increased elasticity of spinal cord injury. T2-weighted images will help to identify areas
of ligaments, creates a relative hypermobility that leads to a predispo- of edema or ischemia, which will have long T2 relaxation times, and
sition for cervical spinal cord injury without plain film radiographic areas of hemorrhage, which will have short T2 relaxation times. The
abnormality (519,520). The greatest amount of motion in the cervical worst prognosis for subsequent spinal cord function is in patients
spine is at C-2 to C-3 in infants and young children; thus, injury is who have intramedullary hemorrhage (539); therefore, it is recom-
most common at this level. The fulcrum of movement moves to C-3 to mended that sagittal or axial T2 gradient-echo images be obtained (see
C-4 by the age of 5 to 6 years, and to C-5 to C-6, the same as in mature Figs. 4-61 and 4-62 in neonatal spine injury section). The next most
adults, by about age 15 years (519,524,587,588). Taking children as a severe MRI sign is the presence of edema extending over more than
whole, approximately 30% of injuries involve the upper cervical spine, one segment; this is best identified on sagittal T2-weighted images. The
15% involve the lower cervical spine, 30% involve the thoracic spine,
and 20% to 25% involve the lumbar spine (587,589–591). The causes
of spinal injury also vary with age. Cervical injuries in infants and tod-
dlers usually result from falls, motor vehicle accidents, and nonacci-
dental trauma; those in children aged 3 to 10 years result from falls,
bicycle accidents, and auto-pedestrian accidents; and those after age
10 years are most often the result of sports and automobile accidents
(588). As populations spend more time in motor vehicles, the causes
and location of cervical spine injuries change. A recent study in the
United States has revealed that the majority of spinal cord injuries in
the pediatric population are due to unrestrained children in motor
vehicles accidents (592). In such groups, more fractures and cervical
spinal cord injures in young children are found in the lower cervical
spine (593). The reason for this change is not known.
As infants and young children have a propensity for spinal cord
injury, an MRI should be obtained in all infants and children in whom
spinal injury is suspected, even in the absence of plain film radiologic
abnormalities. If radiographs are obtained, the radiologist should be
aware that findings in children, especially in the cervical spine, differ
from those in adults and that films taken in the “neutral” position can
be normal despite significant injury. The mobility of the cervical spine
in children is manifest by the large atlantodental space, which can be
as wide as 5 mm in children, and “pseudosubluxation,” a term refer-
ring to the up to 4 mm of anterior motion of C-2 on C-3 and C-3 on
C-4 in up to 40% of children under the age of 8 years (594). As a key
distinguishing feature, alignment of the spinolaminar line is preserved
with pseudosubluxation but misaligned with true subluxation. The
size of the prevertebral soft tissues also differs in children, as compared
with adults. In children less than 15 years old, the retrotracheal space
normally averages 3.5 mm and the retropharyngeal space averages 7 to
9 mm. In general, the prevertebral soft tissues should not exceed two
thirds of the width of the vertebral body of C-2 (524,594), although
this can be greatly affected by the phase of inspiration and patient posi-
tioning. In the adult, the prevertebral distance is normally 5 to 6 mm
at the C-3 level (524). Edema can be seen ventrally (usually a result of FIG. 4-75. Interspinous ligament injury. Sagittal STIR through the mid-
fracture) or dorsally (as a result of ligamentous injury) and, in either line in a 3-month-old infant shows T2 prolongation in the interspinous lig-
location, local soft tissue swelling (T2 hyperintensity and mass effect) aments of the midcervical spine (arrowheads). Focal subcutaneous edema
is most worrisome. and epidural fluid are also present posteriorly.

Barkovich_Chap04.indd 329 5/25/2011 11:39:28 AM


330 Pediatric Neuroimaging

best prognosis is in those patients without hemorrhage or edema and from that of the body of C-2 (608,609). Plain radiographs in active
in those with edema involving one segment or less (535–539). Keep (not passive!) flexion and extension (performed carefully under phy-
in mind that spinal cord edema increases by about one vertebral level sician supervision) will establish the diagnosis, but may be difficult
during the first day or two after injury with the maximum extent being because of patient cooperation and rotation or superimposition of
reached 48 to 72 hours after injury (603). structures; therefore, focal CT with coronal and sagittal reformations
A great deal of literature has been concerned with the concept has become the study of choice for upper cervical spine and atlanto-
of spinal cord injury without radiologic abnormality (SCIWORA) in occipital injuries in children (Figs. 4-76 and 4-77) (610). As discussed
children (519,520). We don’t feel that classification of SCIWORA as a in Chapter 1, newer CT technologies (such as automated dose modula-
separate entity is justified, as long as the radiologist and treating physi- tion algorithms) allow spine CT to be performed at doses lower than
cian recognize the underlying concept that the immature spinal column previous standards (611). It is also important to remember that only
allows significant spinal cord injury to occur without plain radiograph MRI will reliably show an associated spinal cord or brainstem injury
abnormalities and that other studies, such as MR, should be obtained to (Figs. 4-76 and 4-77) (518,536,538,612). As discussed in the section
look for soft tissue injury in appropriate clinical settings. SCIWORA will on neonatal injury, the MRI may show edema, hemorrhage, or atro-
not be addressed as a separate entity in this text. phy, depending upon the severity and chronicity of the injury (532).
Long segments of edema, extensive intramedullary hemorrhages, and
Injuries in Young Children persistent compression of the spinal cord all are predictive of poor neu-
It is important to remember that young children, through the age of rologic outcome (595,613). Diffusion-weighted imaging and/or DTI in
about 8 years, tend to sustain soft tissue injuries without incurring the acute setting may be valuable in the evaluation of traumatic spinal
radiologically apparent fractures. Thus, dislocations, ligamentous avul- cord injury (Fig. 4-77) (614,615).
sions, subluxation without fracture, growth plate injuries, and epiphy- Rotary subluxations accompany rupture of the transverse liga-
sial separations are common. Most spinal injuries in this age group ments and can become fixed. Diagnosis of fixed rotary subluxations is
involve the cervical spine, particularly the upper segments. Injuries, made by performing CT scans with the head in a neutral position and
including fatalities caused by automobile air bags, fall into this cat- then turned 45° to the left and then 45° to the right (see section below
egory, particularly when children are improperly restrained (604,605). on torticollis/rotational injuries of C-1 to C-2). Fractures of the atlas
Upper cervical injuries include atlanto-occipital dislocations, which and axis are uncommon in younger children.
are frequently fatal (606) but, with improvements in on-scene resus- Fractures involving the thoracic or lumbar spine are uncommon
citation, immobilization, and transportation techniques, have shown in children; they tend to involve the T-11 to L-2 levels, where the more
increased rates of survival (607), and atlantoaxial dislocations, which rigid thoracic spine joins the more mobile lumbar segments. Clinical
tend to be less serious because of the more capacious spinal canal at examination for thoracolumbar fracture is reported to be reasonably
C-1 (606). A combination of atlanto-occipital dislocation and atlanto- sensitive and specific, but fractures can be missed (616). Soft tissue
axial dislocations can also be encountered. It is important to remem- injuries, involving the ligaments, cartilage, or growth plates, are more
ber that the synchondrosis between the dens and the body of C-2 does common. Soft tissue injuries can be identified with flexion/extension
not normally fuse until the age of 7 or 8 years; therefore, trauma to radiography or, more reliably, with MRI using sagittal T2-weighted
the odontoid in young children most commonly results not in a frac- images with fat saturation; on the MR sequences, soft tissue damage is
ture through the bony dens, but in a disruption of this synchondro- seen as areas of high signal on T2-weighted images (617). Spinal cord
sis, resulting in separation of the ossification center of the odontoid injury is, of course, best seen on MRI (Figs. 4-78 and 4-79). Seat belt

FIG. 4-76. Upper cervical injury in a 19-month-old infant. A.


Sagittal reformation of C-2 fracture shows fracture (large white
arrow) at the synchondrosis between the dens and body of C-2.
Note also the anterior position of the posterior arch of C-1 (small
white arrows), narrowing markedly the upper cervical canal and
the craniocervical junction. B. Sagittal T2-weighted image after
partial reduction of the fracture shows hyperintensity (white
arrow) at the fracture site. This image also shows a parenchymal
injury (black arrow) at the pontomedullary junction.

Barkovich_Chap04.indd 330 5/25/2011 11:39:28 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 331

FIG. 4-77. Atlanto-axial dissociation. A. Midline sagittal CT reformation shows an abnormally widened atlantodental interval
(small double arrowed line) and the increased distance between the dens and the clivus (large double arrowed line). B. Sagittal
STIR demonstrates abnormal T2 prolongation in the atlantodental interval with stretching of the tectorial membrane (small black
arrow), which allows the tip of the dens to contact the ventral surface of the cervicomedullary junction, disruption of the inters-
pinous ligament at C-1 to C-2 (large black arrow), and abnormal T2 prolongation in the lower cervical spinal cord (white arrow).
C. Sagittal diffusion-weighted imaging shows reduced diffusivity (white arrows) at the site of the traction injury in the lower cer-
vical spinal cord, with a more superior curvilinear hyperintense artifact. D. Sagittal ADC map confirms the abnormally reduced
diffusion in the spinal cord.

injuries, secondary to flexion-distraction (fortunately uncommon in Adolescent Injuries


the current era of shoulder and lap belt systems for all passengers), Adolescents from ages 9 to 16 years sustain spinal injuries at a rate
tend to occur at the L-2 to L-4 levels (Fig. 4-78), lower than in older ten times that of younger children. The incidence is even higher in the
patients (618). Thirty to fifty percent of patients are reported to have 16- to 24-year-old age group; in fact, patients from age 16 to 24 years
associated visceral injury (618,619). These primarily horizontal inju- have the highest incidence of spine trauma of any age group (623). As
ries may be difficult to identify on axial CT images (620,621); routine children get older, they more often injure bone rather than exclusively
reformations in the sagittal and coronal planes will increase detec- soft tissues; these injuries tend to be evenly distributed through the cer-
tion. Burst fractures are uncommon in the pediatric population, with vical levels. The C-5 to C-6 level is most frequently injured in adoles-
thoracic level lesions having a higher incidence of neurologic injury cents (623). Although spinal cord injury can be present without plain
than lumbar lesions (622). The combination of flexion-extension plain radiographic abnormalities in adolescents, bone abnormalities are seen
radiographs and MRI is the most useful combination in the diagnosis more commonly than not. Moreover, the severity of spinal cord injury
of spinal trauma. in adolescents without radiographic abnormalities tends to be less than

Barkovich_Chap04.indd 331 5/25/2011 11:39:28 AM


332 Pediatric Neuroimaging

FIG. 4-78. Spinal cord injury secondary to motor vehicle accident. Plain films
were normal. A. Sagittal T1-weighted image shows no significant abnormality. B.
Sagittal T2-weighted image shows diffuse abnormal hyperintensity of the spinal
cord. Abnormal hyperintensity is present between the spinous processes of L-2
and L-3 (arrows), indicating ligamentous injury at that site.

in those with radiograph abnormalities (524). Noncontiguous second images (512,617). Also, the radiologist should be aware that traumatic
level injuries occur in the adolescent population at a rate similar to that disc herniation in adolescents is frequently associated with fracture
of adults (∼6%) (624). of the adjacent vertebral endplate, a finding that is much more easily
The radiologic evaluation of adolescents with spine trauma is appreciated on CT and MRI than on plain radiographs (625). This
essentially identical to that of adults with a few exceptions. The treating finding is important in surgical planning (626). Although plain radio-
physicians should remain aware of the possibility of soft tissue injury graphs and CT are adequate for evaluation of bone injuries, MRI is
without associated bone injury and, therefore, should obtain flexion- essential to determine the extent of spinal cord injury (Fig. 4-79) and,
extension films, especially in younger adolescents. Sagittal T2-weighted thus, to establish a prognosis (627,628). The presence of hemorrhage
MRI with fat saturation and STIR may be used to detect soft tissue (short T2 relaxation time) or long segments (more than one vertebral
injury if flexion-extension views are judged too dangerous or difficult; level) of edema (long T2 relaxation time) implies a poor prognosis for
areas of soft tissue injury are seen as regions of high signal on these recovery of function (535–538,627,628).
Another syndrome that is relatively unique to children and ado-
lescents is posttraumatic spinal cord infarction (629–631). Typically,
patients have onset of neurological signs several hours to several days
after the injury. Paraparesis or quadriparesis and dissociated sensory
loss then ensue; prognosis is variable. Radiographs and myelography
are normal (630,631). MRI will show anterior spinal artery infarction,
manifest as T2 prolongation in the anterior half to two thirds of the
spinal cord (Fig. 4-80A and B) or as hyperintensity in the ventral gray
matter of the cord. Diffusivity will be reduced in the acute/subacute
phase (Fig. 4-80C). Imaging in the subacute phase may show enhance-
ment of the infarcted region.
Patients with certain genetic abnormalities are predisposed to spi-
nal cord injury as a result of ligamentous laxity, spinal stenosis, spinal
kyphosis, or stenosis of the foramen magnum. Included in this group
are patients with Down syndrome (see Chapter 5), mucopolysacchari-
doses (especially Morquio syndrome, see Chapter 3), achondroplasia
(see Chapter 8), Klippel-Feil syndrome (see Chapter 9), and spondy-
loepiphyseal dysplasia. These patients should be observed for neuro-
logic dysfunction and neck pain in a longitudinal fashion and should
be carefully examined after even minor neck trauma (530,632,633).

Torticollis/Rotational Deformities of C-1 to C-2


FIG. 4-79. Spinal cord injury secondary to motor vehicle accident in a
10-year-old. Radiographs were normal. Sagittal T2-weighted image shows Torticollis is a common problem of childhood. Most cases have no
focal hyperintensity (white arrow) of the lower spinal cord, indicating a radiologic abnormalities and resolve spontaneously when treated con-
contusion. No osseous or ligamentous injury was present. servatively. Rarely, the deformity persists, due to anatomic derangements

Barkovich_Chap04.indd 332 5/25/2011 11:39:29 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 333

FIG. 4-80. Spinal cord infarction; this


child developed quadriparesis after a
sibling walked on his back. A. Sagittal
T2 image on third day after onset of
symptoms shows central hyperintensity
(white arrows) in the cervical spinal cord.
B. Axial T2 image shows hyperintensity
in the ventral gray matter. C. Axial ADC
map shows the reduced diffusivity in the
region of ischemia.

that can be divided into two groups. The first group includes disorders spasm of the sternocleidomastoid muscle. Most cases occur in children
of the sternocleidomastoid muscle and locally painful neck conditions. below the age of 13 years (639). Patients with rotatory fixation typically
Fibromatosis colli, also called congenital fibrosis of the sternocleido- present with sudden onset of torticollis without antecedent symptoms.
mastoid, is a common cause of torticollis in infants that may be the Sometimes, there is a history of minor trauma.
result of injury to the muscle during birth and subsequent scarring and Diagnosis is made by CT (640,641). Plain films are difficult and
shortening of the muscle (634,635). This condition is discussed further confusing because of the inability of the patient to straighten the neck
in Chapter 7. Benign paroxysmal torticollis of infancy is a disorder of (591). The patient is initially scanned with the head in neutral position.
uncertain etiology (possibly migraine-related) that consists of brief, The scan is then repeated, first with the head rotated maximally (up
repeated episodes, but is self-limited; episodes may be preceded by cry- to 45°) in one direction, and then with the head rotated maximally in
ing, agitation, vomiting, or pallor (636). Affected infants have a strong the other direction. If rotatory fixation is present, there will be no sig-
family history of migraine headache and have shown a high incidence nificant motion of C-1 on C-2 in one direction. If the torticollis is due
of gross and fine motor delays (637). Locally painful neck conditions to other causes, some rotation will be present. Sedation may be valu-
that cause torticollis include lymphadenitis and other cervical inflam- able, as muscle spasm may preclude significant motion, and the muscle
matory processes, Chiari I malformations, and tumors of the cervical is relaxed by the sedation. If rotatory fixation is found, the patient is
spine and posterior fossa (591). treated with traction and muscle relaxants; surgery may be necessary if
The other major cause of torticollis in childhood is rotatory fixation the deformity persists, or if neurological deficits are present (642).
of the atlanto-axial joint. Rotation of the head largely relies on motion Fielding and Hawkins have divided rotatory fixation into four cat-
at the atlanto-axial joint; indeed, 50% of total neck rotation occurs at egories for the purposes of prognostication (642). Type I is the most
this level (638,639). In rotatory fixation of C-1 to C-2, the normal rota- common and has the lowest incidence of neurological impairment.
tion becomes limited, possibly due to inflammation of the synovium or Rotatory fixation is present without displacement of C-1. In Type II,

Barkovich_Chap04.indd 333 5/25/2011 11:39:30 AM


334 Pediatric Neuroimaging

rotatory fixation is associated with anterior displacement of 3 to 5 mm As with neonates, MRI (of both brain and pertinent regions of
of C-1. In Type III, anterior displacement of C-1 is more than 5 mm. the spine) should be obtained in all pediatric trauma victims in whom
In Type IV, posterior displacement of C-1 is present; this is necessarily the CT findings do not fully explain the extent of neurologic deficit
associated with deficiency of the odontoid. (532,536,612,617,627). Indeed, magnetic resonance more sensitively
assesses the full extent of damage to the brain (656). MRI has been
Back Pain in Children proven to be more sensitive than CT in the detection of small extra-
Although back pain is a common problem in adults, it is uncommon axial fluid collections and white matter shearing injuries (510–516).
in children and may be a manifestation of a serious underlying prob- Brainstem injuries, which have important prognostic significance, are
lem. Back pain may result from infectious (Chapter 11), congenital better detected by MRI (514,657). Moreover, MRI is extremely sensi-
(Chapter 9), neoplastic (Chapter 10), and traumatic causes. Most of tive to the presence of hemosiderin from old hemorrhage (575,584);
these causes are discussed in other chapters. the presence of old CNS injuries in association with newer ones pres-
Acute traumatic causes of pediatric back pain are discussed earlier ents important evidence in the diagnosis of nonaccidental trauma. In
in this chapter. Nonacute causes include spondylolysis and spondylolis- order to detect hemorrhage with maximal sensitivity, T2 gradient-echo
thesis, intervertebral disc herniation, intervertebral disc degenera- images or susceptibility-weighted images (584,658) should be obtained
tion, and Scheuermann disease. Spondylolysis and spondylolisthesis in all trauma or suspected trauma cases. DTI shows reduced water dif-
are identical in children and adults and will not be discussed here. fusivity in the acute and subacute phase and increased water diffusiv-
Scheuermann disease is a poorly understood phenomenon that is dis- ity with reduced FA of white matter pathways in the chronic phase;
cussed in both pediatric radiology and bone radiology texts and, simi- it should always be considered when MRI is performed in trauma
larly, will not be discussed here. cases (659–662). When performed with many (more than 25) diffu-
Intervertebral disc herniations in children are similar to those in sion-encoding directions, DTI finds many more injuries than FLAIR
adults with three exceptions. (a) The size of disc herniations is larger in or susceptibility-weighted images (661) and may contain important
children than in adults (643–645). (b) When traumatic, disc herniation prognostic information (663–665); DTI may also be valuable for cog-
in adolescents is frequently accompanied by fracture of the adjacent nitive outcome prediction (666,667). Proton MRS can also be useful
vertebral endplate, a finding that is much more easily appreciated on in the subacute and chronic phases to determine the severity of paren-
CT and MRI than on plain radiographs (625). This finding is impor- chymal injury (660,668). Magnetoencephalography (MEG) studies in
tant in surgical planning (626). (c) Pediatric disc herniations may be children and adults with traumatic brain injury show abnormal slow
calcified (646–648), possibly because of accompanying discitis (647); wave (theta and delta) activity, which has been shown to correlate with
the true incidence is likely underestimated (649). The presence of a neurologic function (669–671).
calcified disc does not change treatment, which is almost always con-
servative. It is important to remember two facts when a disc hernia- Extraparenchymal Hematomas
tion is seen in children. First, most children with disc herniations are Epidural hematomas are uncommon in infants and increase slowly
asymptomatic (649,650). Second, the vast majority of these patients in incidence throughout childhood to reach peak incidence in adults
recover completely without surgical intervention. (530). Epidural hematomas in younger children differ from those in
Intervertebral disc degeneration turns out to be a fairly common adults primarily in pathogenesis and clinical manifestations; prognosis
finding in children with low back pain, being seen in 30% to 50% of also appears to be better in children than in adults (672) and in younger
symptomatic patients, as compared with 20% of asymptomatic patients children than in adolescents (673). In young children, epidural hema-
(650,651). MRI shows loss of disc height and diminished signal on tomas are more commonly the result of tears in the dural veins than
T2-weighted images. Adjacent endplate changes have been reported to of laceration of the middle meningeal artery (very rarely, they may be
have a high association with low back pain (651). Although the inci- caused by venous thrombosis [124]); in older children and adolescents,
dence of disc degeneration is higher in adolescents with low back pain, arterial causes are far more common (530). The difference in clinical
the 20% incidence in asymptomatic adolescents is notable. The finding presentation results from two factors: (a) the pediatric skull is more
of disc degeneration has no impact on treatment. pliable than the adult’s and can expand as a posttraumatic hematoma
accumulates (674) and (b) intradural injury is uncommon (673). As a
Head Trauma result, the clinical presentation does not evolve as rapidly. Moreover,
In general, CT is the initial study of choice for older children who in contradistinction to adults, in whom the head injury characteristi-
have suffered head trauma, because CT is fast and easily obtained; it cally induces a loss of consciousness, children more commonly are only
is sensitive in detection of intraparenchymal and extraparenchymal stunned by the injury. This necessitates consideration of early imag-
hematomas, mass effect, and fractures; and it can be used to image ing in the pediatric trauma patient because the clinical manifestations
the spine, chest, and abdomen for traumatic imaging at the same sit- of significant trauma may initially be mild or transient. Although the
ting as the head scan. We obtain 2.5 to 3 mm thick sections from the morbidity and mortality associated with epidural hematoma were his-
vertex to the C-3 vertebral body in all pediatric trauma patients. The torically high, the outcome and prognosis following epidural hema-
reason for obtaining images in the upper cervical spine is the high inci- toma are generally very good after rapid radiological identification and
dence of cervical spine injury that accompanies head injury in chil- surgical decompression (675).
dren (521,583,608,609). Given the special concerns regarding radiation The findings of epidural hematomas in children on CT and MRI
exposure in children, groups have attempted to create a decision rule are identical to those of adults. CT will usually reveal a lentiform
by which those children with a lower risk of clinically significant brain hyperdense extraparenchymal collection that does not cross cranial
injuries could be identified, obviating the need for routine scanning sutures (Figs. 4-81 and 4-82). Skull fractures and soft tissue swelling
in children who do not meet criteria (556,652). While the US trial has are often present overlying the hematoma. Contusions may be pres-
been validated, the Canadian trial has not yet been validated at the time ent in the subjacent brain parenchyma or in a contralateral location
of this writing. Much work is also being done to identify a biomarker (contre-coup injury). If the fracture extends into the mastoid air cells
for pediatric brain injury; however, a sensitive and specific marker or or a paranasal sinus, pneumocephalus may be present (Fig. 4-82). If
panel of markers has yet to be discovered (653–655). the fracture is in the frontal bone and extends into the orbital roof, a

Barkovich_Chap04.indd 334 5/25/2011 11:39:30 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 335

FIG. 4-81. Epidural hematoma. A. Axial CT image in a 3-year-old child shows a lentiform extraparenchymal collection of high
attenuation blood (arrows) in the right anterior cranial fossa. Note that the collection does not cross the metopic or coronal
suture. B. Bone window image shows frontal bone fracture (arrow).

subperiosteal orbital hematoma may develop. Orbital hematomas may images. As with subdural hematomas, a fluid-fluid layer may be present
cause acute proptosis and result in loss of vision unless the hematoma is secondary to layering of blood cells; the upper layer will be bright on
expediently decompressed. On MRI, the hyperacute hematoma is dark T1- and T2-weighted images and the lower level will be isointense to
on T1-weighted images and bright on T2-weighted images. The acute brain on T1-weighted images and dark on T2-weighted images (579).
hematoma is bright on T1-weighted images and dark on T2-weighted Subdural hematomas are most common in infants and elderly
adults and are less common in older children and adolescents (676).
They result from tears of cortical veins bridging the subdural space on
their way to the dural sinuses. The soft consistency of the unmyeli-
nated brain results in increased brain distortion during trauma and
puts increased stress upon these veins (530). In contrast to adults, in
whom traumatic subdural hematomas are usually unilateral, traumatic
subdural hematomas are bilateral in 80% to 85% of infants and are
typically located over the frontoparietal convexity. A history of trauma
is usually elicited. The presence of recent unreported physical trauma
or a history of mild trauma that is inconsistent with the severity of the
injury suggests child abuse (530,674,677–682). Other factors predis-
posing to hematomas include blood dyscrasias and prematurity (683).
Infants with benign enlargement of the subarachnoid spaces may be
at increased risk for subdural hematoma; however, this remains con-
troversial (684–686). Infants with subdural hematomas may present
with seizures, vomiting, hyperirritability, lethargy, or progressive head
enlargement (681,687). Older children typically present with signs
of increased intracranial pressure, such as depressed consciousness,
increased systemic blood pressure with a decreased pulse rate, irregular
respiration, pupillary asymmetry, and hemiparesis (688,689). If chil-
dren have normal level of consciousness and no motor weakness or
pupillary asymmetry, the hemorrhage may be subarachnoid; if so, it
will resolve spontaneously within a few days (690).
FIG. 4-82. Epidural hematoma with pneumocephalus secondary to mas- Imaging studies of subdural hematomas in children are identical
toid fracture. Axial CT image shows a lentiform high attenuation collection to those in adults. In the acute phase, CT will show a high attenuation
of epidural blood (small arrows) overlying the left posterior temporal and extraparenchymal crescentic fluid collection over the frontoparietal
occipital lobes. The presence of extraparenchymal air (large white arrow) convexity that typically crosses cranial sutures (Fig. 4-83). Within 1 to
suggests a basilar skull fracture. 3 weeks after the hemorrhage, the blood becomes isodense with brain

Barkovich_Chap04.indd 335 5/25/2011 11:39:30 AM


336 Pediatric Neuroimaging

FIG. 4-83. Acute traumatic subdural hematoma and parenchymal contusions (from child abuse). A. Axial noncontrast CT image shows
large extraparenchymal high attenuation collection (white arrows). This acute subdural hematoma crosses cranial sutures and lies both
lateral and medial to the left cerebral hemisphere. B. Coronal FLAIR image shows the subdural hematoma (hyperintense crescentic col-
lection, arrows) but shows no evidence of parenchymal injury in the left frontal lobe. C. Axial T2-weighted image shows the subdural
hematoma to be hypointense (white arrows). Parenchymal injury is seen as cortical and white matter hyperintensity (black arrows) in the
left frontal lobe. D. DWI (b = 1000 s/mm2) shows reduced diffusivity, manifested as high intensity (black arrows) in the left lateral frontal
lobe and left posteromedial parietal lobe.

tissue. At this stage, compression of the ventricles and medial displace- described for the epidural hematoma in the previous section (Figs.
ment of the gray-white junction with compression of the white matter 4-81 and 4-83). The changes in signal intensity result from the loss of
will indicate presence of the extra-axial fluid collection (Fig. 4-84A). If oxygen from hemoglobin followed by the oxidation of hemoglobin to
intravenous contrast is given, the inner and outer membranes of the methemoglobin and subsequent breakdown to diamagnetic materials
subdural hematoma will enhance (Fig. 4-84B) (512). It is essential to (576). Fluid-fluid layers are often present as a result of layering of the
recognize shift of the ventricles, medial displacement of the corticome- blood cells in the dependent portion of the subdural collection (Fig.
dullary junction, and medial displacement of the cortical sulci at this 4-83). MRI is also useful in the identification of injury to the under-
state (557,691). Patients with visible subarachnoid spaces on the side of lying parenchyma (such as axonal shearing injuries, contusions, and
the subdural hematoma may have a better prognosis (692). After 2 to 3 infarctions, see subsequent section) that commonly accompanies trau-
weeks, the density of the hematoma will be lower than that of brain and matic subdural hematomas (Fig. 4-83 and 4-85) (555,678–680,689).
closer to that of CSF. At this stage, it is usually called a chronic subdural Unlike intraparenchymal hematomas, in which the blood–brain
hematoma. Because of the development of fibrovascular granulation barrier inhibits their resorption, hemosiderin and ferritin are not
tissue in the periphery of the subdural collection, the outer and inner deposited within the wall of the chronic subdural hematoma (512,575).
membranes of subacute or chronic subdural hematomas enhance Therefore, the absence of subdural hemosiderin does not imply that a
rather intensely after IV contrast is administered (557). On MRI, the subdural hematoma is acute or that a prior subdural hematoma has
signal intensity of the extra-axial fluid collection will evolve as those not been present.

Barkovich_Chap04.indd 336 5/25/2011 11:39:31 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 337

FIG. 4-84. Subacute (isodense) subdural hematoma. A. Axial noncontrast CT image shows compression of the right
lateral ventricle and right hemispheric white matter in addition to medial displacement of the gray matter-white
matter junction (arrows). B. After administration of iodinated contrast, part of the inner membrane of the subdural
hematoma enhances. In addition, many of the dural veins are medially displaced by the subdural collection.

Subarachnoid Hemorrhage hemorrhage may be the harbinger of more severe underlying brain
damage (677,691). Routine T1- and T2-weighted MR sequences are
Subarachnoid hemorrhage often accompanies intraparenchymal
not sensitive for the diagnosis of acute subarachnoid hemorrhage,
damage in both the child and the adult. In the acutely injured child,
probably because the hemoglobin is less concentrated than in clotted
CT is the imaging study of choice to detect the hyperdense subarach-
blood and the high oxygen tension of CSF inhibits the conversion of
noid blood. In the acute phase after injury, when nonhemorrhagic
hemoglobin to deoxyhemoglobin and methemoglobin (575,693,694).
parenchymal lesions are difficult to identify by CT, subarachnoid
FLAIR, T2 gradient-echo, and susceptibility-weighted sequences may

FIG. 4-85. Subarachnoid hemorrhage and cerebral infarction secondary to nonaccidental trauma. A. Axial CT image shows
the high attenuation of blood (small arrow) in the right occipital lobe secondary to contusion. The MCA distribution of the
left hemisphere (arrows) is hypodense secondary to infarction. B. Axial CT at higher level shows bilateral cerebral infarctions
(large arrows) and a thick irregular linear hyperdensity (small arrows) representing subarachnoid blood in the posterior
interhemispheric fissure.

Barkovich_Chap04.indd 337 5/25/2011 11:39:31 AM


338 Pediatric Neuroimaging

be sensitive to both acute and subacute hemorrhage (506,507,695,696) extension of the blood into the cerebral sulci along the medial aspect
and should be utilized to search for subarachnoid blood if an MRI is of the hemisphere (697) (Fig. 4-87). This thick, dense high signal in
obtained. Subarachnoid hemorrhage appears as high signal intensity the interhemispheric fissure has been called the “falx sign.” The reader
in the subarachnoid spaces on FLAIR images, whereas CSF is of low should note that, even in children, the falx can have a high attenuation
intensity; on T2 gradient-echo and susceptibility-weighted imaging, on CT. The falx itself, however, should be thin and regular in contour,
subarachnoid hemorrhage will appear hypointense, in comparison and no extension of the high attenuation into the cortical sulci should
with the hyperintense CSF (696). It is important to remember, how- be seen.
ever, that rapidly flowing CSF (such as that around the foramina of
Monro, the aqueduct of Sylvius, and the prepontine cistern) will com- Injury to the Brain Parenchyma
monly have high signal intensity on FLAIR images; rapidly flowing CSF In the acute phase after either physical trauma or asphyxia, generalized
should not be mistaken for subarachnoid hemorrhage. CT shows acute cerebral swelling is seen more commonly in children than in adults with
subarachnoid hemorrhage as increased attenuation in the subarach- similar types of head injury (698). This generalized cerebral swelling
noid space. Traumatic subarachnoid hemorrhage is most commonly probably results from a combination of edema and a decrease in cere-
seen in the posterior interhemispheric fissure, adjacent to the falx cere- brovascular autoregulation, resulting in vasodilatation and increased
bri (Fig. 4-86), or layering along the tentorium cerebelli (Fig. 4-87); cerebral blood volume (530,699). If an imaging study is obtained in
sometimes, this increased attenuation is the only abnormality seen on the first 12 hours after trauma, it may not demonstrate the edema. By
the CT scan. Blood along the tentorium is best demonstrated by imag- about 24 hours, CT and MRI of the brain show decreased distinction
ing in the coronal plane. When blood is located along the posterior between gray and white matter with compressed, slit-like lateral ven-
falx cerebri, the increased density within the interhemispheric fissure tricles (170,677,691); focal injury may be seen by diffusion-weighted
is thicker and more irregular than the falx itself; a helpful finding is imaging at this time (Fig. 4-86) (700,701). In addition, the cerebral sulci

FIG. 4-86. MRI of severe head trauma; brain CT was normal. A. Cor-
onal fat-suppressed FLAIR image shows two right frontal cortical con-
tusions (white arrows) and bilateral deep white matter axonal shearing
injuries (white arrowheads). B. Axial GRE T2 image shows low inten-
sity foci (arrows) in the bifrontal regions of white matter injury. These
regions also demonstrated reduced diffusion. C. Axial Dav map shows
marked hypointensity (white arrow), indicating reduced diffusivity, in a
contusion of the splenium of the corpus callosum.

Barkovich_Chap04.indd 338 5/25/2011 11:39:32 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 339

FIG. 4-87. Traumatic injury with subarachnoid hem-


orrhage, and parenchymal injury. A. Axial CT image
shows subarachnoid blood in the ambient cisterns
(large solid black arrows), interpeduncular cistern
(white arrow) and along the tentorium cerebelli (small
solid black arrows). Parenchymal hemorrhage (open
arrow), probably secondary to axonal shear injury, is
present in the left temporal lobe. B. Image at the level
of the corpus callosum shows a large hemorrhage
(arrows) at the callosal splenium. C. Image at the level
of the frontoparietal convexities shows subarachnoid
blood in medial hemispheric sulci (curved arrows) and
a widened interhemispheric fissure (straight arrows),
possible secondary to a dural rent or an adjacent
subdural hematoma.

and perimesencephalic cisterns are compressed. Transtentorial hernia- through the skull (Figs. 4-86 and 4-88). Shearing injuries result from
tion of the temporal lobe may cause infarction of the posterior cerebral rotational forces on the skull. When the skull is rapidly rotated, the
artery (PCA) territory by compressing the PCA against the free edge brain lags behind, causing axial stretching and disruption of nerve
of the tentorium in the ambient cistern. Stretching of the thalamoper- fiber tracts (512). Because the unmyelinated brain is less rigid than
forating arteries during transtentorial herniation may cause thalamic the mature brain (and consequently more susceptible to distortion)
infarcts (702). Subfalcine herniation may cause infarcts in the territory and because the subarachnoid spaces are more capacious, shear-
of anterior cerebral artery (ACA) branches (512). ing injuries are a more common consequence of rotational injury in
Parenchymal lesions resulting from head trauma include cere- infants and young children. Shearing injuries most commonly occur
bral contusions and white matter shearing injuries (561,703). Cerebral at the junction of the gray and white matter, in the deep white mat-
contusions are bruises of the brain most commonly caused by decel- ter of the centrum semiovale, in the corpus callosum, the internal
eration injuries in which the brain forcibly contacts the rough edges capsule, the basal ganglia, and the brainstem (Figs. 4-86 and 4-87)
of the skull in the anterior temporal and orbito-frontal regions (Fig. (513,555,561,678,703). Many of the brainstem lesions will be missed
4-87) (555,704). They may also result from direct impact transmitted unless MRI is obtained (657); as the presence of brainstem injuries

Barkovich_Chap04.indd 339 5/25/2011 11:39:33 AM


340 Pediatric Neuroimaging

FIG. 4-88. Trauma in a 10-month-old boy; television fell on his head. A. Axial CT scan shows hyperattenuating blood
(white arrow) in the right cerebellopontine angle cistern and hypoattenuating air (black arrow) in the left cerebellopon-
tine angle cistern. The presence of air suggests a basilar skull fracture. The temporal horn of the right lateral ventricle is
enlarged due to obstructive hydrocephalus from intraventricular hemorrhage. B. Coronal TSE T2 image shows periph-
eral and deep right cerebellar hemisphere contusions, as well as bilateral cerebellar tonsillar contusions (small white
arrowheads). An extradural hematoma overlies the lateral right cerebellar surface (white arrow). A large occipital bone
fracture is evident (large white arrowhead). C. Axial Dav map shows reduced diffusivity (hypointensity, white arrows) in
cerebellar contusion. D. Axial GRE T2*-weighted image shows a hematoma anterior to the cervicomedullary junction
(white arrow) and hemorrhage within the right tonsillar contusion (white arrowhead).

strongly correlates with duration of coma and with outcome, as CT is sensitive in the diagnosis of hemorrhagic cerebral contusion
indicated by the Glasgow Coma Scale, MRI should be considered in in the acute phase because it is sensitive to the presence of large areas
all children with severe head injury or coma (657). As discussed below, of acute blood within the brain (Figs. 4-86 and 4-88) (678,704) and to
DTI might be the best way to assess for shearing injuries in the chronic the accompanying extra-axial hematomas (Figs. 4-81 and 4-82) or air
phase, as the microstructural changes alter both mean diffusivity and (pneumocephalus) that may be present (Figs. 4-82 and 4-88A), and
FA (660,661,700,705,706). Indeed, the number of damaged white the patient is easily monitored during a CT scan. Thus, as stated ear-
matter structures, as quantified by DTI, is significantly correlated lier, it is the imaging study of choice for the evaluation of head injury
with mean reaction time on simple cognitive tasks and the presence in the acutely ill and unstable patient. However, as also stated, CT is
of abnormal slow waves on MEG; no such correlation with traumatic less sensitive than MRI in the detection of contusions (hemorrhagic
microhemorrhages was found (661,669). or nonhemorrhagic, Figs. 4-86, 4-88, and 4-89) and axonal shearing

Barkovich_Chap04.indd 340 5/25/2011 11:39:34 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 341

injuries, particularly in the subacute and chronic phases of injury if imaged at least 3 months after the injury. Interestingly, children with
(678). Therefore, MRI is the imaging modality of choice for assessment frontal lobe lesions were found to be more frequently neurologically
of parenchymal injury. In the acute phase, diffusion images are most and psychologically disabled than those with diffuse injury (711).
sensitive for the detection of contusions and shearing injuries (Figs. When imaged in the chronic phase, 3 months after injury, the finding
4-86 and 4-88) (700, 701). Hyperacute hemorrhage (in the first hours of brainstem injury (T2 prolongation from astrogliosis or T2 shorten-
after injury, when blood largely remains in the oxyhemoglobin con- ing from hemosiderin) portends a poorer prognosis (712).
figuration) will appear on diffusion images as hyperintensity and on As indicated in the previous paragraphs, recent evidence sug-
Dav images as hypointensity, whereas acute hemorrhage (in which gests that MR DTI, MRS, and susceptibility-weighted imaging may
blood is mostly in the deoxyhemoglobin form) will appear on both have a role in the acute phase to differentiate swelling from acute
types of images as hypointensity (578). In the subacute and chronic axonal shear injury (660). Indeed, in the chronic phase, DTI and
phases, T2-weighted scans or a combination of FLAIR and T2 gradi- MRS correlate better with clinical outcome than any anatomic imag-
ent-echo or susceptibility-weighted images are the best choices; these ing sequences (661,705). Diffusion imaging (Figs. 4-83 and 4-88)
methods are sensitive to both the nonhemorrhagic and hemorrhagic demonstrates reduced water diffusion in injured regions of brain
injuries that are present at the site of prior contusions and shear inju- (increased intensity on diffusion images and decreased intensity on
ries (511,516,678,704). Gradient-echo studies with long-echo times Dav images) in the first hours after injury, whereas interstitial edema
(20–30 milliseconds) and susceptibility-weighted images (584,658) and increased blood volume show increased diffusion (decreased
are even more sensitive than T2-weighted spin-echo studies for detect- intensity on diffusion images and increased intensity on Dav images).
ing products of hemorrhage (Figs. 4-88D and 4-90) and, therefore, are Diffusion imaging may detect abnormalities (manifested as reduced
an essential adjunct in the MR examination of trauma victims. Lower diffusion) more than 2 weeks after injury (659). In addition, stud-
numbers of lesions on susceptibility-weighted images seem to cor- ies suggest that reduced FA, as assessed by DTI, may be an early and
relate well with better neurological outcome (660). RARE (fast/turbo more sensitive method of detecting axonal injury (661,700,705,713).
spin echo) and FLAIR sequences are relatively insensitive to the pres- In the chronic phase of injury, the presence of reduced anisotropy in
ence of intraparenchymal blood (Fig. 4-89) at 1.5 T and should not be specific white matter tracts correlates with the Glasgow Coma Scale
relied upon to exclude parenchymal hemorrhage in the setting of trauma. (705) and measures of learning and cognitive performance (661,706).
FLAIR is useful to assess the full extent of white matter injury in older Proton MRS may be useful in differentiating swelling secondary to
children but, in neonates and young infants, FLAIR may be insensi- increased blood volume from swelling secondary to shearing injury.
tive to parenchymal injuries of all kinds (Figs. 4-83 and 4-90). If the Proton MRS is nearly normal in the absence of parenchymal injury
MRI is performed at 3 T, acute and subacute blood may be seen as but shows decreased NAA and increased choline, lactate and gluta-
hypointense on RARE and FLAIR sequences (707); therefore, it is pos- mate/glutamine within the first 2 to 4 days after injury if significant
sible that gradient-echo sequences may be unnecessary if the scans are neuronal injury has occurred (714–718). Indeed, animal studies indi-
performed at 3 T, but further work is necessary before these sequences cate that NAA is reduced as early as 1 hour after injury (719,720).
are eliminated. Subacute hemorrhagic injuries will often show areas NAA/Ch levels seem to correlate well with cognitive outcome, par-
of high intensity on both T1- and T2-weighted images because of the ticularly when the frontal lobes are sampled (721) and lower NAA/
presence of methemoglobin and edema, respectively. Chronic injuries Ch ratios, higher Ch/Cr ratios, and the presence of lactate all cor-
will appear as hyperintense areas on FLAIR images; on T2-weighted relate with poor outcome (660,718,722). Indeed, proton MRS shows
spin-echo images, they may show high signal intensity (astrogliosis, reduced area under the NAA peak even in patients with mild brain
which has more water than normal brain), low signal intensity (resid- injury and normal MRI studies (668). Susceptibility-weighted images,
ual blood breakdown products from previous hemorrhage, Fig. 4-89), a high spatial resolution, 3D gradient-echo technique in which alter-
or both. Both subacute and chronic hemorrhagic injuries have low sig- ations of phase are amplified (723) is even more sensitive to blood
nal intensity on gradient-echo scans with long-echo times (Fig. 4-90) products than standard gradient-echo images, showing tenfold more
and on susceptibility-weighted images (584). Lesions in characteristic hemorrhagic lesions in one study (584,658) and correlations have
locations, such as the orbitofrontal surface of the frontal lobes and the been shown in small groups between number of lesions detected by
anterior temporal lobes, are strongly suggestive of prior trauma. More- susceptibility-weighted images (SWIs), location of lesions, and clini-
over, the location and number of injuries within the brain may be of cal outcome (658,660). SWI may become the sequence of choice for
prognostic value in terms of how much recovery of normal brain func- initial assessment of shearing injuries.
tion can be expected. (Prognosis is probably predicted best by the use
of advanced imaging methods, as described below.) Sequelae of Trauma
Severe swelling of the brain suggests a guarded prognosis (708,709) The sequelae of trauma include infarction from severe brain edema
as does the presence of the “reversal sign” in which cerebral white mat- and vascular injury, infections, leptomeningeal cysts, inflammation,
ter has a higher attenuation than cerebral gray matter (Fig. 4-54) and hydrocephalus (674,689,724). The imaging appearance of these
(456,457). It is difficult in this acute phase to detect axonal shearing conditions is discussed elsewhere in this book and will not be discussed
injuries by CT. Therefore, one cannot determine whether diffuse cere- extensively here.
bral swelling is the result of increased blood volume or axonal shearing Infarctions from severe brain edema and leptomeningeal cysts
injuries (530,710). Proton MRS and DTI may have a role in making this secondary to skull fractures are described earlier in this chapter.
determination (see the last paragraph of this section); in their absence, Hydrocephalus frequently develops after head injuries, possibly as
follow-up scans are necessary. Atrophy will develop if the brain has a result of inflammation and subsequent adhesions caused by suba-
suffered significant injury but the swollen brain will return to a normal rachnoid blood. In children, it is sometimes impossible to differenti-
appearance if significant neuronal damage has not occurred (530,710). ate communicating hydrocephalus from cerebral atrophy, which can
Follow-up standard MR scans of children with mild head injury will also result from trauma, by imaging. Both can appear as dilatation
be normal at 3 months after the injury. Of children with moderate to of the cerebral ventricles and sulci on imaging studies and both can
severe closed head injury, and, therefore, a poorer prognosis, 71% will result from severe cerebral injury (725). Differentiation can sometimes
have parenchymal lesions on MR, predominantly in the frontal lobes, be made by careful analysis of the contours of the third ventricle (see

Barkovich_Chap04.indd 341 5/25/2011 11:39:34 AM


342 Pediatric Neuroimaging

FIG. 4-89. Trauma in a 3-year-old boy; value of T2-weighted images, FLAIR, and diffusion imaging in detecting hemorrhagic injury.
A. Axial CT scan shows areas of hypoattenuation (white arrows) in the orbital surface of the right frontal lobe. Blood is identified
only in the right temporal lobe contusion (white arrowhead). B. Axial FLAIR image shows more contusions and blood (low intensity,
arrow) in the large injury in the right orbital surface. C. Axial T2-weighted image shows many more foci of low attenuation hemor-
rhage bilaterally in the frontal lobes. D. Axial diffusivity (Dav) image shows little damage, probably because of distortion due to sus-
ceptibility artifact from the skull base and blood.

Barkovich_Chap04.indd 342 5/25/2011 11:39:34 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 343

FIG. 4-90. Child abuse in a 2-month-old infant; value of gradient-echo images. A. Axial T1-weighted image
shows large bilateral subacute subdural hematomas with an area of more acute blood clot (curved white arrow). No
definite parenchymal injury is seen. B. Axial FLAIR image is not very useful for identifying brain injury in infants.
C. Axial RARE (FSE/TSE) image shows some evidence of blood (white arrows) in the left posterior temporal
lobe. Note the fluid-fluid layer (black arrows) in the posterior right subdural space. D. Axial gradient-echo image
(TE = 25 milliseconds, q = 15°) image shows multiple foci of hemosiderin (open black arrows), unequivocal
evidence of prior injury.

Chapter 8). Alternatively, diagnosis can be made by monitoring intrac- hydrocephalus will show neurologic improvement after placement of
ranial pressure or by a radionuclide flow study with 99mTc DTPA. The a shunt (674).
technetium compound is injected into the spinal subarachnoid space Vascular complications of head injury include carotid-cavernous
via lumbar puncture. If flow of the radionuclide upward over the cere- fistulas, arterial dissections, and venous sinus occlusions. Signs and
bral convexities is not seen by 24 hours after its injection, or if the radi- symptoms of carotid-cavernous fistulas include pulsating exophthal-
onuclide is seen within the lateral ventricles at 24 hours after injection, mos, deficient ocular motility from multiple cranial nerve palsies, and,
there is very likely a block to CSF flow and hydrocephalus (726,727). sometimes, blindness or subarachnoid hemorrhage. Catheter angiog-
Of course, the most important features are clinical: an enlarging head raphy is necessary for both diagnosis and treatment (see Chapter 12).
size and sutural splitting indicate hydrocephalus, whereas a decreas- Arterial dissections can present with obtundation, mono- or hemipare-
ing head circumference (compared to the normal distribution on head sis, dysphasia, or Horner syndrome (see Chapter 12). T1-weighted MRI
growth charts) suggests atrophy. Identification of hydrocephalus in of the skull base with fat saturation is very sensitive in establishing the
this setting is important, as up to 75% of patients with posttraumatic diagnosis of dissection. A crescentic rim of hyperintensity, presumably

Barkovich_Chap04.indd 343 5/25/2011 11:39:35 AM


344 Pediatric Neuroimaging

methemoglobin, will be seen in the vessel wall. MRA or CTA may show “shaken baby syndrome” (741–743), characterized by retinal hemor-
intimal irregularities that allow the diagnosis to be established. In some rhages, subdural or subarachnoid hemorrhage, cerebral contusion, and
clinical settings, MR diagnosis of dissection may obviate the need for diffuse cerebral edema with minimal evidence of external trauma. The
catheter angiography. CT or MRI is essential to diagnose associated American Academy of Pediatrics recommends use of the term “abusive
cerebral infarction. Venous sinus occlusion can be diagnosed either by head trauma,” rather than describing a single injury mechanism, in the
MRV, CT venography, or intravenous digital subtraction angiography, medical record when encountering the constellation of findings that
as described in Chapter 1 on techniques and Chapter 11 (“Complica- can result from inflicted injury (744).
tions of Meningitis” section).
Infection is an uncommon complication of brain injury. When Mechanisms of Injury
infection does occur, it is usually in the form of meningitis, second- The injuries in child abuse victims can be the result of direct trauma,
ary to bacterial seeding from basilar skull fracture, or cerebritis/abscess shaking injuries, strangulation, or a combination thereof (735). How-
from penetrating injuries. The latter is extremely rare in children and ever, the precise mechanisms by which the injuries occur and the forces
is diagnosed from post contrast CT or MR examinations (see Chap- needed to produce them are not entirely clear due to the lack of a sat-
ter 11). To diagnose a CSF leak from a basilar skull fracture, 3 to isfactory model and the lack of good data in describing mechanisms of
5 mL of nonionic iodinated contrast is injected into the subarachnoid injury in identified cases (745,746). Indeed, most studies have been ret-
space via lumbar or C-1 to C-2 puncture with the patient prone. The rospective, lacking controls, and no definition of nonaccidental trau-
patient is then kept prone and tilted 45° (with head down) for 60 to matic brain injury or shaken baby syndrome for research studies has
90 seconds. The patient is maintained in the prone position while being yet been identified (745,746). Nonetheless, a few basic facts seem to be
transported to CT, where direct coronal less than 2 mm sections are generally accepted. Direct trauma can result in skull fractures, subdural
obtained from the frontal sinuses through the temporal bones. If the hematomas, and cerebral contusion of the coup-contre-coup variety.
patient is actively leaking, contrast will be seen exiting through a defect Vigorous shaking probably causes contusions of the brain (from bang-
in the bone of the skull base. Alternative methods of detection of CSF ing of the brain against the rough edges on the inner surface of the
leakage, including noncontrast 3D-steady state MRI (CISS, FIESTA) calvarium) as well as associated subarachnoid and subdural hemor-
and intrathecal contrast-enhanced MR cisternography, are less mature rhage (from rupture of bridging veins at their weak points) (582,678–
techniques and have not been widely used in children, but have the 680,691,742,743,746–748). Damage to the lower brainstem and upper
advantage of not requiring ionizing radiation and, for the 3D-steady cervical spine can be easily overlooked and likely are underreported
state technique, being entirely noninvasive (728,729). (749) (the cervical spine is particularly at risk in infants—see sections
The role of inflammation in contributing to the neurologic seque- on “Spinal Trauma” and “Injuries in Young Children,” earlier in this
lae of traumatic brain injury is not well understood. Evidence suggests chapter—and should be carefully evaluated if neck injury is detected
that inflammation can have both detrimental and beneficial effects fol- or any question of injury to the structures of the neck is raised [750]).
lowing trauma. While the initial inflammatory cascade can result in Such injuries may result in damage to the respiratory centers and may
vascular leakage and dilation, edema, local hypoxia, and apoptotic cell be associated with subdural and epidural hematomas at the cervicome-
death, the anti-inflammatory and wound healing factors induced by dullary junction that cause further injury; apneic episodes and subse-
the initial inflammatory response may account for some of the benefits quent hypoxia may ensue (745,751). Some axonal injuries, classically
(724). In the future, targeted anti-inflammatory agents may aid in the thought to be diffuse axonal (“shearing”) injuries, have been hypoth-
prevention of secondary insults following traumatic brain injury, but esized to result from diffuse secondary hypoxia and edema that may
they are not currently in clinical practice. be triggered by an apneic episode (750,752,753); this theory remains
controversial.
Nonaccidental Trauma (Child Abuse)
Presentation
Clinical, Mechanistic, and Epidemiological Aspects The clinical presentation of the abused child is variable. The most
Nonaccidental trauma (child abuse) is an increasing problem in pedi- common presentation is that of an irritable or abnormally subdued
atric health care, with an estimated 1.5 million cases reported in the child, refusing meals, perhaps vomiting or having apneic or cyanotic
United States in 1993 (730). Three million cases of suspected abuse attacks (743). The child may present with recurrent encephalopathy,
were reported in the year 2000, and 3.6 million in 2006 (731). Of these, raising suspicion for metabolic disease or encephalitis; anemia is often
nearly half result in disfigurement, permanent neurological or psycho- present and the weight of the child is frequently below 50th percen-
logical deficit, or death (732). In the year 2002, approximately 896,000 tile (681,687,754). Another common presentation is seizures; the child
children were determined to be victims of child abuse or neglect (733). may have an isolated seizure secondary to abusive head injury or may
In 2007, an estimated 1,760 children died from abuse or neglect. Head present in status epilepticus. If the child presents with a history of head
trauma is the leading cause of morbidity and mortality in the abused trauma, the severity of the reported traumatic incident, typically a fall
child, especially in patients under the age of 2 years (734–736). One off of a table or down several stairs, may not match the severity of the
prospective study of head injuries in 100 children under the age of injury detected on the imaging study; epidemiologic studies suggest
22 years revealed that 24% were victims of abuse (737). This study and that serious injury with neurologic deficit is very unlikely from falls
others (738,739) have shown that children who have been abused have of less than 1.2 m (about 4 ft) (681,750,755,756). A prior history of
a higher risk of permanent brain damage and death than those suffer- abuse may be discovered (681,755,756). It is important to remember
ing truly accidental injuries. Radiologists have traditionally played an that children with other developmental problems are at higher risk for
important role in the diagnosis of child abuse by maintaining a high abuse (Fig. 4-91). Other risk factors include young parents, unstable
index of suspicion in children who have multiple instances of unex- family situations, low socioeconomic status, and prematurity of the
plained trauma. Although the diagnosis has classically been made by child (735,745,757).
the use of bone radiographs, which demonstrate multiple fractures Outcome of abused children is variable, but often poor, and their
of different ages (740–742), brain imaging can make an important outcomes are significantly worse than those of children injured in acci-
contribution. Brain imaging is especially important in the so-called dents (739). The estimated mortality rate ranges from 15% to 38%

Barkovich_Chap04.indd 344 5/25/2011 11:39:36 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 345

FIG. 4-91. Child abuse in a child with tuberous sclerosis. A. Axial T2-weighted image shows a left subdural hema-
toma with a focus of acute hypointense blood (solid arrows) within it. Note the right frontal tubers (open arrows).
B. Coronal GE T2* image shows more fully the extent (arrows) of the subdural hematoma.

(757), while 30% to 50% of survivors suffer cognitive or other neu- fractures more than 5 mm wide at presentation, and depressed fractures
rological deficits (758). In the first 4 months after the event, the most should raise suspicion for abusive head trauma (681). Bone scintigra-
notable sequelae are blindness, visual impairment, hemi- or tetraplegia, phy is not particularly useful in the diagnosis of linear skull fractures,
and slowing of head growth (751,754,759,760). By age 18 months, psy- as the osteoblastic response is limited. Moreover, as mentioned earlier,
chomotor retardation may be revealed by deficits in the emergence axial CT sections may miss linear skull fractures if they are parallel to
of language, constructional ability, and spatiotemporal exploration. the plane of section. Therefore, it is essential to look at the scout view of
Between ages 3 and 6 years, major behavioral disorders may become all CT scans obtained in the setting of trauma (Fig. 4-92).
apparent along with a lowering of IQ (761). Both CT and MRI can be of considerable assistance in the assess-
ment of an abused child, both by confirming the diagnosis and by
Choice of Radiological Exams allowing appraisal of the extent of brain injury (678–680,691). With
Skull radiographs are still useful in establishing the diagnosis of abusive increasing concerns regarding radiation exposure in children, the
head trauma, in that the incidence of skull fractures in affected patients American College of Radiology now recommends brain MRI over CT
is 45% (679). Multiple fractures, stellate fractures, bilateral fractures, for children aged 2 years of younger who are suspected to have suffered

FIG. 4-92. Use of scout film and identification of subtle subdural hemorrhage in nonaccidental trauma. A. Scout view
shows horizontal linear fracture (black arrows) in the frontal bone. B. Axial noncontrast CT image shows subdural blood
along the falcotentorial junction (small white arrows) and layering along the right wall of the superior sagittal sinus (white
arrowheads).

Barkovich_Chap04.indd 345 5/25/2011 11:39:36 AM


346 Pediatric Neuroimaging

abusive head trauma—so long as they have no focal signs or symptoms. If assess for evolution of parenchymal injuries. If the early MRI showed
a child up to age 5 years is suspected to have suffered physical abuse and brain parenchymal injury or if neurologic abnormality persists, then a
has neurologic signs, symptoms, or a seizure, then brain CT is recom- follow-up MRI is recommended after 2 to 3 months to look for enlarg-
mended as the most appropriate initial study (762). CT has advantages ing subdural collections, hydrocephalus, or a developing leptomenin-
in the acute setting, as it is sensitive in the detection of subdural (Fig. geal cyst (764).
4-92) and subarachnoid blood and calvarial fractures (Fig. 4-92), and
is more easily performed on an unstable, acutely injured child. Usually, Imaging Findings
CT provides most of the necessary information. MRI is most useful Subdural hemorrhage is the most common intracranial manifestation
when CT has not confirmed a strong clinical suspicion of abuse, and of abuse and its presence in any young child without an appropriate
MRI shows best the full extent of neurological injury (brain and spine), history is strongly suggestive of abusive head trauma. In one series of
which is useful for prognostication of outcome. MR exams should 93 children, aged 3 years and less, with extraparenchymal hematomas,
include diffusion images for the detection of nonhemorrhagic shear 47% with subdural hematomas were ultimately diagnosed as having
injury and hypoxic-ischemic injuries, and susceptibility-weighted been abused (682). In contrast, only 6% of patients with epidural
images, for detection of small intraparenchymal hemorrhages, in addi- hematomas were ultimately diagnosed as having been abused (682).
tion to standard T1- and T2-weighted images. Ultrasound has signifi- Subdural hematomas are particularly suggestive if seen in conjunction
cant limits in the detection of subarachnoid hemorrhage and some with acute and subacute, acute and chronic, or subacute and chronic
subdural hematomas, and is less sensitive in the detection of cerebral intraparenchymal hemorrhages (because the presence of injuries of
parenchymal injury; it has no role in the diagnosis of abusive head different ages suggests repeated trauma) or if the hematoma is mul-
trauma, but may have some use in monitoring the progression of, tiloculated or shows a fluid-fluid layer in the acute phase (suggesting
for example, subdural hematomas in infants receiving intensive care hemorrhage into a preexisting subdural hematoma, Fig. 4-90); subdural
(763). hematomas in association with severe retinal hemorrhage, particularly
Most investigators agree with the ACR position regarding CT as absent signs of impact have been reported to be highly specific, albeit
first line imaging as soon as the child with suspected abusive head not very sensitive, for abusive head trauma (767).
trauma can be stabilized (762,764,765). The utility of MRI is clear when It is sometimes difficult to differentiate bilateral chronic subdural
the CT is abnormal or when the child’s neurological status is abnormal hematomas from benign infantile enlargement of the subarachnoid
in the setting of a normal CT. When the CT is normal and the child is spaces, a condition in which the extra-axial CSF spaces are enlarged as a
without neurological signs or symptoms, the necessity of MRI is less (presumed) result of immaturity of the arachnoid villi (see Chapter 8).
well substantiated. Some favor the utilization of MRI for medical-legal When blood products are present in the subdural collections, the diag-
documentation purposes, including aiding in the timing of hemor- nosis of trauma (but not necessarily abusive head trauma) can confi-
rhages (762,764,765). Others suggest that the high sensitivity of MRI dently be made. However, when the extra-axial fluid is nearly isointense
with diffusion imaging can detect clinically significant injury when CT with CSF on multiple pulse sequences, differentiation is aided by the
findings are subtle (766). Follow up CT or MR scanning (around the presence of associated parenchymal injuries (Figs. 4-90 and 4-94).
10 day postinjury window) may be valuable to check for the devel- Subdural hematomas, subarachnoid hemorrhage and acute cerebral
opment of hydrocephalus or change in subdural hematomas, and to contusions can be visualized by CT (Figs. 4-92 and 4-93). Contusions

FIG. 4-93. Nonaccidental trauma; use of diffusion imaging and spectroscopy. MRI was performed 2 days after the
injury. A. Axial noncontrast CT shows subdural blood layering along the walls of the superior sagittal sinus (black
arrowheads) and over the cerebral convexity (white arrows). A small amount of blood lies in the left occipital horn
(black arrow). Hypodensity of the brain parenchyma is seen in the frontal lobes and left temporal lobe. B. Axial SE
3000/120 image shows the large bilateral subdural collections and the areas of parenchymal hyperintensity in the
bilateral frontal lobes and left temporal lobe.

Barkovich_Chap04.indd 346 5/25/2011 11:39:37 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 347

FIG. 4-93. (Continued) C. Axial ADC image (b = 1000 s/mm2) shows reduced dif-
fusion (low signal intensity) in the regions of parenchymal injury. D. Proton MRS
(TE = 288) shows marked reduction of NAA and elevation of lactate (Lac) in the
injured temporal lobe. E. Follow up CT scan 2 months later shows severe encepha-
lomalacia in the injured brain regions.

may be seen as ovoid areas of low attenuation or as accumulations of rate of degradation varies with size and location of the blood collection
intraparenchymal blood (high attenuation) with surrounding edema (Table 4-9) (680,767,768). It remains to be determined if this general
(Fig. 4-94A). However, small subdurals near the base of the brain and evolution scheme developed from 1.5 T imaging is equally applicable
vertex and small contusions in the anterior temporal lobes and in the for 3.0 T imaging.
orbital surface of the frontal lobes may be difficult to detect on CT. As In addition, subdural hematomas resulting from abusive head
mentioned earlier in the chapter, hemorrhage becomes more difficult trauma are often of mixed density on CT (768–770). These may result
to detect by CT after approximately 1 week, as the blood becomes pro- from single events in which early acute blood mixes with hyperacute
gressively isodense with brain (Table 4-8). hemorrhage; single events in which acute hyperdense hemorrhage
MRI is much more sensitive than CT in diagnosing subacute hema- mixes with hypodense serum and/or CSF; or two or more events with
tomas, both subdural and intraparenchymal, at the vertex (Fig. 4-94) acute and chronic hemorrhage combining to appear as a mixed den-
and in transversely oriented locations (subfrontal, along the tentorium) sity collection (768). Indeed, the only CT features that seem to support
in the middle and posterior cranial fossae (678). While some general a combination of two temporally distinct traumatic events are mem-
concepts from the known evolution of intraparenchymal hemorrhage branes within a collection, large low density collections associated with
can be extrapolated to extra-axial hemorrhage, caution should be exer- expanded underlying subarachnoid spaces (impaired absorption of
cised in trying to date hemorrhages. The MR appearance of hemato- CSF due to old blood products obstructing immature arachnoid gran-
mas varies with location, size, and stage of degradation; moreover, the ulations), and a history of slowly enlarging head circumference (746).

Barkovich_Chap04.indd 347 5/25/2011 11:39:38 AM


348 Pediatric Neuroimaging

FIG. 4-94. Nonaccidental trauma. A. Noncontrast CT shows


hypodense chronic bilateral subdural hematomas, with super-
imposed acute hemorrhage over the left temporal lobe (white
arrow) and along the left tentorial leaf (white arrowhead), with
hemorrhagic contusion (black arrow) in the right frontal lobe.
The quadrigeminal plate cistern is effaced. B. Noncontrast CT
near the vertex shows the more acute, hyperdense bilateral sub-
dural hematomas (a) and the more hypodense chronic subdural
hematomas (c) containing posteriorly layering blood products
(arrows). Soft tissue swelling overlies the left parietal bone frac-
ture and intact right parietal bone. Note the marked parenchy-
mal edema. C. Axial T2-weighted image shows the right anterior
frontal and left posterior deep white matter hemorrhagic injuries.
The chronic subdural hematomas show predominately hyperin-
tense signal, while T2 hypointensity (black arrows) indicates the
more acute hemorrhages over the medial parietal lobes.

Although abusive head trauma is, by far, the most common cause of findings. It has also reported that infants with benign enlargement
subdural hematomas in infants, it is important to keep in mind that of the subarachnoid spaces (see Chapter 8) may develop subdural
metabolic diseases can also cause bilateral subdural hematomas. In hematomas after relatively minor trauma (685). However, all infants
particular, Menkes kinky hair disease, glutaric aciduria Type 1, and and children with subdural hematomas and no satisfactory explana-
Hermansky-Pudlak syndrome can cause both subdural hematomas tion should be referred to pediatricians subspecializing in child abuse,
and retinal hemorrhages (771). These diseases, although very rare, when available, or child protective services, for thorough evaluation, as
should be kept in mind when evaluating infants with such imaging a traumatic cause will very commonly be found (772,773). Radiologists
should be aware that all 50 United States have mandatory reporting
statutes for suspected child abuse and neglect.
Edema and parenchymal injury of the cortex and white matter is
TABLE 4-8 General Evolution of commonly present and is more sensitively detected by MRI than by
Subdural Hematomas on CT CT; in the early phase of injury, diffusion images make the lesions
more conspicuous (766). Common locations for the contusions
include the anterior temporal lobes and anterior frontal lobes (Figs.
Stage Appearance Estimated Age Range
4-93 and 4-94). Noncontusional cerebral cortical/subcortical injury
Hyperacute Isodense <3 h can also occur in child abuse, although the precise mechanism is not
Acute Hyperdense Few Hours to 11 d always elucidated (735,774). These lesions are typically not in classic
vascular territories and they often cross vascular territories; therefore,
Subacute Isodense 1 ½–3 wk it is not certain that they represent infarcts. Single large areas or mul-
Chronic Hypodense More than 3 wk tiple noncontiguous areas may be involved (Figs. 4-93, 4-95, and 4-96).
When large areas of cortical injury are seen in conjunction with sub-
(Adapted from References 767 and 768)
dural or subarachnoid hemorrhage in an infant, the possibility of abu-
sive head trauma should be raised (774). If the MR study is performed

Barkovich_Chap04.indd 348 5/25/2011 11:39:38 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 349

TABLE 4-9 General Evolution of Subdural Hematomas on MRI


Stage T1 Intensity T2 Intensity FLAIR Intensity Estimated Age Range
Hyperacute Iso or hypo Hyper Hypo <24 h
Acute Iso or hypo Hypo Hypo 1–4 d
Early subacute Hyper Iso or hypo Any 2–3 d to 1–2 wk
Late subacute Hyper Hyper Iso to hyper 1–2 wk to 1–2 mo
Chronic (subdural membrane) Iso Hypo Unproven, likely iso Few weeks to years
Chronic (subdural contents) Hypo but brighter Hyper Unproven, likely hypo Few weeks to years
than CSF but brighter than CSF
Note: When sedimentation of blood products occurs, the signal from the sediment is felt to be more accurate for timing of hemorrhage, particularly for T1 and
FLAIR sequences.
(Adapted from References 767 and 768)

soon after the event (during the first day), the injured tissue may be of different magnetic susceptibilities, especially when long (>30 mil-
difficult to detect by routine sequences. Diffusion imaging is invalu- liseconds) echo times are used for GE (Fig. 4-90 and 4-96C). Areas of
able in this setting, as the injured areas will show reduced diffusivity old hemorrhage can be seen on these images as areas of hypointensity
(Figs. 4-93, and 4-96), often in areas that appear normal on standard that are larger and much more apparent than on the T2-weighted spin-
sequences (775,776). On routine sequences, the signal intensity of the echo images. These images are obtained in addition to T2-weighted
injured tissue varies, depending upon the age of the injury and the spin-echo images, not substituted for them. The information they
presence or lack of associated blood. As described in prior sections, provide is complementary to that from spin-echo, RARE, or FLAIR
acute hemorrhage is isointense with brain on T1-weighted spin-echo images; significant parenchymal injury may be missed when GE or
sequences and hypointense on T2-weighted spin-echo and gradient- SWI sequences are employed exclusively. As stated earlier, RARE (fast/
echo sequences with 1.5 T imaging. Subacute blood develops high sig- turbo spin echo) and FLAIR sequences, which are relatively insensitive
nal intensity, first on T1-weighted and then on T2-weighted sequences. to paramagnetic substances (Figs. 4-89, 4-90, and 4-96), should not be
Old hemorrhage within the brain can be identified by the presence of used as primary sequences at 1.5 T static field strength in trauma cases, as
old blood products, predominantly hemosiderin and ferritin, which the presence of old and new hemorrhage is of critical importance. If 3 T
appear as ill-defined areas of very low signal intensity on T2-weighted scanners become common enough that they are used for acute trauma,
spin-echo or gradient-echo sequences (see Fig. 4-90). Gradient-echo it may be necessary to rethink this algorithm, as acute and early sub-
or susceptibility-weighted images (SWI) are especially useful in sus- acute blood seems to be more easily detected as low signal intensity on
pected abusive head trauma cases because of their sensitivity to regions FLAIR and RARE sequences at 3 T (707).

FIG. 4-95. Nonaccidental trauma. A. Axial noncontrast CT image shows right-to-left subfalcine shift, right subdural hematoma (open arrows) and
hypodensity of the posterior cerebral hemispheres bilaterally (solid arrows). B. CT image at higher level shows hypodensity of the entire right cerebral
hemisphere and the anterior and posterior portions of the left hemisphere. C. Bone windows of a CT image superior to (B) shows a depressed skull fracture
(open arrow) and splitting of the coronal sutures (solid arrows).

Barkovich_Chap04.indd 349 5/25/2011 11:39:39 AM


350 Pediatric Neuroimaging

FIG. 4-96. Nonaccidental trauma. Hemorrhage and hypoperfusion


injury in a 3-month old brought to the emergency department because of
seizure. A. Axial fat-suppressed FLAIR shows subdural hematomas over
both cerebral hemispheres (white arrows) that are largely lower in signal
intensity than parenchyma and have a gradient (with posterior more
hyperintense), while more acute subdural blood along the tentorium
(white arrowheads) is significantly more hyperintense than parenchyma.
B. Coronal T2-weighted image shows the heterogeneity of the various
ages of subdural hemorrhage, with hypointense more acute blood over
the medial right parietal cortex (black arrowheads) and hyperintense more
chronic blood over both convexities. Note the membrane in the left sided
chronic subdural hematoma (black arrow). C. Susceptibility-weighted
image demonstrates the intraventricular hemorrhage that layers within
the occipital horns of the lateral ventricles (black arrowheads). This was
not seen on other sequences. D. Axial T1-weighted image near the vertex
shows the subdural hematomas of varying ages, including a very hyper-
intense focus within the posterior interhemispheric fissure, and the loss
of gray-white differentiation in a watershed distribution (white arrows).
E. Axial Dav map shows markedly reduced diffusivity (low signal intensity)
in a watershed distribution, confirming hypoperfusion injury.

Barkovich_Chap04.indd 350 5/25/2011 11:39:39 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 351

Injury to the spine and spinal cord is commonly seen in pathologic 19. Barkovich AJ, Al Ali F, Rowley HA, Bass N. Imaging patterns of neonatal
examinations of abused children. The cervical spine is particularly hypoglycemia. Am J Neuroradiol 1998;19:523–528.
affected by shaking injury and should always be examined if other evi- 20. Filan PM, Inder TE, Cameron FJ, Kean MJ, Hunt RW. Neonatal hypoglyce-
dence of shaking injury is identified. As discussed earlier, infants may mia and occipital cerebral injury. J Pediatr 2006;148:552–555.
21. Frigieri G, Guidi B, Costa Zaccarelli S, et al. Multicystic encephalomalacia
suffer significant spinal cord injury in the absence of radiologically
in term infants. Childs Nerv Syst 1996;12:759–764.
apparent injury to the spinal column. Therefore, MRI is the examina- 22. Fullerton HJ, Chetkovich DM, Wu YW, Smith WS, Johnston SC. Deaths
tion of choice for imaging of the spine. Both sagittal and axial T1- and from stroke in US children, 1979 to 1998. Neurology 2002;59(1):34–39.
T2-weighted images should be obtained to look for spinal cord injury 23. Fullerton HJ, Wu YW, Zhao S, Johnston SC. Risk of stroke in children:
(even cord transection), subdural and epidural hematomas, injuries to ethnic and gender disparities. Neurology 2003;61(2):189–194.
the vertebral bodies, and paraspinous hemorrhage due to the vertebral 24. Green SH. The paediatric overview. Pediatr Radiol 2004;34:5–9.
body injuries. 25. Fullerton HJ, Johnston SC, Smith WS. Arterial dissection and stroke in chil-
Proton MRS obtained in the subacute phase may be useful to assess dren. Neurology 2001;57(7):1155–1160.
prognosis in shaken babies (Fig. 4-93). In particular, the presence of 26. Husson B, Lasjaunias P. Radiological approach to disorders of arterial brain
elevated lactate or the reduction of NAA on MRS studies obtained 5 to vessels associated with childhood arterial stroke-a comparison between
MRA and contrast angiography. Pediatr Radiol 2004;34(1):10–15.
7 days after the injury indicate significant damage; preliminary stud-
27. Ganesan V, Chong WK, Cox TC, Chawda SJ, Prengler M, Kirkham FJ. Poste-
ies suggest that these patients will have a poor prognosis (777). This rior circulation stroke in childhood: risk factors and recurrence. Neurology
observation has been confirmed on a larger patient cohort, and made 2002;59(10):1552–1556.
even more sensitive and specific when correlated with age, initial GCS 28. Ganesan V, Hogan A, Shack N, Gorton A, Isaacs E, Kirkham FJ. Outcome after
score, and presence of retinal hemorrhage (778). ischaemic stroke in childhood. Dev Med Child Neurol 2000;42:455–461.
29. Wu Y, Miller S, Chin K, et al. Multiple risk factors in neonatal sinovenous
thrombosis. Neurology 2002;59:438–440.
REFERENCES 30. Wu Y, Hamrick S, Miller S, et al. Intraventricular hemorrhage in term neo-
nates caused by sinovenous thrombosis. Ann Neurol 2003;54:123–126.
1. Bax M, Tydeman C, Flodmark O. Clinical and MRI correlates of cere- 31. Lee Y-Y, Lin K-L, Wang H-S, et al. Risk factors and outcomes of childhood
bral palsy. The European Cerebral Palsy Study. J Am Med Assn 2006;296: ischemic stroke in Taiwan. Brain Develop 2008;30(1):14–19.
1602–1608. 32. Kirton A, DeVeber G, Pontigon A-M, Macgregor D, Shroff M. Presumed
2. Raybaud C. Destructive lesions of the brain. Neuroradiology 1983;25: perinatal ischemic stroke: Vascular classification predicts outcomes. Ann
265–291. Neurol 2008;63:436–443.
3. Gilles FH. Neuropathologic indicators of abnormal development. In: Free- 33. McQuillen PS, Hamrick SEG, Perez MJ, et al. Balloon Atrial Septostomy Is
man JM, ed. Prenatal and Perinatal Factors Associated with Brain Disorders. Associated With Preoperative Stroke in Neonates With Transposition of the
Bethesda: NIH, 1985:53–107. Great Arteries. Circulation 2006;113(2):280–285.
4. Faden AI, Simon RP. A potential role for excitotoxins in the pathophysiol- 34. Roach ES. Cerebrovascular disease in infants and children. In: Berg BO, ed.
ogy of spinal cord injury. Ann Neurol 1988;23:623–626. Principles of Child Neurology. New York: McGraw-Hill, 1996:901–936.
5. Friede RL. Developmental Neuropathology, 2nd ed. Berlin: Springer-Verlag, 35. Young RSK. Neurologic aspects of cardiovascular disease. In: Berg BO, ed.
1989. Neurologic Aspects of Pediatrics. Boston: Butterworth-Heinemann, 1992:
6. Evrard P, de Saint-Georges P, Kadhim HJ, Gadisseux J-F. Pathology of 320–338.
prenatal encephalopathies. In: French J, ed. Child Neurology and Develop- 36. Lanska MJ, Lanska DJ, Horwitz SJ, Aram DM. Presentation, clinical course,
mental Disabilities. Baltimore: Paul H. Brookes, 1989:153–176. and outcome of childhood stroke. Pediatr Neurol 1991;7:333–341.
7. Yakovlev PI, Wadsworth RC. Schizencephalies. A study of the congenital 37. Mercuri E, Barnett A, Rutherford M, et al. Neonatal cerebral infarction and
clefts in the cerebral mantle. 1. Clefts with fused lips. J Neuropathol Exp neuromotor outcome at school age. Pediatrics 2004;113(1 Pt 1):95–100.
Neurol 1946;5:116–130. 38. Raju TNK, Nelson KB, Ferriero D, Lynch JK, and the N-NPSWP. Ischemic
8. Yakovlev PI, Wadsworth RC. Schizencephalies. A study of the congenital Perinatal Stroke: Summary of a Workshop Sponsored by the National Insti-
clefts in the cerebral mantle. 2. Clefts with hydrocephalus and lips sepa- tute of Child Health and Human Development and the National Institute
rated. J Neuropathol Exp Neurol 1946;5:169–206. of Neurological Disorders and Stroke. Pediatrics 2007;120(3):609–616.
9. Probst FP. The Prosencephalies: Morphology, Neuroradiological Appearances 39. Ramaswamy V, Miller SP, Barkovich AJ, Partridge JC, Ferriero DM. Peri-
and Differential Diagnosis. Berlin: Springer-Verlag, 1979. natal stroke in term infants with neonatal encephalopathy. Neurology
10. Hahn J, Lewis AJ, Barnes PD. Hydranencephaly owing to twin-twin 2004;62:2088–2091.
transfusion: serial fetal ultrasonography and magnetic resonance imaging 40. Abels L, Lequin M, Govaert P. Sonographic templates of newborn perfora-
findings. J Child Neurol 2003;18:367–370. tor strokes. Pediatr Radiol 2006;36:663–669.
11. Basu S, Kumar A, Gupta S, Bhatia BD. A rare association of hydranenceph- 41. Takanashi J, Barkovich A, Ferriero D, Suzuki H, Kohno Y. Widening spec-
aly with congenital rubella. Indian J Pediatr 2007;74:793–794. trum of congenital hemiplegia: Periventricular venous infarction in term
12. Friede RL, Mikolasek J. Postencephalitic porencephaly, hydranencephaly or neonates. Neurology 2003;61:531–533.
polymicrogyria. A review. Acta Neuropathol 1978;43:161–168. 42. Takanashi J, Tada H, Barkovich AJ, Kohno Y. Magnetic resonance imaging
13. Warkany J, Lemire RJ, Cohen MM. Mental Retardation and Congenital Mal- confirms periventricular venous infarction in a term-born child with con-
formations of the Central Nervous System. Chicago: Yearbook, 1981. genital hemiplegia. Dev Med Child Neurol 2005;47:706–708.
14. Hanigan WC, Aldrich WM. MRI and evoked potentials in a child with 43. Sibony O, Stempfle N, Luton D, Oury JF, Blot PH. In utero fetal cerebral
hydranencephaly. Pediatr Neurol 1988;4:185–187. intraparenchymal ischemia diagnosed by nuclear magnetic resonance. Dev
15. Barkovich AJ. Congenital malformations of the brain and skull. In: Med Child Neurol 1998;40:122–123.
Barkovich AJ, ed. Pediatric Neuroimaging, 3rd ed. Philadelphia: Lippincott 44. Canapicchi R, Cioni G, Strigini FA, Abbruzzese A, Bartalena L, Lencioni G.
Williams & Wilkins, 2000:251–381. Prenatal diagnosis of periventricular hemorrhage by fetal brain magnetic
16. Barkovich AJ, Sargent SK. Profound asphyxia in the preterm infant: imag- resonance imaging. Childs Nerv Syst 1998;14:689–692.
ing findings. Am J Neuroradiol 1995;16:1837–1846. 45. de Vries LS, Koopman C, Groenendaal F, et al. COL4A1 mutation in two
17. Barkovich AJ, Hallam D. Neuroimaging in perinatal hypoxic-ischemic preterm siblings with antenatal onset of parenchymal hemorrhage. Ann
injury. Ment Retard Dev Disabil Res Rev 1997;3:28–41. Neurol 2009;65:12–18.
18. Barkovich AJ, Truwit CL. MR of perinatal asphyxia: Correlation of gesta- 46. Williams LS, Garg BP, Cohen M, Fleck JD, Biller J. Subtypes of ischemic
tional age with pattern of damage. Am J Neuroradiol 1990;11:1087–1096. stroke in children and young adults. Neurology 1997;49:1541–1545.

Barkovich_Chap04.indd 351 5/25/2011 11:39:40 AM


352 Pediatric Neuroimaging

47. Riikonen R, Santavuori P. Hereditary and acquired risk factors for childhood 72. Roach ES, Riela AR. Pediatric Cerebrovascular Disorders, 2nd ed. New York:
stroke. Neuropediatrics 1994;25:227–233. Futura, 1995.
48. Ganesan V, Prengler M, McShane MA, Wade AM, Kirkham FJ. Investiga- 73. Moser FG, Miller ST, Bello JA, et al. The spectrum of brain MR abnormali-
tion of risk factors in children with arterial ischemic stroke. Ann Neurol ties in sickle cell disease: a report from the cooperative study of sickle cell
2003;53(2):167–173. disease. Am J Neuroradiol 1996;17:965–972.
49. Lynch JK, Han CJ, Nee LE, Nelson KB. Prothrombotic factors in children 74. Yamada I, Yoshiharu M, Suzuki S. Moyamoya disease: diagnosis with three-
with stroke or porencephaly. Pediatrics 2005;116:447–453. dimensional time-of-flight MR angiography. Radiology 1992;184:773–778.
50. Gunther G, Junker R, Strater R, et al. Symptomatic ischemic stroke in full- 75. Yamada I, Himeno Y, Suzuki S, Matsushima Y. Posterior circulation in moy-
term neonates: role of acquired and genetic prothrombotic risk factors. amoya disease: angiographic study. Radiology 1995;197:239–246.
Stroke 2000;31:2437–2441. 76. Frieden IJ, Reese V, Cohen D. PHACE syndrome. The association of poste-
51. Mercuri E, Cowan F, Gupte G, et al. Prothrombotic Disorders and Abnor- rior fossa brain malformations, hemangiomas, arterial anomalies, coarcta-
mal Neurodevelopmental Outcome in Infants With Neonatal Cerebral tion of the aorta and cardiac defects, and eye anomalies. Arch Dermatol
Infarction. Pediatrics 2001;107(6):1400–1404. 1996;132:307–311.
52. Braun KPJ, Bulder MMM, Chabrier S, et al. The course and outcome of 77. Gould DB, Phalan FC, Breedveld GJ, et al. Mutations in Col4a1 cause peri-
unilateral intracranial arteriopathy in 79 children with ischaemic stroke. natal cerebral hemorrhage and porencephaly. Science 2005;308:1167–1171.
Brain 2009;132:544–557. 78. van der Knaap MS, Smit L, Barkhof F, et al. Neonatal porencephaly and adult
53. Chung B, Wong V. Pediatric stroke among Hong Kong Chinese subjects. stroke related to mutations in collagen IV A1. Ann Neurol 2006;59:504–511.
Pediatrics 2004;114(2):e206–212. 79. Amlie-Lefond C, Kleinschmidt-DeMasters B, Mahalingam R, Davis L,
54. Boardman JP, Ganesan V, Rutherford MA, Saunders DE, Mercuri E, Cowan Gilden D. The vasculopathy of varicella-zoster virus encephalitis. Ann
F. Magnetic resonance imag correlates of hemiparesis after neonatal and Neurol 1995;37:784–790.
childhood middle cerebral artery stroke. Pediatrics 2005;115:321–326. 80. Silverstein F, Brunberg J. Postvaricella basal ganglia infarction in children.
55. Ganesan V, Ng V, Chong WK, Kirkham FJ, Connelly A. Lesion volume, Am J Neuroradiol 1995;16:449–452.
lesion location, and outcome after middle cerebral artery territory stroke. 81. Schievink WI, Mokri B, Piepgras DG. Spontaneous dissection of cervicocepha-
Arch Dis Child 1999;81:295–300. lic arteries in childhood and adolescence. Neurology 1994;44:1607–1612.
56. Ballantyne AO, Spilkin AM, Hesselink J, Trauner DA. Plasticity in the devel- 82. Ogura K, Maegaki Y, Morino S, Ogawa T, Ohno K, Oka A. Vertebral artery
oping brain: intellectual, language and academic functions in children with dissection in an infant: ultrastructural aberrations in connective tissue
ischaemic perinatal stroke. Brain 2008;131:2975–2985. components. Neuropediatrics 2003;34:307–310.
57. Raybaud C, Girard N, Sévely A, Leboucq N. Neuroradiologie pédia- 83. Liu GT, Holmes GL. Varicella with delayed contralateral hemiparesis
trique (I). In: Raybaud C, Girard N, Sévely A, Leboucq N, eds. Radiodi- detected by MRI. Pediatr Neurol 1990;6:131–134.
agnostic—Neuroradiologie-Appariel Locomoteur, vol 31–621-A-05. Paris: 84. Shuper Z, Vining EPG, Freeman JM. CNS vasculitis after chickenpox–cause
Elsevier; 1996:26. or coincidence? Arch Dis Child 1990;65:1245–1248.
58. Giroud M, Lemesle M, Gouyon J, Nivelon J, Milan C, Dumas R. Cerebro- 85. Bodensteiner J, Hille M, Riggs J. Clinical features of vascular thrombosis
vascular disease in children under 16 years in the city of Dijon, France: a following varicella. Am J Dis Child 1992;146:100–102.
study of incidence and clinical features from 1985–1993. J Clin Epidemiol 86. Ganesan V, Kirkham FJ. Mechanisms of ischaemic stroke after chickenpox.
1995;48:1343–1348. Arch Dis Child 1997;76:522–525.
59. Dusser A, Goutières F, Aicardi J. Ischaemic strokes in children. J Child 87. Miaux Y, Ribaud P, Williams M, et al. MR of cerebral aspergillosis in
Neurol 1986;1:131–136. patients who have had bone marrow transplantation. Am J Neuroradiol
60. Israels SJ, Seshia SS. Childhood stroke associated with protein C or S defi- 1995;16:555–562.
ciency. J Pediatr 1987;111:562–564. 88. Nowak-Gottl U, Strater R, Heinecke A, et al. Lipoprotein (a) and genetic
61. Zoller B, Dahlbäck B. Linkage between inherited resistance to activated polymorphisms of clotting factor V, prothrombin, and methylenetetrahy-
protein C and factor V gene mutation in venous thrombosis. Lancet drofolate reductase are risk factors of spontaneous ischemic stroke in child-
1994;343:1536–1538. hood. Blood 1999;94:3678–3682.
62. Brey RL, Coull BM. Cerebral venous thrombosis: role of activated protein C 89. Grow J, Fliman P, Pipe S. Neonatal sinovenous thrombosis associated with
resistance and factor V gene mutation. Stroke 1996;27:1719–1720. homozygous thermolabile methylenetetrahydrofolate reductase in both
63. Göbel U. Inherited or acquired disorders of blood coagulation in children mother and father. J Perinatol 2002;22:175–178.
with neurovascular complications. Neuropediatrics 1994;25:4–7. 90. Heier LA, Carpanzo CR, Mast J, Brill PW, Winchester P, Deck MDF. Mater-
64. van Kuijck MAP, Rotteveel JJ, van Oostrom CG, Novakova I. Neurologi- nal cocaine abuse: the spectrum of radiologic abnormalities in the neonatal
cal complications in children with protein c deficiency. Neuropediatrics CNS. Am J Neuroradiol 1991;12:951–956.
1994;25:16–19. 91. Hoyme HE, Jones KL, Dixon SD, et al. Prenatal cocaine syndrome and fetal
65. de Veber G, Monagle P, Chan A. Prothrombic disorders in infants and chil- vascular disruption. Pediatrics 1990;85:743–747.
dren with cerebral thromboembolism. Arch Neurol 1998;55:1539–1543. 92. Rossi LN, Penzien JM, Deonna T, et al. Does migraine-related stroke occur
66. Steinlin M, Pfister I, Pavlovic J, et al. The first three years of the Swiss neu- in childhood? Dev Med Child Neurol 1990;32:1005–1009.
ropaediatric stroke registry: a population-based study of incidence, symp- 93. Edwards MSB, Hoffman HJ. Cerebral vascular disease in children and ado-
toms and risk factors. Neuropediatrics 2005;36:90–97. lescents Baltimore: Williams and Wilkins, 1989.
67. Schöning M, Klein R, Krägeloh-Mann I, et al. Antiphospholipid antibod- 94. Aviv rI, Benseler SM, Silverman ED, et al. MR imaging and angiogrpahly of
ies in cerebrovascular ischemia and stroke in childhood. Neuropediatrics primary CNS vasculitis of childhood. Am J Neuroradiol 2006;27:192–199.
1994;25:8–14. 95. Benseler S, Schneider R. Central nervous system vasculitis in children. Curr
68. Angelini L, Zibordi F, Nardocci N, Caporali R, Ravelli A, Martini A. Neuro- Opin Rheumatol 2004;16:43–50.
logical disorders, other than stroke, associated with antiphospholipid anti- 96. Sebire G, Meyer L, Chabrier S. Varicella as a risk factor for cerebral infarc-
bodies in childhood. Neuropediatrics 1996;27:149–153. tion in childhood: a case-control study. Ann Neurol 1999;45:679–680.
69. Golomb MR, MacGregor DL, Domi T, et al. Presumed pre-or pernatal arterial 97. Sperl W, Felber S, Skladal D, Wermuth B. Metabolic stroke in carbamyl
ischemic stroke: risk factors and outcomes. Ann Neurol 2001;50:163–168. phosphate synthetase deficiency. Neuropediatrics 1997;28:229–234.
70. Bisset GS, III, Scjwartz DC, Meyer RA, et al. Clinical spectrum and long- 98. Vallée L, Fontaine M, Nuyts J, Ricart G, Krisovic I, Divry P. Stroke,
term follow up of isolated mitral valve prolapse in 119 children. Circulation hemiparesis and deficient mitochondrial beta oxidation. Eur J Pediatr
1980;62:423–427. 1994;153:598–603.
71. Thijs RD, Hazekamp MG, Rijlaarsdam ME, Willems SM, Schutte PJ, Laan 99. van den Berg M, van der Knaap MS, Boers GHJ, Stehouwer CDA, Rauwerda
LA. Unexpected cause of a recurrent cerebral hemorrhage. Neuropediatrics JA, Valk J. Hyperhomocysteinaemia; with reference to its neuroradiological
2005;36:324–327. aspects. Neuroradiology 1995;37:403–411.

Barkovich_Chap04.indd 352 5/25/2011 11:39:40 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 353

100. Wagle WA, Haller JS, Cousins JP. Cerebral infarction in progeria. Pediatr 125. Shevell MI, Silver K, O’Gorman AM, Watter GV, Montes JL. Neonatal
Neurol 1992;8:476–477. dural sinus thrombosis. Pediatr Neurol 1989;5:161–165.
101. Shih VE, Abroms IF, Johnson JL, et al. Sulfite oxidase deficiency. N Engl J 126. Rivkin MJ, Anderson ML, Kaye EM. Neonatal idiopathic cerebral venous
Med 1977;297:1022–1028. thrombosis: an unrecognized cause of transient seizures or lethargy. Ann
102. Gleuck CJ, Daniels SR, Bates S, et al. Pediatric victims of unexplained Neurol 1992;32:51–56.
stroke and their families: familial lipid and lipoprotein abnormalities. 127. Medlock MD, Olivero WC, Hanigan WC, et al. Children with cerebral
Pediatrics 1982;69:308–316. venous thrombosis diagnosed with magnetic resonance imaging and
103. Thompson JE, Smith M, Castillo M, Barrow M, Mukherji SK. MR in chil- magnetic resonance angiography. Neurosurgery 1992;31:870–876.
dren with L-carnitine deficiency. Am J Neuroradiol 1996;17:1585–1588. 128. Leach JL, Strub WM, Gaskill-Shipley MF. Cerebral venous thrombus
104. Küker W, Gaertner S, Nägele T, et al. Vessel wall contrast enhancement: signal intensity and susceptibility effects on gradient recalled echo MR
a diagnostic sign of cerebral vasculitis. Cerebrovasc Dis 2008;26:23–29. imaging. Am J Neuroradiol 2007;28:940–945.
105. de Vries LS, van der Grond J, Van Haastert IC, Groenendaal F. Prediction of 129. Linn J, Ertl-Wagner B, Seelos KC, et al. Diagnostic Value of Multidetector-
outcome in newborn infants with arterial ischemic stroke using diffusion- Row CT Angiography in the Evaluation of Thrombosis of the Cerebral
weighted magnetic resonance imaging. Neuropediatrics 2005;36:12–20. Venous Sinuses. Am J Neuroradiol 2007;28(5):946–952.
106. de Veber G, Andrew M, Adams C, et al. Cerebral sinovenous thrombosis in 130. Ducreux D, Oppenheim C, Vandamme X, et al. Diffusion-weighted imag-
children. N Engl J Med 2001;345:417–423. ing patterns of brain damage associated with cerebral venous thrombosis.
107. James CA, Glasier CM, Angtuaco EE. Altered vertebrobasilar flow in chil- Am J Neuroradiol 2001;22:261–268.
dren: angiographic, MR, and MR angiographic findings. Am J Neuroradiol 131. Yoshikawa T, Abe O, Tsuchiya K, et al. Diffusion-weighted magnetic
1995;16:1689–1695. resonance imaging of dural sinus thrombosis. Neuroradiology 2002;44:
108. Garg BP, Ottinger CJ, Smith RR, Fishman MA. Strokes in children due to 481–488.
vertebral artery trauma. Neurology 1993;43:2555–2558. 132. Forbes K, Pipe J, Heiserman J. Evidence for cytotoxic edema in the patho-
109. Caplan LR. Migraine and vertebrobasilar ischemia. Neurology genesis of central venous infarction. Am J Neuroradiol 2001;22:450–455.
1991;41:55–61. 133. Chiras J, Dubs M, Bories J. Venous infarctions. Neuroradiology 1985;27:
110. Koo B, Becker LE, Chuang SH, et al. Mitochondrial encephalopathy, lactic 593–600.
acidosis, stroke-like episodes (MELAS): clinical radiologic, pathological, 134. Hall DMB. Birth asphyxia and cerebral palsy. Br Med J 1989;299:279–282.
and genetic observations. Ann Neurol 1993;34:25–32. 135. Little WJ. On the influence of abnormal partuition, difficult labour, pre-
111. Rosen L, Phillips S, Enzmann D. Magnetic resonance imaging in MELAS mature birth, and asphyxia neonatorum on mental and physical condi-
syndrome. Neuroradiol 1990;32:168–171. tions of the child, especially in relation to deformities. Trans Obstet Soc
112. Garg BP, DeMyer WE. Ischemic thalamic infarction in children: Clinical London 1862;3:293–344.
presentation, etiology, and outcome. Pediatr Neurol 1995;13:46–49. 136. Nelson KB. What proportion of cerebral palsy is related to birth asphyxia?
113. Blankenberg FG, Norbash AM, Lane B, Stevenson DK, Bracci PM, J Pediatr 1988;112:572–573.
Enzmann DR. Neonatal intracranial ischemia and hemorrhage: diagnosis 137. Wheater M, Rennie JM. Perinatal infection is an important risk factor
with US, CT, and MR imaging. Radiology 1996;199:253–259. for cerebral palsy in very low birthweight infants. Dev Med Child Neurol
114. de Vries LS, Groenendaal F, Eken P, van Haastert IC, Rademaker KJ, Mein- 2000;42:364–367.
ers LC. Infarcts in the vascular distribution of the middle cerebral artery 138. Nelson KB, Dambrosie JM, Grether JK, Phillips TM. Neonatal cytok-
in preterm and full term infants. Neuropediatrics 1997;28:88–96. ines and coagulation factors in children with cerebral palsy. Ann Neurol
115. Taylor GA. Alterations in regional cerebral blood flow in neonatal stroke: 1998;44:665–675.
preliminary findings with color Doppler sonography. Pediar Radiol 139. Grether JK, Nelson KB. Maternal infection and cerebral palsy in infants of
1994;24:111–115. normal birth weight. J Am Med Assn 1998;279:207–211.
116. Cowan FM, Pennock JM, Hanrahan JD, Manji KP, Edwards AD. Early 140. Grether JK, Nelson KB, Emery ES, Cummings SK. Prenatal and peri-
detection of cerebral infarction and hypoxic ischemic encephalopathy in natal factors and cerebral palsy in very low birth weight infants. J Pediatr
neonates using diffusion weighted magnetic resonance imaging. Neurope- 1996;128:407–414.
diatrics 1994;25:172–175. 141. Dammann O, Leviton A. Role of the fetus in perinatal infection and neo-
117. McKinstry R, Miller J, Snyder A, et al. A prospective, longitudinal diffu- natal brain damage. Curr Opin Pediatr 2000;12:99–104.
sion tensor imaging study of brain injury in newborns. Neurology 2002;59: 142. Kraus F, Acheen V. Fetal thrombotic vasculopathy in the placenta: cere-
824–833. bral thrombi and infarcts, coagulopathies, and cerebral palsy. Hum Pathol
118. Dudink J, E. Mercuri E, L. Al-Nakib L, et al. Evolution of unilateral peri- 1999;30:759–769.
natal arterial ischemic stroke on conventional and diffusion-weighted MR 143. Willis TA, Davidson J, Gray RGF, Poulton K, Ramani P, Whitehouse W.
imaging. Am J Neuroradiol 2009;30:998–1004. Cytochrome oxidase deficiency presenting as birth asphyxia. Dev Med
119. Barkovich AJ, Miller SP, Bartha A, et al. MRI, MRS, and DTI of sequen- Child Neurol 2000;42:414–417.
tial studies in neonates with encephalopathy. Am J Neuroradiol 2006;27: 144. Del Toro J, Louis PT, Goddard-Finegold J. Cerebrovascular regulation and
533–547. neonatal brain injury. Pediatr Neurol 1991;7:3–12.
120. Mazumdar A, Mukherjee P, Miller J, Malde H, McKinstry R. Diffusion- 145. Boylan G, Young K, Panerai R, Rennie J, Evans D. Dynamic cerebral auto-
weighted imaging of acute corticospinal tract injury preceding Wal- regulation in sick newborn infants. Pediatr Res 2000;48:12–17.
lerian degeneration in the maturing human brain. Am J Neuroradiol 146. Hill A. Current concepts of hypoxic-ischemic cerebral injury in the term
2003;24:1057–1066. newborn. Pediatr Neurol 1991;7:317–325.
121. Sebire G, Tabarki B, Saunders DE, et al. Cerebral venous sinus thrombosis 147. Volpe JJ. Hypoxic-ischemic encephalopathy: neuropathology and patho-
in children: risk factors, presentation, diagnosis and outcome. Brain genesis. In: Volpe JJ, ed. Neurology of the Newborn. Philadelphia, PA:
2005;128(Pt 3):477–489. Saunders-Elsevier, 2008:347–399.
122. Teksam M, Moharir M, deVeber G, Shroff M. Frequency and topographic 148. Johnston MV, Ishida A, Ishida WN, Matsushita HB, Nishimura A, Tsuji M.
distribution of brain lesions in pediatric cerebral venous thrombosis. Am Plasticity and injury in the developing brain. Brain Dev 2009;31:1–10.
J Neuroradiol 2008;29:1861–1965. 149. Tsuji M, Saul J, du Plessis A, et al. Cerebral intravascular oxygenation cor-
123. Huisman TAGM, Holzmann D, Martin E, Willi UV. Cerebral venous relates with mean arterial pressure in critically ill premature infants. Pedi-
thrombosis in childhood. Eur Radiol 2001;11:1760–1765. atrics 2000;106:625–632.
124. Knopman J, Tsiouris AJ, Souweidane MM. Atraumatic epidural hema- 150. Okumura A, Toyota N, Hayakawa F, et al. Cerebral hemodynamics during
toma secondary to a venous sinus thrombosis: a novel finding. J Neurosurg early neonatal period in preterm infants with periventricular leukomala-
Pediatrics 2008;2:416–419. cia. Brain Dev 2002;24:693–697.

Barkovich_Chap04.indd 353 5/25/2011 11:39:41 AM


354 Pediatric Neuroimaging

151. Volpe JJ. Intracranial hemorrhage: germinal matrix-intraventricular 175. Ashwal S, Majcher JS, Longo L. Patterns of fetal lamb regional cerebral
hemorrhage of the premature infant. In: Volpe JJ, ed. Neurology of the blood flow during and after prologned hypoxia: studies during the post-
Newborn, 5th ed. Philadelphia, PA: Saunders-Elsevier; 2008:517–588. hypoxic recovery period. Am J Obstet Gynecol 1981;139:365–372.
152. Vannucci RC, Yager JY. Glucose, lactic acid, and perinatal hypoxic-ischemic 176. Li AM, Chau V, Poskitt KJ, et al. White matter injury in term newborns
brain damage. Pediatr Neurol 1992;8:3–12. with neonatal encephalopathy. Pediatr Res 2009;65:85–89.
153. Mallard EC, Gunn AJ, Williams CE, Johnston BM, Gluckman PD. Tran- 177. Roland EH, Hill A, Norman MG, Flodmark O, MacNab AJ. Selective
sient umbilical cord occlusion causes hippocampal damage in fetal sheep. brainstem injury in an asphyxiated newborn. Ann Neurol 1988;23:89–92.
Am J Obstet Gynecol 1992;167:1423–1430. 178. Pasternak JF, Predley TA, Mikhael MA. Neonatal asphyxia: vulnerability of
154. Williams CE, Gunn AJ, Mallard EC, Gluckman PD. Outcome after isch- basal ganglia, thalamus, and brainstem. Pediatr Neurol 1991;7:147–149.
emia in the developing sheep brain: an electroencephalographic and his- 179. Rosenbloom L. Dyskinetic cerebral palsy and birth asphyxia. Dev Med
tological study. Ann Neurol 1992;31:14–21. Child Neurol 1994;36:285–289.
155. Ranck JB, Windle WF. Brain damage in the monkey, Macaca mulatta, by 180. Azzarelli B, Meade P, Muller J. Hypoxic lesions in areas of primary myeli-
asphyxia neonatorum. Exp Neurol 1959;1:130–154. nation. Child’s Brain 1980;7:132–145.
156. Windle W. Brain damage by asphyxia at birth. Sci Am 1969;221:77–84. 181. Azzarelli B, Caldemeyer KS, Phillips JP, DeMyer WE. Hypoxic-ischemic
157. Windle WF. Asphyxia at birth, a major factor in mental retardation: sug- encephalopathy in areas of primary myelination: a neuroimaging and PET
gestions for prevention based on experiments in monkeys. Proc Am Psy- study. Pediatr Neurol 1996;14:108–116.
chopathol Assn 1967;56:140–147. 182. Chugani HT, Phelps ME, Mazziotta JC. Positron emission tomography study
158. Myers RE. Two patterns of perinatal brain damage and the conditions of of human brain functional development. Ann Neurol 1987;22:487–497.
occurrence. Am J Obstet Gynecol 1972;112:246–276. 183. Kato T, Okuyama K. Assessment of maturation and impairment of brain
159. Sotrel A, Lorenzo AV. Ultrastructure of blood vessels in the ganglionic by I-123 Iodoamphetamine SPECT and MR Imaging in children. Showa
eminence of premature rabbits with spontaneous germinal matrix hem- University Journal of Medical Sciences 1993;5:99–115.
orrhages. J Neuropathol Exp Neurol 1989;48:462–482. 184. Tokumaru AM, Barkovich AJ, O’uchi T, Matsuo T, Kusano S. The evo-
160. Volpe JJ. Intracranial hemorrhage: Subdural, Primary Subarachnoid, lution of cerebral blood flow in the developing brain: evaluation with
Cerebellar, Intraentricular (Term Infant), and Miscellaneous. In: Volpe Iodine-123 Iodoamphetamine SPECT and MR imaging correlation. Am
JJ, ed. Neurology of the Newborn, 5th ed. Philadelphia: Saunders-Elsevier; J Neuroradiol 1999;20:845–852.
2008:483–516. 185. Vigneron DB, Barkovich AJ, Noworolski SM, et al. Three-Dimensional
161. Azzopardi D, Wyatt JS, Cady EB, et al. Prognosis of newborn infants with Proton MR Spectroscopic Imaging of Premature and Term Neonates.
hypoxic-ischemic brain injury assessed by phosphorus magnetic reso- Am J Neuroradiol 2001;22:1424–1433.
nance spectroscopy. Pediatr Res 1989;25:445–451. 186. Hasegawa M, Houdou S, Mito T, Takashima S, Asanuma K, Ohno T. Devel-
162. Penrice J, Lorek A, Cady EB, et al. Proton magnetic resonance spectros- opment of myelination in the human fetal and infant cerebrum: a myelin
copy of the brain during acute hypoxia-ischemia and delayed cerebral basic protein immunohistochemical study. Brain Dev 1992;14:1–6.
energy failure in the newborn piglet. Pediatr Res 1997;41:795–802. 187. Johnston MV, Trescher WH, Ishida A, Nakajima W. Neurobiology of
163. Qiao M, Malisza KL, Del Bigio MR, Tuor UI. Transient hypoxia-ischemia hypoxic-ischemic injury in the developing brain. Pediatr Res 2001;49:
in rats: changes in diffusion-sensitive MR imaging findings, extracellular 735–741.
space, and Na+-K+ adenosine triphosphatase and cytochrome oxidase 188. Van den Bergh R. Centrifugal elements in the vascular pattern of the deep
activity. Radiology 2002;223:65–75. intracerebral blood supply. Angiologica 1969;20:88–98.
164. Miyasaka N, Kuroiwa T, Zhao FY, et al. Cerebral ischemic hypoxia: dis- 189. Takashima S, Tanaka K. Development of the cerebrovascular architec-
crepancy between apparent diffusion coefficients and histologic changes ture and its relationship to periventricular leukomalacia. Arch Neurol
in rats. Radiology 2000;215:199–204. 1978;35:11–16.
165. Vannucci RC, Towfighi J, Vannucci SJ. Secondary energy failure after cere- 190. deReuck J. The human periventricular arterial blood supply and the anat-
bral hypoxia-ischemia in the immature rat. J Cereb Blood Flow Metabol omy of cerebral infarctions. Europ Neurol 1971; 5:321–334.
2004;24:1090–1097. 191. Kuban KCK, Gilles FH. Human telencephalic angiogenesis. Ann Neurol
166. Steinlin M, Dirr R, Martin E, et al. MRI following severe perinatal asphyxia: 1985;17:539–548.
preliminary experience. Pediatr Neurol 1991;7:164–170. 192. Nelson MD, Jr, Gonzalez-Gomez I, Gilles FH. The search for human
167. Keeney SE, Adcock EW, McArdle CB. Prospective observations of telencephalic ventriculofugal arteries. Am J Neuroradiol 1991;12:215–222.
100 high-risk neonates by high field (1.5 Tesla) magnetic resonance imag- 193. McClure MM, Riddle A, Manese M, et al. Cerebral blood flow heteroge-
ing of the central nervous system: I. Intraventricular and extracerebral neity in preterm sheep: lack of physiologic support for vascular bound-
lesions. Pediatrics 1991;87:421–430. ary zones in fetal cerebral white matter. J Cereb Blood Flow Metabol
168. Keeney S, Adcock EW, McArdle CB. Prospective observations of 100 high- 2008;28:995–1008.
risk neonates by high field (1.5 Tesla) magnetic resonance imaging of 194. Back S, Han B, Luo N, et al. Selective vulnerability of late oligodendrocyte
the central nervous system: II. Lesions associated with hypoxic-ischemic progenitors to hypoxia-ischemia. J Neurosci 2002;22:455–463.
encephalopathy. Pediatrics 1991;87:431–438. 195. Follett PL, Deng W, Dai W, et al. Glutamate receptor-mediated oligoden-
169. Barkovich AJ, Westmark KD, Ferriero DM, Sola A, Partridge JC. Perina- drocyte toxicity in periventriular leukomalacia: a protective role for topi-
tal asphyxia: MR findings in the first 10 days. Am J Neuroradiol 1995;16: ramate. J Neurosci 2004;24:4412–4420.
427–438. 196. Talos DM, Follett PL, Folkerth RD, et al. Developmental regulation of
170. Barkovich AJ. MR and CT evaluation of profound neonatal and infantile alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor
asphyxia. Am J Neuroradiol 1992;13:959–972. subunit expression in forebrain and relationship to regional susceptibility
171. Wiklund L-M, Uvebrant P, Flodmark O. Morphology of cerebral lesions in to hypoxic/ischemic injury. II. Human cerebral white matter and cortex.
children with congenital hemiplegia: a study with computed tomography. J Comp Neurol 2006;497:61–77.
Neuroradiology 1990;32:179–186. 197. Folkerth RD. Neuropathologic substrate of cerebral palsy. J Child Neurol
172. Koeda T, Suganuma I, Kohno Y, Takamatsu T, Takeshita K. MR imaging 2005;20:940–949.
of spastic diplegia: comparative study between preterm and term infants. 198. Back S, Luo N, Borenstein N, Levine J, Volpe J, Kinney H. Late oligodendro-
Neuroradiology 1990;32:187–190. cyte progenitors coincide with the developmental window of vulnerability
173. Rutherford MA, Pennock JM, Dubowitz LMS. Cranial ultrasound and for human perinatal white matter injury. J Neurosci 2001;21:1302–1312.
magnetic resonance imaging in hypoxic-ischemic encephalopathy: a com- 199. Flodmark O, Roland EH, Hill A, Whitfield MF. Periventricular leukomala-
parison with outcome. Dev Med Child Neurol 1994;36:813–825. cia: radiologic diagnosis. Radiology 1987;162:119–124.
174. Okereafor A, Allsop JM, Counsell SJ, et al. Patterns of brain injury in neo- 200. Flodmark O, Lupton B, Li D, et al. MR imaging of periventricular
nates exposed to perinatal sentinel events. Pediatrics 2008;121:906–914. leukomalacia in childhood. Am J Neuroradiol 1989;10:111–118.

Barkovich_Chap04.indd 354 5/25/2011 11:39:41 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 355

201. Ahdab Barmada M, Moossy J, Painter M. Pontosubicular necrosis and 225. Volpe JJ. Hypoxic-ischemic encephalopathy: clinical aspects. In: Volpe JJ,
hyperoxemia. Pediatrics 1980;66:840–847. ed. Neurology of the Newborn, 5th ed. Philadelphia: Saunders-Elsevier;
202. Hashimoto K, Takeuchi Y, Takeshima S. Hypocarbia as a pathogenic factor 2008:400–480.
in pontosubicular necrosis. Brain Dev 1995;13:155–157. 226. Weisglas-Kuperus N, Baerts W, Fetter W, et al. Minor neurological dys-
203. Skullerud K, Westre B. Frequency and prognostic significance of germi- function and quality of movement in relation to neonatal cerebral damage
nal matrix hemorrhage, periventricular leukomalacia and pontosubicular and subsequent development. Dev Med Child Neurol 1994;36:727–735.
necrosis in preterm neonates. Acta Neuropathol Berl 1986;70:257–261. 227. Pinto-Martin JA, Dobson V, Cnaan A, Zhao H, Paneth NS. Vision out-
204. Sohma O, Mito T, Mizuguchi M, Takashima S. The prenatal age critical come at age 2 years in a low birth weight population. Pediatr Neurol
for the development of the pontosubicular necrosis. Acta Neuropathol Berl 1996;14:281–287.
1995;90:155–157. 228. Schenk-Rootlieb AJ, van Nieuwenhuizen O, van der Graaf Y, Wittebol-
205. Kinnala A, Suhonen Polvi H, Aarimaa T, et al. Cerebral metabolic rate Post D, Willemse J. The prevalence of cerebral visual disturbance in chil-
for glucose during the first six months of life: an FDG positron emission dren with cerebral palsy. Dev Med Child Neurol 1992;34:473–480.
tomography study. Arch Dis Child Fetal Neonatal Ed 1996;74:F153-F157. 229. Uggetti C, Egitto ME, Fazzi E, et al. Cerebral visual impairment in
206. Martin JA, Hamilton BE, Ventura SJ, Menacker F, Park MM. Births: final periventricular leukomalacia: MR correlation. Am J Neuroradiol 1996;17:
data for 2000. Natl Vital Stat Rep 2002;50:1–101. 979–985.
207. Tin W, Wariyar U, Hey E. Changing prognosis for babies of less than 230. van den Hout BM, Stiers P, Haers M, et al. Relation between visual per-
28 weeks gestation in the north of England between 1983 and 1994. ceptual impairment and neonatal ultrasound diagnosis of haemorrhagic-
Northern Neonatal Network. Br Med J 1997;314:107–111. ischemic brain lesions in 5 year old children. Dev Med Child Neurol
208. Horwood LJ, Mogridge N, Darlow BA. Cognitive, educational and 2000;42:376–386.
behavoral outcomes at 7 to 8 years in a national very low birthweight 231. Shah DK, Guinane C, August P, et al. Reduced Occipital Regional Volumes
cohort. Arch Dis Child Fetal Neonatal Ed 1998;79:12–20. at Term Predict Impaired Visual Function in Early Childhood in Very Low
209. Emslie HC, Wardle SP, Sims DG, Chiswick ML, D’Souza SW. Increased Birth Weight Infants. Invest Ophthalmol Vis Sci 2006;47(8):3366–3373.
survival and deteriorating developmental outcome in 23–25 weeks gesta- 232. Berman JI, Glass HC, Miller SP, et al. Quantitative fiber tracking analysis
tion infants, 1990–1994 compared with 1984–1989. Arch Dis Child Fetal of the optic radiation correlated with visual performance in premature
Neonatal Ed 1998;78:99–104. newborns. Am J Neuroradiol 2009;30:120–124.
210. Suzuki J, Ito M. Coincidence patterns of cerebral palsy in Shiga Prefecture, 233. Glass HC, Berman JI, Norcia AM, et al. Quantitative fiber tracking of the
Japan, 1977–1991. Brain Dev 2002;24:39–48. optic radiation is correlated with visual-evoked potential amplitude in
211. Wilson-Costello D, Friedman H, Minich N, Fanaroff AA, Hack M. preterm infants. Am J Neuroradiol 2010;31:1424–1429.
Improved Survival Rates With Increased Neurodevelopmental Dis- 234. Jacobson L, Ek U, Fernell E, Flodmark O, Broberger U. Visual impairment
ability for Extremely Low Birth Weight Infants in the 1990s. Pediatrics in preterm children with periventricular leukomalacia—visual, cognitive,
2005;115(4):997–1003. and neuropaediatric characteristics related to cerebral imaging. Dev Med
212. Larroque B, Ancel P, Marret S, et al. Neurodevelopmental disabilities and Child Neurol 1996;38:724–735.
special care of 5-year-old children born before 33 weeks of gestation (the 235. Brodsky M, Fray K, Glasier C. Perinatal cortical and subcortical visual loss:
EPIPAGE study): a longitudinal cohort study. Lancet 2008;371:813–820. mechanisms of injury and associated ophthalmologic signs. Ophthalmol-
213. Hack M, Taylor HG, Drotar D, et al. Chronic conditions, functional ogy 2002;109:85–94.
limitations, and special health care needs of school-aged children born 236. Glass HC, Fujimoto S, Ceppi-Cozzio C, et al. White-Matter Injury is
with extremely low-birth-weight in the 1990s. J Am Med Assn 2005;294: Associated With Impaired Gaze in Premature Infants. Pediatr Neurol
318–325. 2008;38(1):10–15.
214. Miller SP, Ferriero DM, Leonard C, et al. Early Brain Injury in Prema- 237. Roth SC, Baudin J, Pezzani-Goldsmith M, Townsend J, Reynolds EOR,
ture Newborns Detected with Magnetic Resonance Imaging is Associated Stewart AL. Relation between neurodevelopmental status of very preterm
with Adverse Early Neurodevelopmental Outcome. J Pediatr 2005;147(5): infants at one and eight years. Dev Med Child Neurol 1994;36:1049–1062.
609–616. 238. Roth SC, Baudin J, McCormick DC, et al. Relation between ultrasound
215. Wood NS, Marlow N, Costeloe K, Gibson AT, Wilkinson AR. Neurologic appearance of the brain of very preterm infants and neurodevelopmental
and developmental disability after extremely preterm birth. N Eng J Med impairment at eight years. Dev Med Child Neurol 1993;35:755–768.
2000;343:378–384. 239. Bhutta A, Cleves M, Casey P, Cradock M, Anand K. Cognitive and behav-
216. Arnaud C, Daubisse-Marliac L, White-Koning M, et al. Prevalence and ioral outcomes of school-aged children who were born preterm: a meta-
Associated Factors of Minor Neuromotor Dysfunctions at Age 5 Years analysis. JAMA 2002;288:728–737.
in Prematurely Born Children: The EPIPAGE Study. Arch Pediatr Adolesc 240. Hack M, Fanaroff AA. Outcomes of children of extremely low birthweight
Med November 1, 2007 2007;161(11):1053–1061. and gestational age in the 1990’s. Early Hum Dev 1999;53:193–218.
217. Hack M, Flannery D, Schluchter M, Cartar L, Borawski E, Klein N. Out- 241. Vohr B, Wright L, Dusick A, et al. Neurodevelopmental and functional
comes in young adulthood for very-low-birth-weight infants. N Engl J outcomes of extremely low birth weight infants in the National Institute
Med 2002;346:149–157. of Child Health and Human Development Neonatal Research Network,
218. Lindstrom K, Winbladh B, Haglund B, Hjern A. Preterm Infants as Young 1993–1994. Pediatrics 2000;105:1216–1226.
Adults: A Swedish National Cohort Study. Pediatrics 2007;120(1):70–77. 242. Ramaekers G. Embryology and anatomy of the corpus callosum. In: Ramaek-
219. Greene M. Outcomes of very low birth weight in young adults. N Engl J ers G, Njiokikjtien C, eds. Pediatric Behavioural Neurology, Vol 3. The Child’s
Med 2002;346:146–148. Corpus Callosum. Amsterdam, The Netherlands: Suyi, 1991:24–39.
220. Back SA. Perinatal white matter injury: The changing spectrum of pathol- 243. Kostovic I, Judas M. Prolonged coexistence of transient and permanent
ogy and emerging insights into pathogenetic mechanisms. Ment Retard circuitry elements in the developing cerebral cortex of fetuses and preterm
Dev Disabil Res Rev 2006;12(2):129–140. infants. Dev Med Child Neurol 2006;48:388–393.
221. Riddle A, Luo N, Manese M, et al. Spatial heterogeneity in oligodendro- 244. Judas M, Rados M, Jovanov-Milosevic N, Hrabac P, Stern-Padovan R,
cyte lineage maturation and not cerebral blood flow predicts fetal ovine Kostovic I. Structural, immunocytochemical, and MR imaging proper-
periventricular white matter injury. J Neurosci 2006;26(11):3045–3055. ties of periventricular crossroads of growing cortical pathways in preterm
222. Greisen G. Ischemia of the preterm brain. Biol Neonate 1992;62:243–247. infants. Am J Neuroradiol 2005;26:2671–2784.
223. Volpe JJ. Brain injury in the premature infant—current concepts of patho- 245. Tam E, Ferriero DM, Xu D, et al. Cerebellar development in the preterm
genesis and prevention. Biol Neonate 1992;62(4):231–242. neonate: effect of supratentorial brain injury. Pediatr Res 2009;66:102–106.
224. Volpe JJ. Brain injury in premature infants: a complex amalgam of destruc- 246. Limperopoulos C, Bassan H, Gauvreau K, et al. Does cerebellar injury
tive and developmental disturbances. Lancet Neurol 2009;8:110–124. in premature infants contribute to the high prevalence of long-term

Barkovich_Chap04.indd 355 5/25/2011 11:39:41 AM


356 Pediatric Neuroimaging

cognitive, learning, and behavioral disability in survivors? Pediatrics 271. Takashima S, MitokT, Ando Y. Pathogenesis of periventricular white
2007;120(3):584–593. matter hemorrhages in preterm infants. Brain Dev 1986;8:25–30.
247. Mercuri E, He J, Curati WL, Dubowitz LMS, Cowan FM, Bydder GM. Cer- 272. Pikus HJ, Levy ML, Gans W, Mendel E, McComb JG. Outcome, cost analy-
ebellar infarction and atrophy in infants and children with a history of sis, and long-term follow-up in preterm infants with massive grade IV ger-
premature birth. Pediate Radiol 1997;27:139–143. minal matrix hemorrhage and progressive hydrocephalus. Neurosurgery
248. Coakley FV, Hricak H, Filly RA, Barkovich AJ, Harrison MR. Complex 1997;40:983–989.
fetal disorders: effect of MR imaging on management—preliminary clini- 273. Volpe JJ. Hypoxic-ischemic encephalopathy: clinical aspects. In: Volpe JJ, ed.
cal experience. Radiology 1999;213:691–696. Neurology of the Newborn, 3rd ed. Philadelphia: Saunders, 1995:314–369.
249. Fukui K, Morioka T, Nishio S, et al. Fetal germinal matrix and intraven- 274. Glass HC, Bonifacio SL, Chau V, et al. Recurrent postnatal infections are
tricular haemorrhage diagnosed by MRI. Neuroradiology 2001;43:68–72. associated with progressive white matter injury in premature infants. Pedi-
250. Levine D, Barnes P, Madsen JR, Li W, Edelman RR. Fetal central ner- atrics 2008;122:299–305.
vous system anomalies: MR imaging augments sonographic diagnosis. 275. McQuillen PS, Barkovich AJ, Hamrick SEG, et al. Temporal and anatomic
Radiology 1997;204:635–642. risk profile of brain injury with neonatal repair of congenital heart defects.
251. Simon EM, Coakley FV, Goldstein RB, Filly RA, Broderick KS, Barkovich Stroke 2007;38(2):736–741.
AJ. Fast magnetic resonance imaging of fetal central nervous system 276. Shuman RM, Selednik LJ. Periventricular leukomalacia: a one year autopsy
anomalies in utero: 3 year experience. Am J Neuroradiol 2000;21. study. Arch Neurol 1980;37:231–235.
252. de Vries L, Roelants-van Rijn A, Rademaker K, Van Haastert I, Beek F, 277. Hesser U, Katz-Salamon M, Mortensson W, Flodmark O, Forssberg H.
Groenendaal F. Unilateral parenchymal haemorrhagic infarction in the Diagnosis of intracranial lesions in very-low-birthweight infants by ultra-
preterm infant. Eur J Paediatr Neurol 2001;5:139–149. sound: incidence and association with potential risk factors. Acta Pediatr
253. Funato M, Tamai H, Noma K, et al. Clinical events in association with Suppl 1997;419:16–26.
timing of intraventricular hemorrhage in preterm infants. J Pediatr 278. Zupan V, Gonzalez P, Lacaze-Masmonteil T, et al. Periventricular leu-
1992;121:614–619. komalacia: risk factors revisited. Dev Med Child Neurol 1996;38:
254. Funato M, Tamai H, Takeda Z. Moment of intraventricular hemorrhage— 1061–1067.
clinical pathogenic events. In: Lou HC, Greisen G, Falck Larsen J, eds. 279. Claris O, Besnier S, Lapillonne A, et al. Incidence of ischemic-hemorrhagic
Brain Lesions in the Newborn, Alfred Benzon Symposium 37. Copenhagen: cerebral lesions in premature infants of gestational age ≤28 weeks: a pro-
Munksgaard, 1994:456–466. spective ultrasound study. Biol Neonate 1996;70:29–34.
255. Hambleton G, Wigglesworth JS. Origin of intraventricular hemorrhage in 280. Inder T, Anderson N, Spencer C, Wells S, Volpe J. White matter injury in
the pre-term infant. Arch Dis Child 1976; 51:651–660. the premature infant: a comparison between serial cranial sonographic
256. Wigglesworth JS, Pape KE. An integrated model for hemorrhage and isch- and MR findings at term. Am J Neuroradiol 2003;24:805–809.
emic lesions in the newborn brain. Early Hem Dev 1978;2:179–199. 281. de Vries LS, Van Haastert IL, Rademaker KJ, Koopman C, Groenendaal F.
257. Paneth N, Pinto-Martin J, Gardiner J, et al. Incidence and timing of germi- Ultrasound abnormalities preceding cerebral palsy in high-risk preterm
nal matrix/intraventricular hemorrhage in low birth weight infants. Am J infants. J Pediatr 2004;144(6):815–820.
Epidemiol 1993;137:1167–1176. 282. Cornette LG, Tanner SF, Ramenghi LA, et al. Magnetic resonance imag-
258. Kriegstein AR, Noctor SC. Patterns of neuronal migration in the embry- ing of the infant brain: anatomical characteristics and clinical signifi-
onic cortex. Trends Neurosci 2004;27:392–399. cance of punctate lesions. Arch Dis Child Fetal Neonatal Ed 2002;86(3):
259. Rakic P, Sidman R. Histogenesis of cortical layers in human cerebellum, F171–F177.
particularly the lamina dissecans. J Comp Neurol 1970;139:473–500. 283. Miller S, Cozzio C, Goldstein R, et al. Comparing the diagnosis of white
260. Merrill JD, Piecuch RE, Fell SC, Barkovich AJ, Goldstein RB. A new pattern matter injury in premature newborns with serial MR imaging and trans-
of cerebellar hemorrhages in preterm infants. Pediatrics 1998;102:e62. fontanel ultrasonography findings. Am J Neuroradiol 2003;24:1661–1669.
261. Limperopoulos C, Benson CB, Bassan H, et al. Cerebellar hemorrhage in 284. Counsell SJ, Allsop JM, Harrison MC, et al. Diffusion-weighted imag-
the preterm infant: ultrasonographic findings and risk factors. Pediatrics ing of the brain in preterm infants with focal and diffuse white matter
2005;116(3):717–724. abnormality. Pediatrics 2003;112(1):1–7.
262. Steggerda SJ, Leijser LM, Wiggers-de Bruïne FT, van der Grond J, Walther 285. Litt J, Taylor HG, Klein N, Hack M. Learning disabilities in children with
FJ, van Wezel-Meijler G. Cerebellar injury in preterm infants: incidence very low birthweight: Prevalence, neuropsychological correlates, and edu-
and findings on US and MR images. Radiology 2009;252:190–199. cation interventions. J Learn Disabil 2005;38:130.
263. Limperopoulos C, Soul JS, Haidar H, et al. Impaired trophic interactions 286. Verma U, Tehani N, Klein S, et al. Obstetric antecedents of intraventricu-
between the cerebellum and the cerebrum among preterm infants. Pediat- lar hemorrhage and periventricular leukomalacia in the low-birth-weight
rics 2005;116(4):844–850. neonate. Am J Obstet Gynecol 1997;176:275–281.
264. Volpe JJ. Hypoxic-ischemic encephalopathy: neuropathology and patho- 287. Nelson K, Ellenberg J. Antecedents of cerebral palsy: multivariate analysis
genesis. In: Volpe JJ, ed. Neurology of the Newborn, 3rd ed. Philadelphia: of risk. N Engl J Med 1986;315:81–86.
Saunders; 1995:279–313. 288. McQuillen P, Sheldon R, Shatz C, Ferriero D. Selective vulnerability
265. Papile LA, Brustein J, Burstein R, Koffer H. Incidence and evolution of of subplate neurons after early neonatal hypoxia-ischemia. J Neurosci
subependymal and intraventricular hemorrhage: a study of infants with 2003;23:3308–3315.
birth weights less than 1500 gm. J Pediatr 1978;92:529–534. 289. Marret S, Mukendi R, Gadisseux J-F, Gressens P, Evrard P. Effect of iboten-
266. Gould SJ, Howard S, Hope PL, Reynolds EOR. Periventricular intraparen- tate on brain development: an excitotoxic mouse model of microgyria and
chymal cerebral hemorrhage in preterm infants: the role of venous infarc- posthypoxic-like lesions. J Neuropathol Exp Neurol 1995;54:358–370.
tion. J Pathol 1987;151:197–202. 290. Banker BQ, Larroche JC. Periventricular leukomalacia of infancy: a form
267. Volpe JJ. Brain injury in the premature infant: neuropathology, clinical of neonatal anoxic encephalopathy. Arch Neurol 1962;7:386–410.
aspects, and pathogenesis. MRDD Res Rev 1997;3:3–12. 291. Inder TE, Wells SJ, Mogridge MB, Spencer C, Volpe JJ. Defining the nature
268. Bassan H, Benson CB, Limperopoulos C, et al. Ultrasonographic Features of the cerebral abnormalities in the premature infant: a qualitative mag-
and Severity Scoring of Periventricular Hemorrhagic Infarction in Rela- netic resonance imaging study. J Pediatr 2003;143(2):171–179.
tion to Risk Factors and Outcome. Pediatrics 2006;117(6):2111–2118. 292. Woodward LJ, Anderson PJ, Austin NC, Howard K, Inder TE. Neonatal
269. van de Bor M, Ens-Dokkum M, Schreuder AM, Veen S, Brand R, Verloove- MRI to predict neurodevelopmental outcomes in preterm infants. N Eng J
Vanhorick SP. Outcome of periventricular-intraventricular hemorrhage at Med 2006;355:685–694.
five years of age. Dev Med Child Neurol 1993;35:33–41. 293. Sie L, van der Knaap M, van Wezel-Meijler G, Taets van Amerongen A,
270. Whitaker AH, Feldman JF, Van Rossem R, et al. Neonatal cranial ultra- Lafeber H, Valk J. Early MR features of hypoxic-ischemic brain injury in
sound abnormalities in low birth weight infants: relation to cognitive out- neonates with periventricular densities on sonograms. Am J Neuroradiol
comes at six years of age. Pediatrics 1996;98:719–729. 2000;21:852–861.

Barkovich_Chap04.indd 356 5/25/2011 11:39:41 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 357

294. Limperopoulos C, Soul JS, Gauvreau K, et al. Late gestation cerebellar 318. Kubota T, Okumura A, Hayakawa F, et al. Relation between the date of cyst
growth is rapid and impeded by premature birth. Pediatrics 2005;115: formation observable on ultrasonography and the iming of injury deter-
688–695. mined by serial electroencephalography in preterm infants with periven-
295. Srinivasan L, Allsop J, Counsell SJ, Boardman JP, Edwards AD, Rutherford tricular leukomalacia. Brain Dev 2001;23:390–394.
M. Smaller cerebellar volumes in very preterm infants at term-equivalent 319. de Vries LS, Eken P, Dubowitz LMS. The spectrum of leukomalacia using
age are associated with the presence of supratentorial lesions. Am J Neuro- cranial ultrasound. Beh Brain Res 1992;49:1–6.
radiol 2006;27(3):573–579. 320. Dammann O, Leviton A. Duration of transient hyperechoic images of
296. Dumoulin CL, Rohling KW, Piel JE, et al. Magnetic resonance imaging white matter in very-low-birthweight infants: a proposed classification.
compatible neonate incubator. Concepts Magn Res (Magn Reson Eng). Dev Med Child Neurol 1997;39:2–5.
2002;15(2):117–128. 321. Schouman-Clays E, Henry-Feugeas M-C, Roset F, et al. Periventricular
297. Miller S, Vigneron D, Henry R, et al. Serial quantitative diffusion tensor leukomalacia: correlation between MR imaging and autopsy findings dur-
MRI of the premature brain: development in newborns with and without ing the first 2 months of life. Radiology 1993;189:59–64.
injury. J Magn Reson Imaging 2002;16:621–632. 322. Leijser L, de Bruïne F, van der Grond J, Steggerda S, Walther F, van
298. Siebert JJ, Avva R, Hronas TN, et al. Use of power Doppler in pediatric Wezel-Meijler G. Is sequential cranial ultrasound reliable for detection of
neurosonography: a pictorial essay. Radiographics 1998;18:879–890. white matter injury in very preterm infants? Neuroradiology 2010;52(5):
299. Zuerrer M, Martin E, Boltshauser E. MR imaging of intracranial hem- 397–406.
orrhage in neonates and infants at 2.35 Tesla. Neuroradiology 1991;33: 323. Felderhoff-Mueser U, Rutherford M, Squier W, et al. Relationship between
223–229. MR imaging and histopathologic findings of the brain in extremely sick
300. Fukumizu M, Takashima S, Becker LE. Neonatal posthemorrhagic hydro- preterm infants. Am J Neuroradiol 1999;20:1349–1357.
cephalus: neuropathologic and immunohistochemical studies. Pediatr 324. Khwaja O, Volpe JJ. Pathogenesis of cerebral white matter injury of prema-
Neurol 1995;13:230–234. turity. Arch Dis Child Fetal Neonatal Ed 2008;93:F153–F161.
301. Dudink J, Lequin M, Weisglas-Kuperus N, Conneman N, Goudoever Jv, 325. Cheong JLY, Thompson DK, Wang HX, et al. Abnormal white matter sig-
Govaert P. Venous subtypes of preterm periventricular haemorrhagic nal on MR imaging is related to abnormal tissue microstructure. Am J
infarction. Arch Dis Child Fetal Neonatal Ed 2008;93:F201–206. Neuroradiol 2009;30:623–628.
302. Govaert P, Smets K, Matthys E, Oostra A. Neonatal focal temporal lobe 326. Counsell S, Edwards AD, Chew AT, et al. Specific relations between neu-
or atrial wall haemorrhagic infarction. Arch Dis Child Fetal Neonatal Ed rodevelopmental abilities and white matter microstructure in children
1999;81(3):F211–F216. born preterm. Brain 2008;131:3201–3208.
303. Taylor G. Effect of germinal matrix hemorrhage on terminal vein position 327. Sie LTL, Hart AAM, van Hof J, et al. Predictive value of neonatal MRI
and patency. Pediatr Radiol 1995;25:S37–S40. with respect to late MRI findings and clinical outcome. A study in infants
304. Garg A, Berman JI, Xu D, et al. Abnormal white matter and gray matter mat- with periventricular densities on neonatal ultrasound. Neuropediatrics
uration in premature neonates with periventricular hemorrhagic infarction: 2005;36:78–89.
a diffusion tensor imaging study. ASNR Annual Meeting, June 2008 328. Anderson NG, Laurent I, Cook N, Woodward L, Inder TE. Growth
305. Leijser LM, Liauw L, Veen S, de Boer IP, Walther FJ, van Wezel-Meijler G. rate of corpus callosum in very premature infants. Am J Neuroradiol
Comparing brain white matter on sequential cranial ultrasound and MRI 2005;26:2685–2690.
in very preterm infants. Neuroradiology 2008;50:799–811. 329. Counsell SJ, Rutherford MA, Cowan FM, Edwards AD. Magnetic reso-
306. Schellinger D, Grant EG, Richardson JD. Cystic periventricular leukomal- nance imaging of preterm brain injury. Arch Dis Child Fetal Neonatal Ed.
acia: sonographic and CT findings. Am J Neuroradiol 1984;5:439–445. 2003;88(4):F269–F274.
307. Vannucci RC, Christensen MA, Yager JY. Nature, time-course, and extent 330. Childs AM, Ramenghi LA, Evans DJ, et al. MR features of developing
of cerebral edema in perinatal hypoxic-ischemic brain damage. Pediatr periventricular white matter in preterm infants: evidence of glial cell
Neurol 1993;9:29–34. migration. Am J Neuroradiol 1998;19(5):971–976.
308. Grant EG, Schellinger D, Richardson JD, Coffey ML, Smirniotopoulous JG. 331. Garel C. New advances in fetal MR neuroimaging. Pediatr Radiol
Echogenic periventricular halo: normal sonographic finding or neonatal 2006;36:621–625.
cerebral hemorrhage? Am J Neuroradiol 1983;4:43–46. 332. Kostovic I, Judas M, Radios M, Hrabac P. Laminar organization of the
309. Dipietro MA, Brody BA, Teele RL. Periventricular echogenic “blush” on cra- human fetal cerebrum revealed by histochemical markers and magnetic
nial sonography: pathologic correlates. Am J Neuroradiol 1986;7:305–310. resonance imaging. Cereb Cortex 2002;12:536–544.
310. Baarsma R, Laurini RN, Baerts W, Okken A. Reliability of sonogra- 333. Girard NJ, Raybaud CA. In vivo MRI of fetal brain cellular migration.
phy in non-hemorrhagic periventricular leucomalacia. Pediatr Radiol J Comput Assist Tomogr 1992;16:265–267.
1987;17:189–191. 334. Bartha AI, Yap KRL, Miller SP, et al. The Normal Neonatal Brain: MR
311. Fazzi E, Orcesi S, Caffi L, et al. Neurodevelopmental outcome at 5–7 years Imaging, Diffusion Tensor Imaging, and 3D MR Spectroscopy in Healthy
in preterm infants with periventricular leukomalacia. Neuropediatrics Term Neonates. Am J Neuroradiol 2007;28(6):1015–1021.
1994;25:134–139. 335. Inder T, Huppi P, Zientara G, et al. Early detection of periventricular leu-
312. O’Shea T, Kuban K, Allred E, et al. Neonatal cranial ultrasound lesions and komalacia by diffusion-weighted magnetic resonance imaging techniques.
developmental delays at 2 years of age among extremely low gestational J Pediatr 1999;134:631–634.
age children. Pediatrics 2008;122:e662–e669. 336. Bonifacio SL, Glass HC, Chau V, et al. Extreme premature birth is not
313. Horsch S, Muentjes C, Franz A, Roll C. Ultrasound diagnosis of brain associated with impaired development of brain microstructure. J Pediatr
atrophy is related to neurodevelopmental outcome in preterm infants. 2010;157:726–732.
Acta Paediatrica 2005;94:1815–1821. 337. Liauw L, van Wezel-Meijler G, Veen S, van Buchem M, van der Grond J.
314. Carson SC, Hertzberg BS, Bowie JD, Burger PC. Value of sonography in Do apparent diffusion coefficient measurements predict outcome in chil-
the diagnosis of intracranial hemorrhage and periventricular leukomala- dren with neonatal hypoxic-ischemic encephalopathy? Am J Neuroradiol
cia: a postmortem study of 35 cases. Am J Neuroradiol 1990;11:677–684. 2009;30:264–270.
315. Paneth N, Rudelli E, Monte W, et al. White matter necrosis in very low 338. Berman JI, Mukherjee P, Partridge SC, et al. Quantitative diffusion ten-
birth weight infants: neuropathologic and ultrasonographic findings in sor MRI fiber tractography of sensorimotor white matter development in
infants surviving six days or longer. J Pediatr 1990;116:975–984. premature infants. NeuroImage 2005;27(4):862–871.
316. Sauerbrei EE. Serial brain sonography in two children with leukomalacia 339. Bassi L, Ricci D, Volzone A, et al. Probabilistic diffusion tractography of
and cerebral palsy. J Can Assoc Radiol 1984;35:164–167. the optic radiations and visual function in preterm infants at term equiva-
317. Dubowitz LMS, Bydder GM, Mushin J. Developmental sequence of lent age. Brain 2008;131:573–582.
periventricular leukomalacia: correlation of ultrasound, clinical, and 340. Baker LL, Stevenson DK, Enzmann DR. End stage periventricular
nuclear magnetic resonance functions. Arch Dis Child 1985;60:349–355. leukomalacia: MR imaging evaluation. Radiology 1988;168:809–815.

Barkovich_Chap04.indd 357 5/25/2011 11:39:41 AM


358 Pediatric Neuroimaging

341. van de Bor M, Guit GL, Schreuder AM, et al. Does very preterm birth 363. Daneman A, Epelman M, Blaser S, Jarrin JR. Imaging of the brain in
impair myelination of the central nervous system? Neuropediatrics full term neonates: does sonography still play a role? Pediatr Radiol
1990;21(1):37–39. 2006;36:636–646.
342. van de Bor M, Guit G, Schreuder A, Wondergem J, Vielvoye GJ. Early 364. Adcock LM, Moore PJ, Schlesinger AE, Armstrong DL. Correlation of
detection of delayed myelination in preterm infants. Pediatrics 1989;84: ultrasound with postmortem neuropathologic studies in neonates. Pediatr
407–411. Neurol 1998;19:263–271.
343. Skranes J, Vik T, Nilsen G, et al. Cerebral magnetic resonance imaging and 365. Barkovich AJ, Westmark KD, Bedi HS, Partridge JC, Ferriero DM, Vign-
mental and motor function of very low birth weight infants at one year of eron DB. Proton spectroscopy and diffusion imaging on the first day
corrected age. Neuropediatrics 1993;24:256–262. of life after perinatal asphyxia: preliminary report. Am J Neuroradiol
344. Mercuri E, Jongmans M, Henderson S, et al. Evaluation of the corpus cal- 2001;22:1786–1794.
losum in clumsy children born prematurely: a functional and morpho- 366. Robertson R, Ben-Sira L, Barnes P, et al. MR line scan diffusion weighted
logical study. Neuropediatrics 1996;27:317–322. imaging of term neonates with perinatal brain ischemia. Am J Neuroradiol
345. Barkovich AJ, Norman D. Anomalies of the corpus callosum: Correlation 1999;20:1658–1670.
with further anomalies of the brain. Am J Neuroradiol 1988;9:493–501. 367. Dubois J, Hertz-Pannier L, Dehaene-Lambertz G, Cointepas Y, Le Bihan D.
346. Stewart A, Rifkin L, Amess P, et al. Brain structure and neurocognitive and Assessment of the early organization and maturation of infants’ cerebral
behavioural function in adolescents who were born very preterm. Lancet white matter fiber bundles: A feasibility study using quantitative diffusion
1999;353:1653–1657. tensor imaging and tractography. NeuroImage 2006;30(4):1121–1132.
347. Melhem E, Hoon A Jr, Ferrucci J Jr, et al. Periventricular leukomalacia: 368. Partridge SC, Mukherjee P, Berman JI, et al. Tractography-based quan-
relationship between lateral ventricular volume on brain MR images titation of diffusion tensor imaging parameters in white matter tracts of
and severity of cognitive and motor impairment. Radiology 2000;214: preterm newborns. J Magn Reson Imaging 2005;22(4):467–474.
199–204. 369. Hull J, Dodd KL. Falling incidence of hypoxic-ischemic encephalopathy in
348. Hüppi PS, Murphy B, Maier SE, et al. Microstructural brain development term infants. Br J Obstet Gynecol 1992;99:386–391.
after perinatal cerebral white matter injury assessed by diffusion tensor 370. Evans D, Levene M. Evidence of selection bias in preterm survival studies:
magnetic resonance imaging. Pediatrics 2001;107:455–460. a systematic review. Arch Dis Child Fetal Neonatal Ed 2001;82:F79–F84.
349. Hoon Jr A, Lawrie Jr W, Melhem E, et al. Diffusion tensor imaging of 371. du Plessis A, Volpe J. Perinatal brain injury in the preterm and term new-
periventricular leukomalacia shows affected sensory cortex white matter born. Curr Opin Neurol 2002;15:151–157.
pathways. Neurology 2002;59:752–756. 372. Pierrat V, Haouari N, Liska A, et al. Prevalence, causes, and outcomes at
350. Nagae LM, Hoon AH Jr., Stashinko E, et al. Diffusion Tensor Imaging 2 years of age of newborn encephalopathy: population based study. Arch
in Children with Periventricular Leukomalacia: Variability of Injuries to Dis Child Fetal Neonatal Ed 2005;90(3):F257–F261.
White Matter Tracts. Am J Neuroradiol 2007;28(7):1213–1222. 373. Blair E, Stanley J. Intrapartum asphyxia: a rare cause of cerebral palsy.
351. Nagy Z, Westerberg H, Skare S, et al. Preterm children have disturbances J Pediatr 1988;112:515–519.
of white matter at 11 years of age as shown by diffusion tensor imaging. 374. Truwit CL, Barkovich AJ, Koch TK, Ferriero DM. Cerebral palsy: MR find-
Pediatr Res 2003;54:672–679. ings in 40 patients. Am J Neuroradiol 1992;13:67–78.
352. Dudink J, Lequin M, van Pul C, et al. Fractional anisotropy in white 375. Miller SP, Ramaswamy V, Michelson D, et al. Patterns of brain injury in
matter tracts of very-low-birth-weight infants. Pediatric Radiology term neonatal encephalopathy. J Pediatr 2005;146:453–460.
2007;37(12):1216–1223. 376. Gonzalez FF, Miller SP. Does perinatal asphyxia impair cognitive func-
353. de la Monte SM, Hsu FI, Hedley-Whyte ET, Kupsky W. Morphometric tion without cerebral palsy? Arch Dis Child Fetal Neonatal Ed 2006;91:
analysis of the human infant brain: Effects of intraventricular hemorrhage F454–F459.
and periventricular leukomalacia. J Child Neurol 1989;4:101–110. 377. Steinman KJ, Gorno-Tempini ML, Glidden DV, et al. Neonatal watershed
354. Evrard P, Belpaire MC, Boog G, et al. Diagnostic antenatal des affections brain injury on magnetic resonance imaging correlates with verbal IQ at
du syteme nerveux central: resultats preliminaires d’une etude multi- 4 years. Pediatrics 2009;123:1025–1030.
centrique europeenne. Journal Français d’Echographie 1985;2:123–126. 378. Mercuri E, Atkinson J, Braddick O, et al. Visual function in full-term infants
355. Evrard P, Gadisseux JF, Gerriere G, Lyon G. Le developpment prena- with hypoxic-ischemic encephalopathy. Neuropediatrics 1997;28:155–161.
tal du systeme nerveux central et ses perturbations. In: Ferriere G, ed. 379. Foran A, Cinnante C, Groves AM, Azzopardi DV, Rutherford MA, Cowan
La Deficience Mentale: Causes, Prevention, Et Traitement Brussels: Prodim, FM. Patterns of brain injury and outcome in term neonates present-
1989:11–93. ing with postnatal collapse. Arch Dis Child Fetal Neonatal Ed 2008;94:
356. Messerschmidt A, Brugger PC, Boltshauser E, et al. Disruption of Cerebel- F168–F177.
lar Development: Potential Complication of Extreme Prematurity. Am J 380. Shankaran S, Pappas A, Laptook A, et al. Outcomes of safety and effec-
Neuroradiol 2005;26(7):1659–1667. tiveness in a multicenter randomized, controlled trial of whole-body
357. Shah DK, Anderson PJ, Carlin JB, et al. Reduction in cerebellar volumes hypothermia for neonatal hypoxic-ischemic encephalopathy. Pediatrics
in preterm infants: relationship to white matter injury and neurodevelop- 2008;122:e791–e798.
ment at two years of age. Pediatr Res 2006;60(1):97–102. 381. Fauchère JC, Dame C, Vonthein R, Arri SJ, Koller B, Bucher HU. Erythro-
358. Müller H, Beedgen B, Schenk JP, Troger J, Linderkamp O. Intracerebellar poitin for neuroprotection in preterm infants: feasibility and safety study.
hemorrhage in premature infants: sonographic detection and outcome. Paper presented at: European Society for Paediatric Research, October 7,
J Perinat Med 2007;35:67–70. 2007, 2007; Prague.
359. Aldinger KA, Lehmann OJ, Hudgins L, et al. FOXC1 is required for normal 382. Perlman JM. Intervention strategies for neonatal hypoxic-ischemic injury.
cerebellar development and is a major contributor to chromosome 6p25.3 Clin Ther 2006;28(9):1353–1365.
Dandy-Walker malformation. Nat Genet 2009;41:1037–1042. 383. Gilles FH, Leviton A, Dooling EC. The Developing Human Brain. Boston,
360. Logitharajah P, Rutherford MA, Cowan F. Hypoxic-ischaemic enceph- MA: John Wright-PSG, 1983.
alopathy in preterm infants: antecedent factors, brain imaging and out- 384. Barkovich AJ, Baranski K, Vigneron D, et al. Proton MR spectroscopy in
come. Pediatr Res 2009;66(2):222–229. the evaluation of asphyxiated term neonates. Am J Neurorad 1999;20:1399–
361. Leech RW, Alvord EC Jr. Anoxic ischemic encephalopathy in the human 1405.
neonatal period: The significance of brain stem involvement. Arch Neurol 385. Miller SP, Newton N, Ferriero DM, et al. Predictors of 30-month outcome
1977;34:109–113. following perinatal depression: role of proton MRS and socio-economic
362. Barr LL, McCullough PJ, Ball WS, Krasner BH, Garra BS, Deddens JA. factors. Pediatr Res 2002;52:71–77.
Quantitative sonographic feature analysis of clinical infant hypoxia: a 386. Inder TE. How low can I go? The impact of hypoglycemia on the imma-
pilot study. Am J Neuroradiol 1996;17:1025–1031. ture brain. Pediatrics 2008;122:440–441.

Barkovich_Chap04.indd 358 5/25/2011 11:39:42 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 359

387. Miyasaka N, Nagaoka T, Kuriowa T, et al. Histopathologic correlates of 411. Phillips R, Brandberg G, Hill A, Roland E, Poskitt K. Prevalence and prog-
temporal diffusion changes in a rat model of cerebral hypoxia/ischemia. nostic value of abnormal CT findings in 100 term asphyxiated newborns.
Am J Neuroradiol 2000;21:60–66. Radiology 1993;189P:287.
388. Vermeulen RJ, van Schie PE, Hendrikx L, et al. Diffusion-weighted and 412. Connolly B, Kelehan P, O’Brien N, et al. The echogenic thalamus in
conventional MR imaging in neonatal hypoxic ischemia: two-year hypoxic ischaemic encephalopathy. Pediatr Radiol 1994;24:268–271.
follow-up study. Radiology 2008;249:631–639. 413. Allison JW, Faddis LA, Kinder DL, Roberson PK, Glasier CM, Seibert JJ.
389. Hanrahan JD, Sargentoni J, Azzopardi D, et al. Cerebral metabolism within Intracranial resistive index (RI) values in normal term infants during the
18 hours of birth asphyxia: a proton magnetic resonance spectroscopy first day of life. Pediatr Radiol 2000;30:618–620.
study. Pediatr Res 1996;39:584–590. 414. Rutherford M, Counsell S, Allsop J, et al. Diffusion weighted magnetic
390. Hanrahan JD, Cox IJ, Azzopardi D, et al. Relation between proton mag- resonance imaging in term perinatal brain injury: a comparison with site
netic resonance spectroscopy within 18 hours of birth asphyxia and neu- of lesion and time from birth. Pediatrics 2004;114:1004–1014.
rodevelopment at 1 year of age. Dev Med Child Neurol 1999;41:76–82. 415. Boichot C, Walker PM, Durand C, et al. Term neonate prognoses after peri-
391. Barkovich AJ, Hajnal BL, Vigneron D, et al. Prediction of neuromotor natal asphyxia: contributions of MR imaging, MR spectroscopy, relaxation
outcome in perinatal asphyxia: evaluation of MR scoring systems. Am J times, and apparent diffusion coefficients. Radiology 2006;239(3):839–848.
Neuroradiol 1998;19:143–150. 416. Sargent MA, Poskitt KJ, Roland EH, Hill A, Hendson G. Cerebellar verm-
392. Johnson MA, Pennock JM, Bydder GM, et al. Clinical MR imaging of ian atrophy after neonatal hypoxic-ischemic encephalopathy. Am J Neuro-
the brain in children: normal and neurologic disease. Am J Roentgenol radiol 2004;25:1008–1015.
1983;141:1005–1018. 417. Connolly DJA, Widjaja E, Griffiths PD. Involvement of the anterior lobe
393. Johnson MA, Pennock JM, Bydder GM, et al. Serial MR imaging in neona- of the cerebellar vermis in perinatal profound hypoxia. Am J Neuroradiol
tal cerebral injury. Am J Neuroradiol 1987;8:83–92. 2007;28:16–19.
394. Byrne P, Welch R, Johnson MA, Darrah J, Piper M. Serial magnetic reso- 418. van Bogaert P, Baleriaux D, Christophe C, Szliwowski HB. MRI of patients
nance imaging in neonatal hypoxic-ischemic encephalopathy. J Pediatr with cerebral palsy and normal CT scan. Neuroradiology 1992;34:52–56.
1990;117:694–700. 419. Hope PL, Costello AM, Cady EB, et al. Cerebral energy metabolism stud-
395. Kuenzle C, Baenziger O, Martin E, et al. Prognostic value of early MR ied with phosphorus NMR spectroscopy in normal and birth-asphyxiated
imaging in term infants with severe perinatal asphyxia. Neuropediatrics infants. Lancet 1984;2:366–370.
1994;25:191–200. 420. Laptook AR, Corbett RJ, Uauy R, Mize C, Mendelsohn D, Nunnally RL.
396. Takashima S, Tanaka K. Subcortical leukomalacia, relationship to devel- Use of 31P magnetic resonance spectroscopy to characterize evolving brain
opment of the cerebral sulcus, and its vascular supply. Arch Neurol damage after perinatal asphyxia. Neurology 1989;39(5):709–712.
1978;35:470–476. 421. Moorcraft J, Bolas NM, Ives NK, et al. Spatially localized magnetic reso-
397. Roland EH, Poskitt K, Rodriguez E, Lupton BA, Hill A. Perinatal hypoxic- nance spectroscopy of the brains of normal and asphyxiated newborns.
ischemic thalamic injury: clinical features and neuroimaging. Ann Neurol Pediatrics 1991;87:273–282.
1998;44:161–166. 422. Martin E, Buchli R, Ritter S, et al. Diagnostic and prognostic value of cere-
398. Griffiths PD, Radon MR, Crossman AR, Zurakowski D, Connolly DJ. Ana- bral 31-P magnetic resonance spectroscopy in neonates with perinatal
tomic Localization of Dyskinesia in Children with “Profound” Perinatal asphyxia. Pediatr Res 1996;40:749–758.
Hypoxic-Ischemic Injury. Am J Neuroradiol 2010;31(3):436–441. 423. Peden CJ, Cowan FM, Bryant DJ, et al. Proton MR spectroscopy of the
399. Johnston MV, Hoon Jr A. Possible mechanisms in infants for selective basal brain in infants. J Comput Assist Tomogr 1990;14:886–894.
ganglia damage from asphyxia, kernicterus, or mitochondrial encephal- 424. Leth H, Toft PB, Peitersen B, Lou HC, Henriksen O. Use of brain lac-
opathies. J Child Neurol 2000;15:588–591. tate levels to predict outcome after perinatal asphyxia. Acta Paediatr
400. Pasternak JF, Gorey MT. The syndrome of acute near total intrauterine 1996;85:859–864.
asphyxia in the term infant. Pediatr Neurol 1998;18:391–398. 425. Groenendaal F, Veenhoven EH, van der Grond J, Jansen GH, Witkamp
401. Schneider H, Ballowitz L, Schachinger H, Hanefeld F, Droszus JU. Anoxic TD, de Vries L. Cerebral lactate and N-acetyl-aspartate/choline ratios in
encephalopathy with predominant involvement of basal ganglia, brain asphyxiated full-term neonates demonstrated in-vivo using proton mag-
stem, and spinal cord in the perinatal period. Acta Neuropathologica netic resonance spectroscopy. Pediatr Res 1994;35:148–151.
1975;32:287–298. 426. Holshouser BA, Ashwahl S, Luh GY, et al. Proton MR spectroscopy after
402. Mercuri E, Rutherford M, Barnett A, et al. MRI lesions and infants with acute central nervous system injury: outcome prediction in neonates,
neonatal encephalopathy. Is the Apgar score predictive? Neuropediatrics infants, and children. Radiology 1997;202:487–496.
2002;33:150–156. 427. Amess PN, Penrice J, Wylezinska M, et al. Early brain proton magnetic
403. Krägeloh-Mann I, Helber A, Mader I, et al. Bilateral lesions of thalamus resonance spectroscopy and neonatal neurology related to neurodevelop-
and basal ganglia: origin and outcome. Dev Med Child Neurol 2002;44: mental outcome at 1 year in term infants after presumed hypoxic-ischae-
477–484. mic brain injury. Dev Med Child Neurol 1999;41:436–445.
404. Menkes J, Curran JG. Clinical and magnetic resonance imaging cor- 428. Cheong JL, Cady EB, Penrice J, Wyatt JS, Cox IJ, Robertson NJ. Proton MR
relates in children with extrapyramidal cerebral palsy. Am J Neuroradiol spectroscopy in neonates with perinatal cerebral hypoxic-ischemic injury:
1994;15:451–457. metabolite peak-area ratios, relaxation times, and absolute concentra-
405. Kerrigan JF, Chugani HT, Phelps ME. Regional cerebral glucose metabo- tions. Am J Neuroradiol 2006;27:1546–1554.
lism in clinical subtypes of cerebral palsy. Pediatr Neurol 1991;7:415–425. 429. Penrice J, Cady EB, Lorek A, et al. Proton magnetic resonance spectros-
406. Hayashi M, Satoh J, Sakamoto K, Morimatsu Y. Clinical and neuropatho- copy of the brain in normal preterm and term infants and early changes
logical findings in severe athetoid cerebral palsy: a comparative study of after perinatal hypoxia-ischemia. Pediatr Res 1996;40:6–14.
globo-luysian and thalamo-putaminal groups. Brain Dev 1991;13:47–51. 430. McGuinness G, Weisz S, Bell W. CSF lactate levels in neonates.
407. Barnett A, Mercuri E, Rutherford M, et al. Neurological and perceptual- Effect of asphyxia, gestational age, and postnatal age. Am J Dis Child
motor outcome at 5–6 years of age in children with neonatal encephalopathy: 1983;137:48–50.
relationship with neonatal brain MRI. Neuropediatrics 2002;33:242–248. 431. Fernandez F, Verdu A, Quero J, et al. Cerbrospinal fluid lactate levels in
408. Arvidsson J, Hagberg B. Delayed onset dyskinetic “cerebral palsy”—a late term infants with perinatal hypoxia. Pediatr Neurol 1986;2:39–42.
effect of perinatal asphyxia? Acta Pediatr Scand 1990;79:1121–1123. 432. Auld KL, Ashwal S, Holshouser B, et al. Proton magnetic resonance spec-
409. Burke RE, Fahn S, Gold AP. Delayed-onset dystonia in patients with troscopy in children with acute central nervous system injury. Pediatr
“static” encephalopathy. J Neurol Neurosurg Psychiatry 1980;43:789–797. Neurol 1995;12:323–334.
410. Saint-Hilaire MH, Burke RE, Bressman SB, et al. Delayed-onset dystonia 433. Leth H, Toft PB, Pryds O, Petersen B, Lou HC, Henriksen O. Brain lactate
due to perinatal or early childhood asphyxia. Neurology 1991;41:216–221. in preterm and growth-retarded neonates. Acta Pediatr 1995;84:495–499.

Barkovich_Chap04.indd 359 5/25/2011 11:39:42 AM


360 Pediatric Neuroimaging

434. Kreis R, Ernst T, Ross BD. Absolute quantitation of water and metabolites in 460. Levi S. Ultrasonic assessment of the high rate of human multiple pregnancy
the human brain. II. Metabolite concentrations. J Magn Res 1993;102:9–15. in the first trimester. J Clin Ultrasound 1976;4:3–5.
435. Kreis R, Ernst T, Ross BD. Development of the human brain: in vivo quan- 461. Pharoah P, Adi Y. Consequences of in-utero death in a twin pregnancy.
tification of metabolite and water content with proton magnetic reso- Lancet 2000;355:1597–1602.
nance spectroscopy. Mag Res Med 1993;30:424–437. 462. Larroche J-C, Droulle P, Delezoide AL, et al. Brain damage in monozygous
436. Pu Y, Li Q-F, Zeng C-M, et al. Increased detectability of alpha brain gluta- twins. Biol Neonate 1990;57:261–278.
mate/glutamine in neonatal hypoxic-ischemic encephalpathy. Am J Neu- 463. Scheller JM, Nelson KB. Twinning and neurologic morbidity. Am J Dis
roradiol 2000;21:203–212. Child 1992;146:1110–1113.
437. Groenendaal F, Roelants-Van Rijn AM, van Der Grond J, Toet MC, de 464. Behar R, Wozniak P, Allard M, et al. Antenatal origin of neurologic
Vries LS. Glutamate in cerebral tissue of asphyxiated neonates during the damage in newborn infants. 1. Preterm infants. Am J Obstet Gynecol
first week of life demonstrated in vivo using proton magnetic resonance 1988;159:357–363.
spectroscopy. Biol Neonate 2001;79:254–257. 465. Grafe MR. Antenatal cerebral necrosis in monochorionic twins. Pediatr
438. Zhu W, Zhong W, Qi J, Yin P, Wang C, Chang L. Proton magnetic reso- Pathol 1993;13:15–19.
nance spectroscopy in neonates with hypoxic-ischemic injury and its 466. Williams K, Hennessy E, Alberman E. Cerebral palsy: effects of twin-
prognostic value. Translational Res 2008;152:225–232. ning, birthrate, and gestational age. Arch Dis Child Fetal Neonatal Ed
439. Roland EH, Flodmark O, Hill A. Thalamic hemorrhage with intraventric- 1996;75:F178–F182.
ular hemorrhage in the full-term newborn. Pediatrics 1990;85:737–742. 467. Wadhawan R, Oh W, Perritt RL, et al. Twin gestation and neurodevel-
440. Mitchell W, O’Tuama L. Cerebral intraventricular hemorrhages in infants: opmental outcome in extremely low birth weight infants. Pediatrics
a widening age spectrum. Pediatrics 1980;65:35–39. 2009;123:220–227.
441. Blanchette VS. Neonatal alloimmune thrombocytopenia. In: Stockman 468. Luu TM, Vohr B. Twinning on the brain: the effect on neurodevelopmental
JA, ed. Developmental and Neonatal Hematology. New York: Raven, 1988: outcomes. Am J Med Genet C Semin Med Genet 2009;151C:142–147.
145–168. 469. Benirschke K. The contribution of placental anastomoses to prenatal twin
442. Zalneraitis EL, Young RS, Krisnamoorthy KS. Intracranial hemorrhage damage. Hum Pathol 1992;23:1319–1320.
in utero as a complication of isoimmune thrombocytopenias. J Pediatr 470. Harkness UF, Crombleholme TM. Twin-twin transfusion syndrome:
1979;95:611–614. where do we go from here? Semin Perinatol 2005;29:296–304.
443. Serrarens-Janssen V, Semmekrot B, Novotny V, et al. Fetal/neonatal allo- 471. Fisk NM, Duncombe GJ, Sullivan MH. The basic and clinical science of
immune thrombocytopenia (FNAIT): past, present, and future. Obstet twin-twin transfusion syndrome. Placenta 2009;30:379–390.
Gynedol Surv 2008;63:239–252. 472. Norman MG, McGillivray BC, Kalousek DK, Hill A, Poskitt KJ. Congenital
444. Shapiro S, Campbell R, Scully T. Hemorrhagic dilation of the fourth ven- Malformations of the Brain: Pathologic, Embryologic, Clinical, Radiologic
tricle: an ominous predictor. J Neurosurg 1994;80:805–809. and Genetic Aspects. Oxford: Oxford University Press, 1995.
445. Huang A, L. RR. Spontaneous superficial parenchymal and leptomenin- 473. Volpe JJ. Hypoglycemia and brain injury. In: Volpe JJ, ed. Neurology of the
geal hemorrhage in term neonates. Am J Neuroradiol 2004;25:469–475. Newborn. Philadelphia, PA: Elsevier, 2008:591–618.
446. Hanigan W, Powell F, Palagallo G, Miller T. Lobar hemorrhages in full 474. Sexson WR. Incidence of neonatal hypoglycemia: a matter of definition.
term neonates. Child’s Nerv Syst 1995;11:276–280. J Pediatr 1984;105:149–153.
447. Dale S, Coleman L. Neonatal alloimmune thrombocytopenia: antenatal 475. Cornblath M, Schwartz P. Disorders of Carbohydrate Metabolism in Infancy,
and postnatal imaging findings in the pediatric brain. Am J Neuroradiol 3rd ed. Boston: Blackwell Scientific, 1991.
2002;23:1457–1465. 476. Montassir H, Maegaki Y, Ogura K, et al. Associated factors in neonatal
448. Bussel J. Immune thrombocytopenia in pregnancy: autoimmune and hypoglycemic brain injury. Brain Dev 2009;31:649–656.
alloimmune. J Reprod Immunol 1997;37:35–61. 477. Vannucci RC, Randis EE, Vannucci SJ. Cerebral metabolism during hypo-
449. Bussel J, Zubusky M, Berkowitz R, McFarland J. Fetal alloimmune throm- glycemia and asphyxia in newborn dogs. Biol Neonate 1980;38:276–283.
bocytopenia. N Engl J Med 1997;337:22–26. 478. Hernandez M, Vannucci R, Salcedo A, Brennan R. Cerebral blood flow
450. Millet V, Bartoli J, Lacroze V, Raybaud C, Unal D, Girard N. Predictive and metabolism during hypoglycemia in modern dogs. J Neurochem
significance of magnetic resonance imaging at 4 months of adjusted age in 1980;35:622–626.
infants after a perinatal neurologic insult. Biol Neonate 1998;73:207–219. 479. Vannucci RC, Nardis EE, Vannucci SJ, Campbell PA. Cerebral carbohy-
451. Dubowitz DJ, Bluml S, Arcinue E, Dietrich RB. MR of hypoxic enceph- drate and energy metabolism during hypoglycemia in newborn dogs.
alopathy in children after near drowning: correlation with quantita- Am J Physiol 1981;240:192–198.
tive proton MR spectroscopy and clinical outcome. Am J Neuroradiol 480. Anderson JM, Milner RDG, Strich SJ. Effects of neonatal hypoglycaemia
1998;19:1617–1627. on the nervous system: a pathological study. J Neurol Neurosurg Psychiat
452. Andronikou S, Van Toorn R. The DWI “reversal sign” of white matter 1967;30:295–310.
hypoxic ischaemic injury in older children: an unusual MRI pattern for 481. Anderson J, Milner R, Strich S. Pathological changes in the nervous system
age. Pediatr Radiol 2009;39:293–298. in severe neonatal hypoglycaemia. Lancet 1966;2:372–375.
453. Christophe C, Fonteyne C, Ziereisen F, et al. Value of MR imaging of the 482. Banker BQ. The neuropathological effects of anoxia and hypoglycemia in
brain in children with hypoxic coma. Am J Neuroradiol 2002;23(4):716–723. the newborn. Dev Med Child Neurol 1967;9:544–550.
454. Fitch SJ, Gerald B, Magill HL, Tonkin ILD. Central nervous system hypoxia 483. Spar JA, Lewine JD, Orrison WW Jr. Neonatal hypoglycemia: CT and MR
in children due to near drowning. Radiology 1985;156:647–650. findings. Am J Neuroradiol 1994;15:1477–1478.
455. Kjos BO, Brant-Zawadzki M, Young RG. Early CT findings of global central 484. Kim S, Goo H, Lim K, Kim S, Kim K. Neonatal hypoglycaemic encephal-
nervous system hypoperfusion. Am J Roentgenol 1983;141:1227–1232. opathy: diffusion-weighted imaging and proton MR spectroscopy. Pediatr
456. Han BK, Towbin RB, De Courten-Myers G, McLaurin RL, Ball WS, Jr. Radiol 2006;36(2):144.
Reversal sign on CT: effect of anoxic/ischemic cerebral injury in children. 485. Volpe JJ. Bilirubin and brain injury. In: Volpe JJ, ed. Neurology of the
Am J Neuroradiol 1989;10:1191–1198. Newborn, 5th ed. Philadelphia: Saunders-Elseier, 2008:619–651.
457. Bird CR, Drayer BP, Gilles FH. Pathophysiology of reverse edema in global 486. Maisels MJ. Jaundice. In: MacDonald MG, Mullett MD, Seshia MMK, eds.
cerebral ischemia. Am J Neuroradiol 1989;10:95–98. Neonatology: Pathophysiology and Management of the Newborn, 6th ed.
458. Kline-Fath BM, Calvo-Garcia MA, O’Hara SM, Crombleholme TM, Raca- Philadelphia: Lippincott Williams & Wilkins; 2005:768–846.
dio JM. Twin–twin transfusion syndrome: cerebral ischemia is not the 487. Bhutani VK, Johnson LH, Shapiro SM. Kernicterus in sick and preterm
only fetal MR imaging finding. Pediatr Radiol 2007;37(1):47–56. infants (1999–2002): a need for an effective preventive approach. Semin
459. Landy HJ, Weiner S, Carson S, et al. The “vanishing twin”: ultrasono- Perinatol 2004;28:319–325.
graphic assessment of fetal disappearance in the first trimester. Am J 488. Govaert P, Lequin M, Swarte R, et al. Changes in globus pallidus with (pre)
Obstet Gynecol 1986;155:14–19. term kernicterus. Pediatrics 2003;112:1256–1263.

Barkovich_Chap04.indd 360 5/25/2011 11:39:42 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 361

489. Gkoltsiou K, Tzoufi M, Counsell S, Rutherford M, Cowan F. Serial brain 514. Gentry LR, Godersky JC, Thompson BH. Traumatic brain stem injury:
MRI and ultrasound findings: relation to gestational age, bilirubin level, MR imaging. Radiology 1989;171:177–187.
neonatal neurologic status and neurodevelopmental outcome in infants at 515. Gentry LR, Godersky JC, Thompson B, et al. Prospective comparative
risk of kernicterus. Early Hum Dev 2008;84:829–838. study of intermediate-field MR and CT in the evaluation of closed head
490. Penn A, Enzmann D, Hahn J, Stevenson D. Kernicterus in a full term trauma. Am J Neuroradiol 1988;9:101–110.
infant. Pediatrics 1994;93:1003–1006. 516. Snow RB, Zimmerman RD, Gandy SE, Deck MDF. Comparison of MRI
491. Coskun A, Yikilmaz A, Kumandas S, Karahan OI, Akcakus M, Manav A. and CT in the evaluation of head injury. Neurosurgery 1986;18:45–52.
Hyperintense globus pallidus on T1-weighted MR imaging in acute ker- 517. Sullivan CR, Bruwer AJ, Harris LE. Hypermobility fo the cervical spine
nicterus: is it common or rare? Eur Radiol 2005;15:1263–1267. in children: a pitfall in the diagnosis of cervical dislocation. Am J Surg
492. Oakden WK, Moore AM, Blaser S, Noseworthy MD. H-1 MR spectro- 1958;95:636–640.
scopic characteristics of kernicterus: a possible metabolic signature. Am 518. Rossitch E, Jr, Oakes WJ. Perinatal spinal cord injury: clinical radiographic
J Neuroradiol 2005;26:1571–1574. and pathologic features. Pediatr Neurosurg 1992;18:149–152.
493. Wang X, Wu W, Hou BL, et al. Studying neonatal bilirubin encephalopathy 519. Pang D, Wilberger JE Jr. Spinal cord injury without radiographic abnor-
with conventional MRI, MRS, and DWI. Neuroradiology 2008;50:885–893. malities in children. J Neurosurg 1982;57:114–129.
494. Mahle W, Tavani F, Zimmerman R, et al. An MRI study of neuro- 520. Osenbach RK, Menezes AH. Spinal cord injury without radiographic
logical injury before and after congenital heart surgery. Circulation abnormality in children. Pediatr Neurosci 1989;15:168–174.
2002;106:I109–I114. 521. McGrory BJ, Klassen RA, Chao EYS, Staeheli JW, Weaver AL. Acute frac-
495. Limperopoulos C, Majnemer A, Rosenblatt B, Shevell M, Rohlicek C, tures and dislocations of the cervical spine in children and adolescents.
Tchervenkov C. Neurologic status of newborns with congenital heart J Bone Joint Surg [Am] 1993;75:988–995.
defects before open heart surgery. Pediatrics 1999;103:402–408. 522. Leventhal HR. Birth injuries of the spinal cord. J Pediatr 1960;56:447–453.
496. Forbess J, Visconti K, Bellinger D, Jonas R. Neurodevelopmental outcomes 523. Byers RK. Spinal cord injuries during birth. Dev Med Child Neurol
in children after the fontan operation. Circulation 2001;104:1127–1132. 1975;17:103–110.
497. Wright J, Hicks R, Newman D. Deep hypothermic arrest: observations on 524. Dickman CA, Rekate HL, Sonntag VKH, Zabramski JM. Pediatric spinal
later development in children. J Thorac Cardiovasc Surg 1979;77:466–468. trauma: vertebral column and spinal cord injuries in children. Pediatr
498. Miller SP, McQuillen PS, Hamrick S, et al. Abnormal Brain Devel- Neurosci 1989;15:237–256.
opment in Newborns with Congenital Heart Disease. N Engl J Med 525. Koch BM, Eng GM. Neonatal spinal cord injury. Arch Phys Med Rehabil
2007;357(19):1928–1938. 1979;60:378–381.
499. van Houten J, Rothman A, Bejar R. High incidence of cranial ultrasound 526. Ford FR. Breech delivery in its possible relations to injury of the spinal
abnormalities in full-term infants with congenital heart disease. Am J Peri- cord, with special reference to infantile paraplegia. Arch Neurol Psychiatry
natol 1996;13:47–53. 1925;14:742–750.
500. Finberg L. Current concepts: hypernatremic (hypertonic) dehydration in 527. Norman MG, Wedderburn LC. Fetal spinal cord injury with cephalic
infants. N Eng J Med 1973;289:196–198. delivery. Obstet Gynecol 1973;42:355–357.
501. Macaulay D, Watson M. Hypernatraemia in infants as a cause of brain 528. MacKinnon JA, Perlman M, Kirpalani H, et al. Spinal cord injury at
damage. Arch Dis Child 1967;42:485–491. birth: diagnostic and prognostic data in twenty-two patients. J Pediatr
502. Finberg L, Luttrell C, Redd H. Pathogenesis of lesions in the nervous sys- 1993;122:431–437.
tem in hypernatremic states. II. Experimental studies of gross anatomic 529. Towbin A. Spinal cord and brain stem injury at birth. Arch Pathol
changes and alterations of chemical composition of the tissues. Pediatrics 1964;77:620–632.
1959;23:46–53. 530. Edwards MSB, Cogen PH. Craniospinal trauma in children. In: Berg BO,
503. Karadag A, Uras N, Odemis E, Tunc B, Tatli MM. Superior sagittal sinus ed. Neurological Aspects of Pediatrics. Boston: Butterworth-Heinemann,
thrombosis: a rare but serious complication of hypernatremic dehydra- 1992:595–613.
tion in newborns. J Pernat Med 2007;35:82–83. 531. Castillo M, Quencer RM, Green BA. Cervical spinal cord injury after trau-
504. Righini A, Ramenghi L, Zirpoli S, Mosca F, Triulzi F. Brain Apparent Dif- matic breech delivery. Am J Neuroradiol 1989;10:S99.
fusion Coefficient Decrease During Correction of Severe Hypernatremic 532. Felsberg GJ, Tien RD, Osumi AK, Cardenas CA. Utility of MR imaging in
Dehydration. Am J Neuroradiol August 1, 2005 2005;26(7):1690–1694. pediatric spinal cord injury. Pediatr Radiol 1995;25:131–135.
505. Han BH, Lee M, Yoon HK. Cranial ultrasound and CT findings in infants 533. Fotter R, Sorantin E, Schneider U, Ranner G, Fast C, Schober P. Ultrasound
with hypernatremic dehydration. Pediatr Radiol 1997;27:739–742. diagnosis of birth-related spinal cord trauma: neonatal diagnosis and fol-
506. Noguchi K, Ogawa T, Seto H, et al. Subacute and chronic subarachnoid low-up and correlation with MRI. Pediatr Radiol 1994;24:241–244.
hemorrhage: diagnosis with fluid-attenuated inversion recovery MR 534. Filippigh P, Clapuyt P, Debauche C, Claus D. Sonographic evaluation
imaging. Radiology 1997;203:257–262. of truamatic spinal cord lesions in the newborn infant. Pediatr Radiol
507. Noguchi K, Ogawa T, Inugami A, et al. Acute subarachnoid hemorrhage: 1994;24:245–247.
MR imaging with fluid-attenuated inversion recovery pulse sequences. 535. Kulkarni MV, McArdle CB, Kopanicky D, et al. Acute spinal cord injury:
Radiology 1995;196:773–777. MR imaging at 1.5T. Radiology 1987;164:837–843.
508. Tavani F, Zimmerman R, Clancy R, Licht D, Mahle W. Incidental intracra- 536. Silberstein M, Tress BM, Hennessy O. Prediction of neurologic outcome
nial hemorrhage after uncomplicated birth: MRI before and after neonatal in acute spinal cord injury: the role of CT and MR. Am J Neuroradiol
heart surgery. Neuroradiology 2003;45:253–258. 1992;13:1597–1608.
509. Rooks VJ, Eaton JP, Ruess L, Petermann GW, Keck-Wherley J, Pedersen 537. Schaefer DM, Flanders AE, Osterholm JL, Northrup BE. Prognostic
RC. Prevalence and Evolution of Intracranial Hemorrhage in Asymptom- significance of magnetic resonance imaging in the acute phase of cervical
atic Term Infants. Am J Neuroradiol 2008;29:1082–1089. spine injury. J Neurosurg 1992;76:218–223.
510. Kelly AB, Zimmerman RD, Snow RB, Gandhi SE, Heier LA, Deck MDF. 538. Flanders AE, Spettell CM, Tartaglino LM, Friedman DP, Hervison GJ.
Head trauma: Comparison of MR and CT—experience in 100 patients. Forecasting motor recovery after cervical spinal cord injury: value of MR
Am J Neuroradiol 1988;9:699–708. imaging. Radiology 1996;201:649–655.
511. Han JS, Kaufman B, Alfidi RJ, et al. Head trauma evaluated by mag- 539. Miyanji F, Furlan JC, Aarabi B, Arnold PM, Fehlings MG. Acute cervi-
netic resonance and computed tomography: a comparison. Radiology cal traumatic spinal cord injury: mr imaging findings correlated with
1984;150:71–77. neurologic outcome—prospective study with 100 consecutive patients.
512. Gean A. Imaging of Head Trauma. New York, NY: Raven, 1994. Radiology 2007;243(3):820–827.
513. Gentry LR, Godersky JC, Thompson B. MR imaging of head trauma: 540. Smith AB, Gupta N, Strober J, Chin C. Magnetic resonance neurogra-
Review of the distribution and radiopathologic features of traumatic phy in children with birth-related brachial plexus injury. Pediatr Radiol
lesions. Am J Neuroradiol 1988;9:101–110. 2008;38:159–163.

Barkovich_Chap04.indd 361 5/25/2011 11:39:42 AM


362 Pediatric Neuroimaging

541. Volpe JJ. Injuries of extracranial, cranial, intracranial, spinal cord, periph- 568. Looney CB, Smith JK, Merck LH, et al. Intracranial hemorrhage in asymp-
eral nervous system structures. In: Volpe JJ, ed. Neurology of the Newborn, tomatic neonates: prevalence on mr images and relationship to obstetric
3rd ed. Philadelphia, PA: Saunders; 1995:769–792. and neonatal risk factors. Radiology 2006;242(2):535–541.
542. Gordon M, Rich H, Deutschberger J, Green M. The immediate and long 569. Perrin RG, Rutka JT, Drake JM, et al. Management and outcomes of poste-
term outcome of obstetric birth trauma. I. Brachial plexus paralysis. Am J rior fossa subdural hematomas in neonates. Neurosurgery 1997;40:1190–
Obstet Gynecol 1973;117:51–61. 1200.
543. Eng GD. Brachial plexus palsy in newborn infants. Pediatrics 1971;48: 570. Huang C-C, Shen E-Y. Tentorial subdural hemorrhage in term new-
18–26. borns: ultrasonographic diagnosis and clinical correlates. Pediatr Neurol
544. Moczygemba CK, Paramsothy P, Meikle S, et al. Route of delivery and neo- 1991;7:171–177.
natal birth trauma. Am J Obstet Gynecol 2010;202:361.e1–6. 571. Koch TK, Jahnke S, Edwards MSB, Davis RL. Posterior fossa hemorrhage
545. Laurent JP, Lee RT, Shenaq S, et al. Neurosurgical correction of upper bra- in term newborns. Pediatr Neurol 1985;1:96–102.
chial plexus birth injuries. J Neurosurg 1993;79:197–203. 572. Currarino G. Occipital osteodiastasis: presentation of four cases and
546. Hoeksma AF, ter Steeg AM, Nelissen RG, van Ouwerkerk WJ, Lankhorst review of the literature. Pediatr Radiol 2000;30:823–829.
GJ, de Jong BA. Neurological recovery in obstetric brachial plexus injuries: 573. Hernansanz J, Munoz F, Rodriquez M, et al. Subdural hematomas of the
an historical cohort study. Dev Med Child Neurol 2004;46(2):76–83. posterior fossa in normal-weight newborns. J Neurosurg 1984;61:972–977.
547. Pondaag W, Malessy MJ, van Dijk JG, Thomeer RT. Natural history of 574. Lau LSW, Pike JW. The CT findings of peritentorial subdural hemorrhage.
obstetric brachial plexus palsy: a systematic review. Dev Med Child Neurol Radiology 1983;146:699–701.
2004;46(2):138–144. 575. Barkovich AJ, Atlas SW. MRI of intracranial hemorrhage. Radiol Clin
548. Filler AG, Howe FA, Hayes CE, et al. Magnetic resonance neurography. North America 1988;26:801–820.
Lancet 1993;341:659–661. 576. Gomori JM, Grossman RI, Goldberg HI, Zimmerman RA, Bilaniuk
549. Howe FA, Filler AG, Bell BA, et al. Magnetic resonance neurography. Magn LP. Intracranial hematomas: Imaging by high field MR. Radiology
Reson Med 1992;28:328–338. 1985;157:87–93.
550. Medina LS, Yaylali I, Zurakowski D, Ruiz J, Altman NR, Grossman JA. 577. Hayman LA, Taber KH, Ford JJ, Bryan RN. Mechanisms of MR signal
Diagnostic performance of MRI and MR myelography in infants with a alteration by acute intracerebral blood: old concepts and new theories.
brachial plexus birth injury. Pediatr Radiol 2006;36(12):1295–1299. Am J Neuroradiol 1991;12:899–907.
551. Volpe JJ. Neurology of the Newborn, 4th ed. Philadelphia: Saunders, 2001. 578. Silvera S, Oppenheim C, Touze E, et al. Spontaneous intracerebral hema-
552. Caughey AB, Sandberg PL, Zlatnik MG, Thiet MP, Parer JT, Laros RK Jr. toma on diffusion-weighted images: influence of T2-shine-through and
Forceps compared with vacuum: rates of neonatal and maternal morbid- T2-blackout effects. Am J Neuroradiol Feb 2005;26(2):236–241.
ity. Obstet Gynecol 2005;106(5 Pt 1):908–912. 579. Fobbin J, Grossman R, Atlas S. MR characterization of subdural hemato-
553. Thornbury JR, Campbell JA, Masters SJ, Fryback DG. Skull fracture and mas and hygromas at 1.5T. Am J Neuroradiol 1989;10:687–693.
the low risk of intracranial sequelae in minor head trauma. Am J Roent- 580. Schreiber MS. Some observations on certain head injuries in infants and
genol 1984;143:661–664. children. Med J Australia 1957;2:930–936.
554. Masters SJ, McClean PM, Arcarese JS, et al. Skull x-ray examination after 581. Rivera FP, Kamitsuka MD, Quan L. Injuries to children younger than one
head trauma: recommendations by a multidisciplinary panel and valida- year of age. Pediatrics 1988;81:93–97.
tion study. N Eng J Med 1987;316:84–91. 582. Matschke J, Voss J, Obi N, et al. Nonaccidental head injury is the most
555. Lindenberg R. Pathology of craniocerebral injuries. In: Newton TH, Potts common cause of subdural bleeding in infants <1 year of age. Pediatrics
DG, eds. Anatomy and Pathology, vol 3. St. Louis: Mosby, 1977:3049– 2009;124(6):1587–1594.
3087. 583. Garton HJ, Hammer MR. Detection of Pediatric Cervical Spine Injury.
556. Kuppermann N, Holmes JF, Dayan PS, et al. Identification of children at Neurosurgery 2008. 62:700–708.
very low risk of clinically-important brain injuries after head trauma: a 584. Tong K, Ashwal S, Holshouser B, et al. Hemorrhagic shearing lesions
prospective cohort study. Lancet Oct 3 2009;374(9696):1160–1170. in children and adolescents with posttraumatic diffuse axonal injury:
557. Chuang SH, Fitz CR. CT of head trauma. In: Gonzales CF, Grossman improved detection and initial results. Radiology 2003;227:332–339.
CB, Masdeu JC, eds. CT of the Head and Spine New York: Wiley, 1985: 585. Hadley MN, Zabramski JM, Browner CM, Rekate H, Sonntag VKH. Pedi-
523–536. atric spinal trauma. Review of 122 cases of spinal cord and vertebral col-
558. Harwood-Nash DC, Hendrick EB, Hudson AR. The significance of umn injuries. J Neurosurg 1988;68:18–24.
skull fracture in children. A study of 1187 patients. Radiology 1971;101: 586. Wilberger JE Jr. Spinal Cord Injuries in Children. Mt. Kisco: Futura; 1986.
151–160. 587. Anderson JM, Schutt AH. Spinal injury in children. A review of 156 cases
559. Hughes CA, Harley EH, Milmoe G, Bala R, Martorella A. Birth trauma in the seen from 1950 through 1978. Mayo Clin Proc 1980;55:499–504.
head and neck. Archiv Otolaryngol Head Neck Surg 1999;125(2):193–199. 588. McCall T, Fassett D, Brockmeyer D. Cervical spine trauma in children: a
560. Mastrapa TL, Fernandez LA, Alvarez MD, Storrs BB, Flores-Urueta A. review. Neurosurg Focus 2006;20(2):E5.
Depressed skull fracture in Ping Pong: elevation with Medeva extractor. 589. Turgut M, Akpinar G, Akala N, et al. Spinal injuries in the pediatric age
Childs Nerv Syst 2007;23(7):787–790. group: a review of 82 cases of spinal cord and vertebral column injuries.
561. Adams JH. Head injury. In: Adams JH, Corsellis JAN, Duchen LW, eds. Eur Spine J 1996;5:148–152.
Greenfield’s Neuropathology. New York: Wiley, 1984:85–124. 590. Hamilton MG, Myles ST. Pediatric spinal injury: review of 174 hospital
562. Eames FA, Waldman JB. CT of posttraumatic intradiploic meningocele of admissions. J Neurosurg 1992;77:700–704.
the skull base: a case report. Am J Neuroradiol 1991;12:985–987. 591. Roche C, Carty H. Spinal trauma in children. Pediatr Radiol 2001;31:677–700.
563. Numerow LM, Krcek JP, Wallace CJ, Tranmer BI, Auer RN, Fong TC. 592. Vitale MG, Goss JM, Matsumoto H, Roye DP Jr. Epidemiology of pediatric
Growing skull fracture simulating a rounded lytic calvarial lesion. Am J spinal cord injury in the United States: years 1997 and 2000. J Pediat
Neuroradiol 1991;12:783–784. Orthop 2006;26(6):745–749.
564. Husson B, Pariente D, Tammam S, Zerah M. The value of MRI in the early 593. Polk-Williams A, Carr BG, Blinman TA, Masiakos PT, Wiebe DJ, Nance
diagnosis of growing skull fracture. Pediatr Radiol 1996;26:744–747. ML. Cervical spine injury in young children: a National Trauma Data
565. Brouwer A, Groenendaal F, Koopman C, Nievelstein R-J, Han S, de Vries L. Bank review. Journal of pediatric surgery 2008;43(9):1718–1721.
Intracranial hemorrhage in full-term newborns: a hospital-based cohort 594. Cattell HS, Filtzer DL. Pseudosubluxation and other normal variations in
study. Neuroradiology 2010;52(6):567–576. the cervical spine in children. A study of one hundred and sixty children.
566. Aicardi J. Diseases of the Nervous System in Childhood. 1992; MacKeith J Bone Joint Surg 1965;47A:1295–1309.
Press, London. 1408pp. 595. Selden NR, Quint DJ, Patel N, d’Arcy HS, Papadopoulos SM. Emergency
567. Choux M, Grisoli F, Peragut J. Extradural hematomas in children. magnetic resonance imaging of cervical spinal cord injuries: clinical cor-
104 cases. Childs Brain 1975;1:337–347. relation and prognosis. Neurosurgery 1999;44:785–793.

Barkovich_Chap04.indd 362 5/25/2011 11:39:42 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 363

596. Eleraky MA, Theodore N, Adams M, Rekate HL, Sonntag VK. Pediatric 620. Sivit CJ, Taylor GA, Newman KD, et al. Safety belt injuries in children
cervical spine injuries: report of 102 cases and review of the literature. with lap-belt ecchymosis: CT findings in 61 patients. Am J Roentgenol
J Neurosurg 2000;92(1 Suppl):12–17. 1991;157:111–114.
597. Wang MY, Hoh DJ, Leary SP, Griffith P, McComb JG. High rates of neu- 621. Taylor GA, Eggli KD. Lap-belt injuries of the lumbar spine in children: a
rological improvement following severe traumatic pediatric spinal cord pitfall in CT diagnosis. Am J Roentgen 1988;150:1355–1358.
injury. Spine 2004;29(13):1493–1497; discussion E1266. 622. Vander Have KL, Caird MS, Gross S, et al. Burst fractures of the tho-
598. Keiper MD, Zimmerman RA, Bilaniuk LT. MRI in the assessment of the racic and lumbar spine in children and adolescents. J Pediat Orthoped
supportive soft tissues of the cervical spine in acute trauma in children. 2009;29(7):713–719.
Neuroradiology 1998;40:359–363. 623. Kraus JF. Epidemiological aspects of acute spinal cord injury. A review of
599. Frank JB, Lim CK, Flynn JM, Dormans JP. The efficacy of mag- incidence, prevalence, causes, and outcome. In: Becker DP, Povlishock JT,
netic resonance imaging in pediatric cervical spine clearance. Spine eds. Central Nervous System Trauma Status Report. Bethesda, MD: NIH,
2002;27(11):1176–1179. 1985:313–322.
600. Pieretti-Vanmarcke R, Velmahos GC, Nance ML, et al. Clinical clearance 624. Mahan ST, Mooney DP, Karlin LI, Hresko MT. Multiple level injuries in
of the cervical spine in blunt trauma patients younger than 3 years: a pediatric spinal trauma. J Trauma 2009;67(3):537–542.
multi-center study of the american association for the surgery of trauma. 625. Banerian KG, Wang A-Y, Samberg LC, Kerr HH, Wesolowski DP. Asso-
The Journal of trauma 2009;67(3):543–549; discussion 549–550. ciation of vertebral end plate fracture with pediatric lumbar interverte-
601. Viccellio P, Simon H, Pressman BD, Shah MN, Mower WR, Hoffman JR. A bral disk herniation: value of CT and MR imaging. Radiology 1990;177:
prospective multicenter study of cervical spine injury in children. Pediat- 763–765.
rics 2001;108(2):E20. 626. Gennuso R, Humphries RP, Hoffman HJ, Hendrick EB, Drake JM. Lum-
602. Grabb BC, Frye TA, Hedlund GL, Vaid YN, Grabb PA, Royal SA. MRI bar intervertebral disc disease in the pediatric population. Pediatr Neurosci
diagnosis of suspected atlanto-occipital dissociation in childhood. Pediatr 1992;18:282–286.
Radiol 1999;29:275–281. 627. Flanders AE, Schaefer DM, Doan HT, Mishkin MM, Gonzalez CF,
603. Leypold BG, Flanders AE, Burns AS. The early evolution of spinal cord Northrup BE. Acute cervical spine trauma: correlation of MR imaging
lesions on MR imaging following traumatic spinal cord injury. Am J Neu- findings with degree of neurologic deficit. Radiology 1990;177:25–33.
roradiol 2008;29:1012–1016. 628. Hackney DB. Denominators of spinal cord injury. Radiology
604. Willis BK, Smith JL, Falkner LD, Vernon DD, Walker ML. Fatal air bag 1990;177:18–20.
mediated craniocervical trauma in a child. Pediatr Neurosurg 1996;24: 629. Ahmann PA, Smith SA, Schwartz JF, et al. Spinal cord infarction due to
323–327. minor trauma in children. Neurology 1975;25:301–307.
605. Quinones-Hinojosa A, Jun P, Manley GT, Knudson MM, Gupta N. Airbag 630. Blennow G, Starck L. Anterior spinal artery syndrome: report of seven
deployment and improperly restrained children: a lethal combination. cases in childhood. Pediatr Neurosci 1987;13:32–37.
J Trauma 2005;59(3):729–733. 631. Choi J-U, Hoffman HJ, Hendrick EB, Humphreys RP, Keith WS. Traumatic
606. Fielding JW. Cervical spine injuries in children. In: The Cervical Spine infarction of the spinal cord in children. J Neurosurg 1986;65:608–610.
Research Society, ed. The Cervical Spine. Philadelphia: Lippincott, 632. Hecht JT, Butler IJ. Neurologic morbidity associated with achondoplasia.
1983:268–281. J Child Neurol 1990;5:84–97.
607. Steinmetz MP, Lechner RM, Anderson JS. Atlantooccipital dislocation 633. Kao SCS, Waziri MH, Smith WL, Sato Y, Yuh WTC, Franken EA, Jr. MR
in children: presentation, diagnosis, and management. Neurosurg Focus imaging of the craniovertebral junction, cranium, and brain in children
2003;14(2):ecp1. with achondoplasia. Am J Roentgenol 1989;153:565–569.
608. Sherk H, Nicholson J, Chung S. Fractures of the odontoid process in young 634. MacDonald D. Sternomastoid tumor and muscular tortocollis. J Bone
children. J Bone Joint Surg [Am] 1978;60:921–924. Joint Surg 1969;51B:432–437.
609. Ruge JR, Sinson GP, McLone DG, Cerullo LJ. Pediatric spinal injury: the 635. Tom LW, Rossiter JL, Sutton LN, Davidson RS. Torticollis in Children.
very young. J Neurosurg 1988;68:25–30. Otolaryngol Head Neck Surg 1991;105:1–9.
610. Lustrin E, Karakas S, Ortiz A, et al. Pediatric cervical spine: normal anat- 636. Duchowny M. Nonepileptic paroxysmal disorders. In: Berg BO, ed. Prin-
omy, variants, and trauma. Radiographics 2003;23:539–560. ciples of Child Neurology. New York: McGraw-Hill, 1996:285–296.
611. Smith AB, Dillon WP, Lau BC, et al. Radiation dose reduction strategy 637. Rosman NP, Douglass LM, Sharif UM, Paolini J. The neurology of benign
for CT protocols: successful implementation in neuroradiology section. paroxysmal torticollis of infancy: report of 10 new cases and review of the
Radiology 2008;247(2):499–506. literature. J Child Neurol 2009;24(2):155–160.
612. Connolly B, Emery D, Armstrong D. The odontoid synchondrotic slip: an 638. Phillips WA, Hensinger RN. The management of rotatory atlanto-axial
injury unique to young children. Pediatr Radiol 1995;25:S129-S133. subluxation in children. J Bone Joint Surg Am 1989;71:664–668.
613. Andreoli C, Colaiacomo MC, Rojas Beccaglia M, Di Biasi C, Casciani E, 639. Mathern G, Batzdorf U. Grisel’s syndrome. Clin Orthop Rel Res
Gualdi G. MRI in the acute phase of spinal cord traumatic lesions: Rela- 1989;244:131–146.
tionship between MRI findings and neurological outcome. La Radiologia 640. Dvorak J, Hayek J, Zehnder R. CT-functional diagnostics of the rotatory
medica 2005;110(5–6):636–645. instability of the upper cervical spine. Spine 1987;12:726–731.
614. Shanmuganathan K, Gullapalli RP, Zhuo J, Mirvis SE. Diffusion tensor MR 641. Maheshwaran S, Sgouros S, Jeyapalan K, Chapman S, Chandy J, Flint G.
imaging in cervical spine trauma. Am J Neuroradiol 2008;29(4):655–659. Imaging of childhood torticollis due to atlanto-axial rotatory fixation.
615. Vargas MI, Delavelle J, Jlassi H, et al. Clinical applications of diffusion ten- Childs Nerv Syst 1995;11:667–671.
sor tractography of the spinal cord. Neuroradiology 2008;50(1):25–29. 642. Fielding JW, Hawkins RJ. Atlanto-axial rotatory fixation. J Bone Joint Surg
616. Junkins EP, Jr., Stotts A, Santiago R, Guenther E. The clinical presenta- Am 1977;59:37–44.
tion of pediatric thoracolumbar fractures: a prospective study. J Trauma 643. Bunnell WP. Back pain in children. Ortho Clin North America 1982;13:
2008;65(5):1066–1071. 587–604.
617. Benzel EC, Hart BL, Ball PA, Baldwin NG, Orrison WW, Espinosa MC. 644. Kurihara A, Kataoka O. Lumbar disc herniation in children and adoles-
Magnetic resonance imaging for the evaluation of patients with occult cents. Spine 1980;5:443–448.
cervicalspine injury. J Neurosurg 1996;85:824–829. 645. Webb JH, Svien HJ, Kennedy RL. Protruded lumbar intervertebral discs in
618. Johnson DL, Falci S. The diagnosis and treatment of pediatric lum- children. J Am Med Assn 1954;154:1153–1154.
bar spine injuries caused by rear seat lap belts. Neurosurgery 1990;26: 646. Cronqvist S, Mortensson W. Protrusion of calcified cervical discs into
434–441. the spinal canal in children. a report of two cases. Neuroradiology 1975;9:
619. Mulpuri K, Reilly CW, Perdios A, Tredwell SJ, Blair GK. The spectrum of 223–225.
abdominal injuries associated with chance fractures in pediatric patients. 647. Dias MS, Pang D. Juvenile intervertebral disc calcification: recognition,
Eur J Pediatr Surg 2007;17(5):322–327. management, and pathogenesis. Neurosurgery 1991;28:130–135.

Barkovich_Chap04.indd 363 5/25/2011 11:39:42 AM


364 Pediatric Neuroimaging

648. Newton TH. Cervical intervertebral disc calcification in children. J Bone 672. Mohanty A, Sastry Kolluri VR, Subbakrishna DK, Satish S, Chandra Mouli
Joint Surg [Am] 1958;40A:107–113. BA, Das BS. Prognosis of extradural hematomas in children. Pediatr Neu-
649. Beluffi G, Fiori P, Sileo C. Intervertebral disc calcifications in children. rosurg 1995;23:57–63.
La Radiologia medica 2009;114(2):331–341. 673. Dhellemmes P, Lejeune J-P, Christiaens J-L, Combelles G. Traumatic extra-
650. Erkintalo MO, Salminen JJ, Alanen AM, Paajanen HEK, Kormano MJ. dural hematomas in infancy and childhood: experience with 144 cases.
Development of degenerative changes in the lumbar intervertebral disk: J Neurosurg 1985;62:861–864.
results of a prospective MR imaging study in adolescents with and without 674. Humphries RP. Complications of pediatric head trauma. Pediatr Neuro-
low-back pain. Radiology 1995;196:529–533. surg 1991–92;17:274–278.
651. Kjaer P, Leboeuf-Yde C, Sorensen JS, Bendix T. An epidemiologic study of 675. Gerlach R, Dittrich S, Schneider W, Ackermann H, Seifert V, Kieslich
MRI and low back pain in 13-year-old children. Spine 2005;30(7):798–806. M. Traumatic epidural hematomas in children and adolescents: out-
652. Osmond MH, Klassen TP, Wells GA, et al. CATCH: a clinical decision rule come analysis in 39 consecutive unselected cases. Pediatr Emerg Care
for the use of computed tomography in children with minor head injury. 2009;25(3):164–169.
CMAJ 2010;182(4):341–348. 676. Luerssen TG, Klauber MR, Marshall LF. Outcome from head injury related
653. Geyer C, Ulrich A, Grafe G, Stach B, Till H. Diagnostic value of S100B and to the patients’s age. J Neurosurg 1988;68:409–416.
neuron-specific enolase in mild pediatric traumatic brain injury. J Neuro- 677. Merten DF, Osborne DRS, Radkowski MA, et al. Craniocerebral trauma
surg 2009;4(4):339–344. in the child abuse syndrome: radiological observations. Pediatr Radiol
654. Lo TY, Jones PA, Minns RA. Pediatric brain trauma outcome prediction 1984;14:272–277.
using paired serum levels of inflammatory mediators and brain-specific 678. Sato Y, Yuh WTC, Smith WL, Alexander RC, Kao SCS, Ellerbroek CJ.
proteins. J Neurotrauma 2009;26(9):1479–1487. Head injury in child abuse: evaluation with MR imaging. Radiology
655. Bechtel K, Frasure S, Marshall C, Dziura J, Simpson C. Relationship of 1989;173:653–660.
serum S100B levels and intracranial injury in children with closed head 679. Harwood-Nash DC. Abuse to the pediatric central nervous system. Am J
trauma. Pediatrics 2009;124(4):e697–e704. Neuroradiol 1992;13:569–575.
656. Bruce DA. Imaging after head trauma: why, when, and which. Child’s Nerv 680. Ball WS, Jr. Nonaccidental craniocerebral trauma (child abuse): MR imag-
Syst 2000;16:755–759. ing. Radiology 1989;173:609–610.
657. Woischneck D, Klein S, Reissberg S, et al. Prognosis of brain stem lesion in 681. Brown J, Minns R. Non-accidental head injury, with particular reference
children with head injury. Childs Nerv Syst 2003;19:174–178. to whiplash shaking injury and medicolegal aspects. Dev Med Child Neurol
658. Tong K, Ashwal S, Holshouser B, et al. Diffuse axonal injury in children: 1993;35:849–869.
clinical correlation with hemorrhagic lesions. Ann Neurol 2004;56:36–50. 682. Shugerman RP, Paez A, Grossman DC, Feldman KW, Grady MS. Epidural
659. Liu AY, Maldjian JA, Bagley LJ, Sinson GP, Grossman RI. Traumatic hemorrhage: is it abuse? Pediatrics 1996;97:664–668.
brain injury: diffusion weighted MR imaging findings. Am J Neuroradiol 683. Moyes PD. Subdural effusions in infancy. Can Med Assoc J 1969;100:
1999;20:1636–1641. 231–235.
660. Ashwal S, Holshouser BA, Tong K. Use of advanced neuroimaging tech- 684. Hellbusch LC. Benign extracerebral fluid collections in infancy: clinical pre-
niques in the evaluation of pediatric traumatic brain injury. Dev Neurosci sentation and long-term follow-up. J Neurosurg 2007;107(2 Suppl):119–125.
2006;28(4):309–326. 685. McNeely PD, Atkinson JD, Saigal G, O’Gorman AM, Farmer JP. Sub-
661. Niogi SN, Mukherjee P, Ghajar J, et al. Extent of microstructural white dural hematomas in infants with benign enlargement of the subarach-
matter injury in postconcussive syndrome correlates with impaired cogni- noid spaces are not pathognomonic for child abuse. Am J Neuroradiol
tive reaction time: a 3T diffusion tensor imaging study of mild traumatic 2006;27(8):1725–1728.
brain injury. Am J Neuroradiol 2008;29:967–973. 686. Papasian NC, Frim DM. A theoretical model of benign external hydro-
662. Kraus MF, Susmaras T, Caughlin BP, Walker CJ, Sweeney JA, Little DM. cephalus that predicts a predisposition towards extra-axial hemorrhage
White matter integrity and cognition in chronic traumatic brain injury: a after minor head trauma. Pediatric neurosurgery 2000;33(4):188–193.
diffusion tensor imaging study. Brain 2007;130(Pt 10):2508–2519. 687. Neglect CoCAa. Shaken baby syndrome: inflicted cerebral trauma. Pediat-
663. Wilde EA, McCauley SR, Hunter JV, et al. Diffusion tensor imaging of acute rics 1993;92:872–875.
mild traumatic brain injury in adolescents. Neurology 2008;70(12):948–955. 688. Kraus JF, Fife D, Conroy C. Pediatric brain injuries: the nature, clinical
664. Levin HS, Wilde EA, Chu Z, et al. Diffusion tensor imaging in relation to course, and early outcomes in a defined United States population. Pediat-
cognitive and functional outcome of traumatic brain injury in children. rics 1987;79:501–508.
J Head Trauma Rehabil 2008;23(4):197–208. 689. Bell WO. Pediatric Head Trauma. In: Arensman RM, ed. Pediatric Trauma.
665. Caeyenberghs K, Leemans A, Geurts M, et al. Brain-behavior relationships Philadelphia: Lippencott-Raven; 1995:101–119.
in young traumatic brain injury patients: DTI metrics are highly corre- 690. Duhaime A-C, Christian C, Armonda R, Hunter J, Hertle R. Disappearing
lated with postural control. Hum Brain Map 2010;31:992–1002. subdural hematomas in children. Pediatr Neurosurg 1996;25:116–122.
666. Babikian T, Asarnow R. Neurocognitive outcomes and recovery after 691. Zimmerman RA, Bilaniuk LT, Bruce D, et al. Computed tomography of
pediatric TBI: meta-analytic review of the literature. Neuropsychology cerebral injury in the abused child. Radiology 1979;130:687–690.
2009;23(3):283–296. 692. Domenicucci M, Strzelecki JW, Delfini R. Acute posttraumatic subdural
667. Babikian T, Tong KA, Galloway NR, Freier-Randall MC, Obenaus A, hematomas: “intradural” computed tomographic appearance as a favor-
Ashwal S. Diffusion-weighted imaging predicts cognition in pediatric able prognostic factor. Neurosurgery 1998;42:51–55.
brain injury. Pediatr Neurol 2009;41(6):406–412. 693. Grossman RI, Gomori JM, Goldberg HI, et al. MR imaging of hemorrhagic
668. Cohen BA, Inglese M, Rusinek H, Babb JS, Grossman RI, Gonen O. Proton conditions of the head and neck. Radiographics 1988;8(3):441–454.
MR Spectroscopy and MRI-Volumetry in Mild Traumatic Brain Injury. 694. Gomori JM, Grossman RI, Steiner I. High-field magnetic resonance imag-
Am J Neuroradiol 2007;28(5):907–913. ing of intracranial hematomas. Isr J Med Sci 1988;24(4–5):218–223.
669. Huang M, Theilmann RJ, Robb A, et al. Integrated imaging approach with 695. da Rocha AJ, da Silva CJ, Gama HP, et al. Comparison of magnetic reso-
MEG and DTI to Detect Mild Traumatic Brain Injury in Military and nance imaging sequences with computed tomography to detect low-grade
Civilian Patients. J Neurotrauma 2009;26:1213–1226. subarachnoid hemorrhage: role of fluid-attenuated inversion recovery
670. Lewine JD, Davis JT, Bigler ED, et al. Objective documentation of trau- sequence. J Comput Assist Tomogr 2006;30(2):295–303.
matic brain injury subsequent to mild head trauma: multimodal brain 696. Wu Z, Li S, Lei J, An D, Haacke EM. Evaluation of traumatic subarach-
imaging with MEG, SPECT, and MRI. J Head Trauma Rehabil 2007;22(3): noid hemorrhage using susceptibility-weighted imaging. Am J Neuroradiol
141–155. 2010;31:1302–1310.
671. Lewine JD, Davis JT, Sloan JH, Kodituwakku PW, Orrison WW Jr. Neuro- 697. Dolinskas C, Zimmerman RA, Bilaniuk LT. A sign of subarachnoid bleed-
magnetic assessment of pathophysiologic brain activity induced by minor ing on cranial computed tomography of pediatric head trauma patients.
head trauma. Am J Neuroradiol 1999;20(5):857–866. Radiology 1978; 126:409–411.

Barkovich_Chap04.indd 364 5/25/2011 11:39:42 AM


Chapter 4 • Brain and Spine Injuries in Infancy and Childhood 365

698. Aldrich EF, Eisenberg HM, Saydjari C, et al. Diffuse brain swelling in 720. Cecil KM, Lenkinski RE, Meaney DF, McIntosh TK, H. SD. High field
severely head-injured children. A report from the NIH Traumatic Coma proton magnetic resonance spectroscopy of a swine model for axonal
Data Bank. J Neurosurg 1992;76:450–454. injury. J Neurochem 1998;70:2038–2044.
699. Vavilala M, Muangman S, Tontisirin N, et al. Impaired cerebral auto- 721. Hunter JV, Thornton RJ, Wang ZJ, et al. Late proton MR spectroscopy in
regulation and 6-month outcome in children with severe traumatic brain children after traumatic brain injury: correlation with cognitive outcomes.
injury: preliminary findings. Dev Neurosci 2006;28:348–353. Am J Neuroradiol 2005;26:482–488.
700. Huisman TA, Schwamm LH, Schaefer PW, et al. Diffusion tensor imaging 722. Holshouser B, Ashwal S, Tong K. Proton MR spectroscopic imaging
as potential biomarker of white matter injury in diffuse axonal injury. Am depicts diffuse axonal injury in children with traumatic brain injury. Am J
J Neuroradiol Mar 2004;25(3):370–376. Neuroradiol 2005;26:1276–1285.
701. Schaefer PW, Huisman TAGM, Sorensen AG, Gonzalez RG, Schwamm 723. Reichenbach J, Venkatesan R, Yablonskiy D, Thompson M, Lai S, Haacke E.
LH. Diffusion weighted MR imaging in closed head injury: high correla- Theory and application of static field inhomogeneity effects in gradient-
tion with initial Glasgow coma scale score and score on modified Rankin echo imaging. J Magn Reson Imaging 1997;7:266–279.
scale at discharge. Radiology 2004;233:58–66. 724. Cederberg D, Siesjo P. What has inflammation to do with traumatic brain
702. Endo M, Ichikawa F, Miyasaka Y, Yada K, Ohwada T. Capsular and thal- injury? Childs Nerv Syst 2010;26(2):221–226.
amic infarction caused by tentorial herniation subsequent to head trauma. 725. Fitz CR, Harwood-Nash DC. CT of hydrocephalus. Comput Tomogr
Neuroradiology 1991;33:296–299. 1978;2:91–108.
703. Adams JH, Doyle D, Ford I, Gennarelli TA, Graham DI, McLellan DR. 726. Guertin SR. Cerebrospinal fluid shunts. Evaluation, complications, and
Diffuse axonal injury in head injury: definition, diagnosis and grading. crisis management. Pediatr Clin North America 1987;34:203–217.
Histopathology 1989;15(1):49–59. 727. Howman-Giles R, McLaughlin A, Johnston I, Whittle I. A radionuclide
704. Hesselink JR, Dowd CF, Healy ME, et al. MR imaging of brain contusions: method of evaluating shunt function and CSF circulation in hydrocepha-
A comparative study with CT. Am J Neuroradiol 1988;9:269–278. lus. Technical note. J Neurosurg 1984;61:604–605.
705. Yuan W, Holland SK, Schmithorst VJ, et al. Diffusion tensor MR imag- 728. Algin O, Hakyemez B, Gokalp G, Ozcan T, Korfali E, Parlak M. The
ing reveals persistent white matter alteration after traumatic brain injury contribution of 3D-CISS and contrast-enhanced MR cisternography in
experienced during early childhood. Am J Neuroradiol 2007;28(10): detecting cerebrospinal fluid leak in patients with rhinorrhoea. Br J Radiol
1919–1925. 2010;83(987):225–232.
706. Salmond CH, Menon DK, Chatfield DA, et al. Diffusion tensor imaging 729. Selcuk H, Albayram S, Ozer H, et al. Intrathecal gadolinium-enhanced
in chronic head injury survivors: correlations with learning and memory MR cisternography in the evaluation of CSF leakage. Am J Neuroradiol
indices. NeuroImage 2006;29(1):117–124. 2010;31(1):71–75.
707. Allkemper T, Tombach B, Schwindt W, et al. Acute and subacute intrac- 730. Sedlak AJ, Broadhurst DD. Executive Summary of the Third National Inci-
erebral hemorrhages: comparison of MR imaging at 1.5 and 3.0 T—initial dence Study of Child Abuse and Neglect. 1996; http://www.childwelfare.
experience. Radiology 2004;232:874–881. gov/pubs/statsinfo/nis3.cfm#national. Accessed July 3, 2008.
708. Levin H, Aldrich E, Saydjari C, et al. Severe head injury in chil- 731. U.S. Department of Health and Human Services AoC, Youth, and Fami-
dren: experience of the traumatic coma data bank. Neurosurgery lies. Child Maltreatment 2006. [Washington, DC: US. Government Printing
1992;31:435–444. Office] available at http://www.childwelfare.gov. 2008.
709. Aldrich EF, Eisenberg HM, Saydjari C, et al. Diffuse brain swelling in 732. Child Protection Division of the American Humane Association. National
severely head-injured children: a report from the NIH Traumatic Coma Analysis of Official Child Neglect and Abuse Reporting. Denver, CO: Ameri-
Data Bank. J Neurosurg 1992;76:450–454. can Humane Association, 1982.
710. Bruce DA, Alavi A, Bilaniuk L, et al. Diffuse cerebral swelling following 733. System NCAaND. Summary of child maltreatment 2002. 2004; http://
head injuries in children: the syndrome of “malignant brain edema”. www.acf.dhhs.gov/programs/cb/publications/cm02/summary.htm.
J Neurosurg 1981;54:170–178. 734. Billmire ME, Myers PA. Serious head injury in infants: accident or abuse?
711. Mendelsohn D, Levin HS, Burce D, et al. Late MRI after head injury in Pediatrics 1985;75:340–342.
children: relationship to clinical features and outcome. Child’s Nerv Syst 735. Duhaime A-C, Christian CW, Rorke LB, Zimmerman RA. Nonacciden-
1992;8:445–452. tal head injury in infants—the “shaken-baby syndrome”. N Eng J Med
712. Levin HS, Mendelsohn D, Lilly MA, et al. Magnetic resonance imaging in 1998;338:1822–1829.
relation to functional outcome of pediatric closed head injury: a test of the 736. Goldstein B, Kelly MM, Bruton D, Cox C. Inflicted vs. accidental head
Ommaya-Gennarelli model. Neurosurgery 1997;40:432–441. injury in critically injured children. Crit Care Med 1993;21:1328–1332.
713. Arfanakis K, Haughton V, Carew J, Rogers B, Dempsey R, Meyerand M. 737. Duhaime A, Alario A, Lewander W, et al. Head injury in very young chil-
Diffusion tensor MR imaging in diffuse axonal injury. Am J Neuroradiol dren: mechanisms, injury types, and ophthalmological findings in 100
2002;23(5):794–802. hospitalized patients younger than 2 years. Pediatrics 1992;90:179–185.
714. Sutton LN, Wang Z, Duhaime AC, Costarino D, Sauter R, Zimmerman 738. Perez-Arjona E, Dujovny M, DelProposto Z, et al. Late outcome fol-
RA. Tissue lactate in pediatric head trauma: a clinical study using H-1 lowing central nervous system injury in child abuse. Child Nerv Syst
NMR spectroscopy. Pediatr Neurosurg 1995;22:81–87. 2003;19:69–81.
715. Choe BY, Suh TS, Choi KH, Shinn KS, Park CK, Kang JK. Neuronal dys- 739. Adamo MA, Drazin D, Smith C, Waldman JB. Comparison of accidental and
function in patients with closed head injury evaluated by in vivo 1H mag- nonaccidental traumatic brain injuries in infants and toddlers: demographics,
netic resonance spectroscopy. Invest Radiol 1995;30:502–506. neurosurgical interventions, and outcomes. J Neurosurg 2009;4(5):414–419.
716. Stinson G, Bagley LJ, Cecil KM, et al. Magnetization transfer imaging 740. Kempe CH, Silverman FN, Steele BF, et al. The battered child syndrome.
and proton MR sepctroscopy in the evaluation of axonal injury: correla- JAMA 1962;181:17–24.
tion with clinical outcome after traumatic brain injury. Am J Neuroradiol 741. Caffey J. On the theory and practice of shaking infants: its potential resid-
2001;22:143–151. ual effects of permanent brain damage and mental retardation. Am J Dis
717. Schuhmann M, Stiller D, Skardelly M, et al. Metabolic changes in the Child 1972;124:161–169.
vicinity of brain contusions: a proton magnetic resonance spectroscopy 742. Caffey J. The whiplash shaken infant syndrome: manual shaking by the extremi-
and histology study. J Neurotrauma 2003;20:725–743. ties with whiplash induced intractranial hemorrhage linked with residual per-
718. Govindaraju V, Gauger GE, Manley GT, Ebel A, Meeker M, Maudsley AA. manent brain damage and mental retardation. Pediatrics 1974;54:396–403.
Volumetric proton spectroscopic imaging of mild traumatic brain injury. 743. Duhaime A-C, Gennarelli TA, Thibault LE, Bruce DA, Margulies SS, Wiser
Am J Neuroradiol 2004;25:730–737. R. The shaken baby syndrome: a clinical, pathological, and biomechanical
719. Rubin Y, Cecil KM, Wehrli S, McIntosh TK, Lenkinski RE, Smith DH. High study. J Neurosurg 1987;66:409–415.
resolution 1H NMR spectroscopy following experimental brain trauma. 744. Christian CW, Block R. Abusive head trauma in infants and children. Pedi-
J Neurotrauma 1997;14:441–449. atrics 2009;123(5):1409–1411.

Barkovich_Chap04.indd 365 5/25/2011 11:39:42 AM


366 Pediatric Neuroimaging

745. Gerber P, Coffman K. Nonaccidental head trauma in infants. Child’s Nerv Criteria for Imaging and Treatment Decisions. Reston: American College of
Syst 2007;23(5):499–507. Radiology, 2005.
746. Jaspan T. Current controversies in the interpretation of non-accidental 763. Chen CY, Huang CC, Zimmerman RA, et al. High-resolution cranial ultra-
head injury. Pediatr Radiol 2008;38(Suppl 3):S378–S387. sound in the shaken baby syndrome. Neuroradiology 2001;43:653–661.
747. Maxeiner H, Wolff M. Pure subdural hematomas: a postmortem analysis 764. Jaspan T, Griffiths P, McConachie N, Punt J. Neuroimaging for non-
of their form and bleeding points. Neurosurgery 2002;50:503–509. accidental head injury in childhood: a proposed protocol. Clin Radiol
748. Yamashima T, Friede R. Why do bridging veins rupture into the virtual 2003;58:44–53.
subdural space. J Neurol Neurosurg Psychiatr 1984;47:121–127. 765. Kemp AM, Rajaram S, Mann M, et al. What neuroimaging should be per-
749. Ghatan S, Ellenbogen R. Pediatric spine and spinal cord injury after formed in children in whom inflicted brain injury (iBI) is suspected? A
inflicted trauma. Neurosurg Clin North America 2002;13:227–233. systematic review. Clin Radiol 2009;64(5):473–483.
750. Oehmichen M, Meissner C, Saternus KS. Fall or shaken: traumatic brain 766. Parizel PM, Ceulemans B, Laridon A, Ozsarlak O, Jorens PG. Cortical
injury in children caused by falls or abuse at home—a review on biome- hypoxic-ischemic brain damage in shaken-baby (shaken impact) syn-
chanics and diagnosis. Neuropediatrics 2005;36:240–245. drome: value of diffusion-weighted MRI. Pediatr Radiol 2003;33:868–871.
751. Hadley M, Sonntag V, Rekate H, Murphy A. The infant whiplash- 767. Vinchon M, de Foort-Dhellemmes S, Desurmont M, Delestret I. Confessed
shake injury syndrome: a clinical and pathological study. Neurosurgery abuse versus witnessed accidents in infants: comparison of clinical, radio-
1989;24:536–540. logical, and ophthalmological data in corroborated cases. Childs Nerv Syst
752. Geddes JF, Hackshaw AK, Vowles GH, Nickols CD, Whitwell HL. Neuro- 2010;26:637–645.
pathology of inflicted head injury in children I. Patterns of brain damage. 768. Vezina G. Assessment of the nature and age of subdural collections in
Brain 2001;124:1290–1298. nonaccidental head injury with CT and MRI. Pediatr Radiol 2009;39(6):
753. Geddes JF, Vowles GH, Hackshaw AK, Nickols CD, Scott IS, Whitwell HL. 586–590.
Neuropathology of inflicted head injury in children II. Microscopic brain 769. Tung GA, Kumar M, Richardson RC, Jenny C, Brown WD. Comparison of
injury in infants. Brain 2001;124:1299–1306. accidental and nonaccidental traumatic head injury in children on non-
754. Wilkinson WS, Han D, Rappley MD, Owings C. Retinal hemorrhage pre- contrast computed tomography. Pediatrics 2006;118(2):626–633.
dicts neurologic injury in the shaken baby syndrome. Arch Ophthalmol 770. Zouros A, Bhargava R, Hoskinson M, Aronyk KE. Further characterization
1989;107:1472–1474. of traumatic subdural collections of infancy. Report of five cases. J Neuro-
755. Williams RA. Injuries in infants and small children resulting from wit- surg 2004;100(5 Suppl Pediatrics):512–518.
nessed and corroborated free falls. J Trauma 1991;31:1350–1352. 771. Menkes JH. Commentary: subdural hematoma, nonaccidental head injury
756. Kravitz H, Driessen G, Gomberg R, Korach A. Accidental falls from ele- or…? Eur J Ped Neurol 2001;5:175–176.
vated surfaces in infants from birth to one year of age. Pediatrics 1969;44: 772. Morris MW, Smith S, Cressman J, Ancheta J. Evaluation of Infants With
869–876. Subdural Hematoma Who Lack External Evidence of Abuse. Pediatrics
757. Keenan HT, Runyan DK, Marshall SW, Nocera MA, Merten DF, Sinal SH. 2000;105(3):549–553.
A population-based study of inflicted traumatic brain injury in young 773. Sun DT, Zhu XL, Poon WS. Non-accidental subdural haemorrhage in
children. J Am Med Assn 2003;290:621–626. Hong Kong: incidence, clinical features, management and outcome. Childs
758. Case ME, Graham MA, Handy TC, Jentzen JM, Monteleone JA. Position Nerv Syst 2006;22(6):593–598.
paper on fatal abusive head injuries in infants and young children. Am 774. Bird CR, McMahan JR, Gilles FH, Senac MO, Apthorp JS. Strangulation in
J Forensic Med Pathol 2001;22:112–122. child abuse: CT diagnosis. Radiology 1987;163:373–375.
759. Frank Y, Zimmerman RD, Leeds NM. Neurological manifestations 775. Blumenthal I. Shaken baby syndrome. Postgrad Med J 2002;78:732–735.
in abused children who have been shaken. Dev Med Child Neurol 776. Suh D, Davis P, Hopkins K, Fajman N, Mapstone T. Nonaccidental pedi-
1985;27:312–316. atric head injury: diffusion-weighted imaging findings. Neurosurgery
760. Ludwig S, Warman M. Shaken baby syndrome: a review of 20 cases. Ann 2001;49:309–318.
Emerg Med 1984;13:104–107. 777. Haseler LJ, Phil M, Arcinue E, Danielsen E, Bluml S, Ross BD. Evidence
761. Bonnier C, Nassogne M-C, Evrard P. Outcome and prognosis of whiplash from proton magnetic resonance spectroscopy for a metabolic cascade of
shaken infant syndrome: late consequences after a symptom-free interval. neuronal damage in shaken baby syndrome. Pediatrics 1997;99:4–14.
Dev Med Child Neurol 1995;37:943–956. 778. Aaen GS, Holshouser BA, Sheridan C, et al. Magnetic resonance spectros-
762. American College of Radiology Task Force on Appropriateness Criteria. copy predicts outcomes for children with nonaccidental trauma. Pediatrics
Imaging of the child with suspected physical abuse. In: ACR Appropriateness 2010;125:295–303.

Barkovich_Chap04.indd 366 5/25/2011 11:39:42 AM


CHAPTER

Congenital Malformations
5 of the Brain and Skull
A. JAMES BARKOVICH AND CHARLES A. RAYBAUD

Basic concepts of brain development Anomalies of the craniocervical junction (the Chiari
Concepts: causes, classification, and basic embryology malformations)
Early brain development Chiari I malformations
Anomalies of dorsal prosencephalon development Chiari II malformations
Anomalies of the cerebral commissures Chiari III malformations
Malformations of cerebral cortical development Anomalies of the mesenchyme (meninges and skull)
Anomalies of ventral prosencephalon development Cephaloceles and other calvarial and skull base defects
Holoprosencephaly Intracranial lipomas
Arrhinia/arrhinencephaly Arachnoid cysts
Septooptic dysplasia Craniosynostosis
Isolated absence of the septum pellucidum Chromosomal anomalies
Anomalies of the hypothalamic-pituitary axis Trisomy 21
Anomalies of the eyes Trisomy 18
Anomalies of midbrain-hindbrain development Trisomy 13
Overview of midbrain and hindbrain development Fragile X syndrome
Classification of midbrain-hindbrain malformations Inversion duplication of short arm of chromosome 8
Wolf-Hirschhorn syndrome

bnormal brain development, leading to dysplasia or BASIC CONCEPTS OF BRAIN DEVELOPMENT


A malformation, is a common finding in the neuroimaging stud-
ies of children with developmental delay, mental retardation, Concepts: Causes, Classification,
or epilepsy (1–3). This chapter discusses brain malformations, includ- and Basic Embryology
ing the essential embryology and genetics involved in the development
As will become evident in the sections that follow, most structures
of the malformation, the clinical findings, the optimal imaging tech-
of the brain form at about the same time. Therefore, events leading
niques to aid in diagnosis, and imaging findings. In addition, it discusses
to malformation of the developing brain often result in anomalies of
basic embryology of the brain, the disturbance of which results in mal-
more than one structure. For example, malformations of cerebral cor-
formations. This information should allow the reader to diagnose most
tex development may have associated cerebellar anomalies, cephaloce-
disorders of brain development.
les often have associated gray matter heterotopia or anomalies of the
The organization of this chapter has changed from that in the prior
corpus callosum, and holoprosencephalies frequently have associated
editions of this book. In order to help the reader locate the malforma-
anomalies of the corpus callosum and the cerebral cortex. The reader
tion, it is only necessary to figure out what part of the brain or skull is
should keep this high incidence of multiple anomalies in mind when
affected. If the malformation involves the cerebral hemispheres or the
reviewing imaging studies; as a wise radiologist once said, the hardest
commissures connecting them (corpus callosum, anterior commissure,
brain malformation to find is the second one.
hippocampal commissure), it will be found in “Anomalies of Dorsal
Classification of brain malformations is also difficult. One reason
Prosencephalon Development.” If it primarily involves the basal ganglia,
for this, pointed out by Norman et al. (4), is that no two brain mal-
hypothalamus-pituitary axis, the olfactory structures, or the orbits, it will
formations are exactly the same. Indeed, differences are seen even in
be found in “Anomalies of Ventral Prosencephalon Development.” If it
the malformations of siblings with identical chromosomal mutations,
mainly involves the brain stem or cerebellum, look in “Anomalies of Mid-
probably as a result of epigenetic (5) and environmental (6) factors. It
brain-Hindbrain development.” The Chiari malformations are discussed
is, nonetheless, important to try to put similar malformations into a
in “Anomalies of the Craniocervical Junction.” Anomalies of the calva-
single group whenever possible; without some sort of organization, it
rium, skull base, subarachnoid space (lipomas and cysts), and leptom-
would be extremely difficult to assemble information about prognosis,
eninges are discussed in “Anomalies of the Mesenchyme.” Hopefully, this
optimal therapy, and the chances that future siblings will be similarly
organization will facilitate navigating this chapter. Before discussing the
affected. Another difficulty in classifying brain anomalies results from
anomalies themselves, it is important to understand the basics of brain
the aforementioned frequency of multiple anomalies. Should a cepha-
development. This knowledge will help the reader to recognize malfor-
locele with heterotopic gray matter and callosal agenesis be classified as
mations and to distinguish them from distortion of normal brain.
367

Barkovich_Chap05.indd 367 5/6/2011 8:58:32 PM


368 Pediatric Neuroimaging

a disorder of neural tube closure, a malformation of cerebral cortical gestation. The caudal end of the neural tube, the posterior neuropore,
development, or a disorder of commissuration? Or are there enough closes at about 27 to 28 days of gestation.
patients with that combination of findings to define it as a malforma- At the time of closure of the anterior neuropore, three brain ves-
tion syndrome? These questions have no easy answers and, as a result, icles develop secondary to focal dilations in the rostral cavity of the
this chapter mainly describes anomalies of the various structures of the neural tube. These three early subdivisions are the prosencephalon
brain (i.e., corpus callosum, cerebral cortex, cerebellum) and its cover- (forebrain), the mesencephalon (midbrain), and the rhombencepha-
ings; we discuss the embryology of those structures but do not propose lon (hindbrain) (Fig. 5-1). The rhombencephalon is separated from
an overall classification of the disorders, as is done on anomalies of the the mesencephalon by the cephalic flexure and from the cervical spi-
spine in Chapter 9. A framework for classification based upon mor- nal cord by the cervical flexure. The prosencephalon divides into the
phology, genetics, and embryology is introduced for malformations caudal diencephalon, which forms the pretectum, thalami, substan-
of cortical development (7) and malformations of the midbrain and tial nigra-ventral tegmental area, and the prerubral tegmentum, and
hindbrain (8–10). When these disorders have malformations of mul- the more rostral protosegment, from which the hypothalamus, optic
tiple structures, all are discussed together whenever possible. vesicles, and telencephalon (which will form the cerebral hemispheres)
One final important concept concerns the causes of brain anoma- arise (21). The cells that compose the brain arise primarily within the
lies. The time during which specific structures of the brain are formed germinal zones in the walls of the ventricles (22–28). Many secondary
is usually known. An injury (of any type) to the brain at the time a par- germinal zones (e.g., the subventricular zone (SVZ) in the cerebrum,
ticular structure is forming will result in an anomaly of that structure; a the external granular layer [EGL] in the cerebellum) form when some
mutated gene can cause an identical anomaly if the protein product of daughter cells migrate a short way before stopping and duplicating
the gene functions in the formation of that same structure at the same again (27,29).
time. In addition, one protein may be involved in many pathways and, At the time of closure of the anterior neuropore, the optic vesicles
thus, be necessary for the formation of different structures at differ- have already begun to bud from the protosegment; these will interact
ent times; as a consequence, a single mutation may cause anomalies of with overlying ectoderm to form the optic nerves and ocular globes
several structures that form at different times. Finally, some of the same (which are really rostral extensions of the diencephalon). As the tel-
chemicals responsible for promoting cell growth or guiding cell/axon encephalon develops, the cerebral hemispheres grow posteriorly to
migration in utero have functions regulating brain metabolism after cover the diencephalon, mesencephalon, and portions of the rhomb-
birth; therefore, some patients with inborn errors of metabolism may encephalon (Fig. 5-1); accompanying this growth of the hemispheres,
have brain malformations. Although only a few such associations are the corpus callosum seems to grow from anterior to posterior (see sec-
described in the literature, more are likely to be discovered. One exam- tion “Anomalies of the Cerebral Commissures”). Further details of the
ple of this is the presence of callosal dysgenesis in patients with pyru- development of the telencephalon are discussed in sections “Anomalies
vate dehydrogenase complex deficiency (11). Other examples include of the Cerebral Commissures” and “Malformations of Cerebral Corti-
the presence of abnormal sulcation in the Zellweger syndrome (12,13) cal Development.”
and the presence of cerebellar hypoplasia in mitochondrial respiratory The rhombencephalon will eventually divide into the myelenceph-
chain dysfunction (14) and adenylosuccinate lyase deficiency (15). alon, which will form the pons and cerebellum (a dorsal extension of
the pons), and the metencephalon, which will form the medulla oblon-
gata. This process of development is discussed further in the section
Early Brain Development
“Anomalies of Midbrain–Hindbrain Development.”
On about the 15th day of life, ectodermal cells proliferate along the sur-
face of the embryo to form a plate of tissue, the primitive streak. A rap-
idly proliferating group of cells, known as Hensen node, forms at one
ANOMALIES OF DORSAL
end of the primitive streak and defines its cephalic end. From Hensen PROSENCEPHALON DEVELOPMENT
node, cells that will form the notochord migrate rostrally and allow dif-
Anomalies of the Cerebral Commissures
ferentiation of dorsal midline ectoderm into neuroectoderm. This plate-
like condensation of neuroectoderm is known as the neural plate. The cerebral commissures are bundles of axons that arise from corti-
At approximately seventeen days of gestation, the lateral portions cal neurons and connect the two cerebral hemispheres by traversing
of the neural plate begin to thicken bilaterally, forming the neural folds; the developing hemisphere and crossing the midline. Alterations of
the process of bending elevates these folds and brings them to the dor- their morphology, as assessed by imaging, may be identified in many
sal midline (16). At about 20 days, these folds meet in the midline at the hemispheric disorders, destructive or developmental, that impair cel-
level of the rhombencephalon. This junction of the neural folds begins lular migration, axon guidance, or crossing of the midline. As they can
the formation of the neural tube (17–19). be altered by so many processes, abnormalities of the cerebral com-
As the neural tube closes, the neuroectoderm, which will form the missures are among the most common developmental brain anoma-
central nervous system, separates from the overlying ectoderm, which lies, being found in 1.8 per 10,000 live births (30). The incidence is
will become the skin. Until recently, most experts believed that neural increased in prematurely born children and in children born to moth-
tube closure proceeds cranially and caudally in a zipper-like fashion. ers of advanced age, as well as in children with chromosomal disorders
More recent evidence, however, suggests that neurulation begins sepa- (17.3%), those with accompanying somatic disorders (musculoskeletal
rately at two, or possibly three, different levels in humans, when cellular 33.5% and cardiac 27.6%), and those with other CNS malformations
protrusions (possibly cilia) project medially from the most dorsal cells (49.5%) (30). As many as 3% to 4% of patients referred for cognitive
of the neural folds on either side. Cell recognition and adhesion occur or motor delay are found to have commissural anomalies, most often
under the influence of many molecules (20), closing the tube at each of the corpus callosum (31). Commissural anomalies were identified
point. This information may provide a unifying concept for certain in 33% of fetal brain malformations found on MRI by Raybaud et al.
developmental malformations, such as cephaloceles. The cephalic end (32); this figure may be low, as mild commissural anomalies may be
of the neural tube, the anterior neuropore, closes at about 25 days of missed by prenatal ultrasonography and, therefore, not referred for

Barkovich_Chap05.indd 368 5/6/2011 8:58:32 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 369

FIG. 5-1. Early development of the nervous system. A.


At the time of closure of the anterior neuropore, three
brain vesicles develop in the rostral cavity of the neural
tube. These three subdivisions are the prosencephalon,
mesencephalon, and rhombencephalon. The rhomben-
cephalon is separated from the mesencephalon by the
cephalic flexure and from the cervical spinal cord by
the cervical flexure. B. The pontine (rhombic) flexure,
which is the site of the future fourth ventricle and is
very important in the development of the cerebellum,
develops after the cervical and cephalic flexures. C. As
the telencephalon develops, the cerebral hemispheres
grow posteriorly to cover the midbrain and portions of
the hindbrain. The cerebellar hemispheres grow from
the dorsal, rostral portion of the hindbrain, which sit at
the cranial edge of the developing fourth ventricle (the
site of the pontine flexure).

Barkovich_Chap05.indd 369 5/6/2011 8:58:32 PM


370 Pediatric Neuroimaging

MRI (32). Although a few affected patients have normal cognition or connects the precentral and postcentral gyri as well as the primary
normal neurological examinations, the vast majority has neuromotor auditory cortices (Fig. 5-3). It contains large, heavily myelinated axons.
and cognitive impairment (31,33). Therefore, it is important to analyze Anterior to it, the body, genu, and lamina rostralis contain the small
the cerebral commissures in all patients referred for fetal or postnatal myelinated commissural axons of the frontal lobe association areas:
imaging. premotor cortex/supplementary motor area and prefrontal cortex.
Posterior to the isthmus, the splenium contain axons from the parietal,
Anatomy and Embryology medial occipital, and medial temporal cortices. The midline portion of
The cerebral commissures include the anterior commissure, which the hippocampal commissure is fused to the inferior, concave aspect
connects the olfactory cortex (paleocortex) and the lateral and inferior of the splenium, behind the body of the fornix (Fig. 5-2); it cannot be
temporo-occipital neocortex of the two hemispheres; the hippocam- distinguished from the callosal splenium on midline sagittal images.
pal commissure (psalterium), which connects the fornices and, thus, The anterior commissure is located in the upper portion of the lamina
the hippocampal cortex (archicortex); and the corpus callosum, which terminalis (the anterior wall of the third ventricle), very close to the
connects most of the hemispheric neocortex (principally neurons in junction of the lamina rostralis and the lamina terminalis (Fig. 5-2)
the intermediate cortical layers, Fig. 5-2). The corpus callosum, an evo- and the bifurcation of the fornix (where it divides into precommissural
lutionary acquisition of placental mammals (34), develops by second- [septal] and postcommissural [mamillary] tracts). The septum pellu-
ary fusion of the hemispheres in the midline at the site of the cavum cidum is bounded superiorly by the callosal body, anterior-inferiorly
septi pellucidi (35). Although the corpus callosum is the main pathway by the lamina rostralis, and posterior-inferiorly by the body of the
through which neocortical commissural fibers cross the midline, some fornix (Fig. 5-2) (38).
neocortical axons (from the lateral and inferior temporo-occipital In order to understand commissural anomalies, it is essential to
lobes) also cross via the anterior commissure (36). The septum pellu- understand the basics of commissural development. During the sev-
cidum is central to this process. It is surrounded by the three commis- enth week of gestation, the upper portion of the lamina terminalis (at
sures, being formed by the midline juxtaposition of the layer of white the rostral end of the neural tube) thickens to become the lamina reu-
matter that forms the medial border of each lateral ventricle, anterior niens or commissural plate (Fig. 5-4A); it corresponds to the future
to the hippocampal commissure. It contains myelinated, presumably septum or septal area (35), and it establishes continuity between the
septocingulate, axons but no gray matter (37,38). The septum pellu- two hemispheres. The pioneer axons of the anterior commissure cross
cidum is distinct from the septal area, also called the septum verum or the midline through cellular glial tunnels in its ventral portion dur-
septum granulosum, a gray matter structure that is connected to the ing the eighth week (Fig. 5-4B) (39). During weeks 9 to 10, the lamina
hypothalamus and located below the rostrum of the corpus callosum, reuniens develops a groove, the sulcus medianus telencephali medii
anterior to the lamina terminalis. The septum pellucidum is located (SMTM, Fig. 5-5A) in its dorsal surface. The lips of this groove con-
above the rostrum and forms the medial wall of the anterior parts of tain the early axons of the fornix that connect the septal area with the
the lateral ventricles. future hippocampus; some of these forniceal axons cross the midline
The corpus callosum is the largest and most easily visualized of in week 11, either within the dorsal lamina reuniens itself (35) or
the commissures. It is composed of five sections: the rostrum (lamina along its surface under the meningeal covering (40), to form the early
rostralis), genu, body, isthmus, and splenium (Fig. 5-2). The isthmus hippocampal commissure (Fig. 5-4C). Then the walls of the SMTM

FIG. 5-2. The normal cerebral commissures. Sagittal T1-weighted image (A) shows the anterior commissure (white asterisk) in the upper portion
of the anterior wall of the third ventricle (lamina terminalis). The hippocampal commissure (black asterisk) connects the columns of the fornices
and blends with the callosal splenium in the midline. The parts of the corpus callosum are the lamina rostralis or rostrum (small white arrow), genu
(large white arrow), body (large black arrow), isthmus (double white arrow), and splenium (triple white arrow). Coronal image (B) better shows the
separation between the callosal axons (cc) connecting the cerebral hemispheric white matter and the hippocampal commissure (hc) connecting the
fornices.

Barkovich_Chap05.indd 370 5/6/2011 8:58:35 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 371

FIG. 5-3. Callosal defect secondary to parenchymal injury (history of neonatal hypoxic-ischemic injury at term). A. Axial FLAIR
image demonstrates encephalomalacia in the depth of the central sulcus and underlying white matter bilaterally. B. Sagittal midline
T2 image demonstrates focal atrophy (white arrow) of the corpus callosum at the level of the isthmus (destruction of the sensorimo-
tor commissural axons). This example illustrates that the rostrum, genu, and body contain frontal commissural fibers, while the
splenium contains parietal, occipital, and temporal fibers.

LR
LT

A. 7 w
FIG. 5-4. Development of the lamina reuniens and of the corpus
LR callosum, sagittal midline. A. During week 7, the upper portion of
the lamina terminalis (LT), which connects the hemispheres across
AC
LT the midline, thickens and forms the lamina reuniens (LR) of His. B.
In the following week, olfactory commissural fibers cross the mid-
B. 8 w line through the ventral aspect of the lamina reuniens to form the
anterior commissure (AC). C. In the following weeks, fibers develop
B between the anterior mediobasal cortex (septal nuclei) and the
future hippocampus to form the ipsilateral fornix (FO); about week
11, some forniceal fibers cross the midline in the dorsal portion of
HC the lamina reuniens and form the hippocampal commissure (HC).
FO D. During week 12, the corticoseptal boundary becomes defined at
the medial edge of the future neocortex and a glial sling forms along
this boundary (see Fig. 5-5B). E. By week 13, three commissural sites
have been established: anterior commissure, hippocampal commis-
LR sure, and glial sling. Depending on their origins, early neocortical
AC commissural fibers cross the midline along the anterior commissure
C. 11 w
(temporo-occipital fibers), the glial sling (frontal fibers) or the hip-
pocampal commissure (parietooccipitotemporal fibers). F. The cor-
HC pus callosum grows by addition of further commissural fibers and
FO
forms a single continuous structure stretched between the anterior
commissure and the hippocampal commissure; it circumscribes the
glial sling future septum pellucidum. Later, the prominent development of the
frontal lobes results in posterior growth of the anterior callosum,
which displaces the hippocampal commissure and the splenium
AC D. 12 w
backward above the velum interpositum (roof above the third ven-
tricle), stretching the body of the fornix.

Barkovich_Chap05.indd 371 5/6/2011 8:58:35 PM


372 Pediatric Neuroimaging

C D

HC and posterior
callosum
HC
FO
AC FO

AC
glial sling and anterior
callosum

septum pellucidum
corpus callosum

septum pellucidum

FIG. 5-4. (Continued)

come into contact in the midline along a line (called the corticoseptal that compose the sling express surface molecules and secrete chemicals
boundary, Fig. 5-5A) that marks the limit between the developing cor- into the extracellular space that help to guide axons across the midline
tical plate at one end and the septal area and the fornix at the other (39,47,50–53). Within the developing hemispheres, two specialized
(41–44). Specialized cells that originate from the SVZ of the germinal glial structures secrete chemical signals that direct the commissural
matrix migrate toward this line, pierce the brain surface and invade axons toward the midline and the glial sling. One, located just above
the developing subarachnoid space (meninx primitiva) (Fig. 5-4) to the corticoseptal boundary is the indusium griseum glia, and the other
form an interhemispheric bridge across the midline, which is called the located just below it at the dorsomedial aspect of the lateral ventricle is
“glial sling” (41,45–47), or “midline glial zipper” (42,48,49). The cells called the glial wedge (Fig. 5-5B) (43,44,53).

FIG. 5-5. Midline glia, corpus callosum, and cavum septi pellucidi coronal
diagrams. A. A deep sulcus (sulcus medianus telencephali medii, SMTM)
develops in the superior aspect of the lamina reuniens. A clear demarcation
(the corticoseptal boundary) appears between the banks of this sulcus and
the neocortical plate. B. Specialized glial structures develop in the vicinity
of the corticoseptal boundary; the glial sling (or midline zipper glia) forms
a bridge between the hemispheres; on each side of the midline, the indu-
sium griseum glia and the glial wedge form above and below the corticoseptal
boundary. C. The pioneer commissural fibers are channeled from the cingu-
late gyrus toward the midline by the repelling effect of the indusium griseum
glia and the glial wedge, and guided across the midline by the glial sling.
At the same time, the ipsilateral fornix and septocingulate (septum pellu-
cidum) fibers develop in the banks of the SMTM, which become the leaves
of the septum pellucidum (not shown); the space within the SMTM becomes
enclosed superiorly by the developing corpus callosum to form the cavum
septi pellucidi.

Barkovich_Chap05.indd 372 5/6/2011 8:58:37 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 373

The first callosal axons to cross along the glial sling arise from the splenium forms over the hippocampal commissure after the anterior
developing cingulate gyri (Fig. 5-5C) (47,54,55). These cingulate axons corpus callosum and it enlarges later (35,58,59).
act as pioneer fibers, guiding later axons from neocortical neurons. At The septum pellucidum forms in close association with the anterior
this stage, three separate sites of commissuration can be recognized corpus callosum. Its lower margin contains the early axons (week 10) of
(Fig. 5-4D and F): the ventral lamina reuniens (forming the anterior the fornix between the septal area and the hippocampus, (week 11). The
commissure), the dorsal lamina reuniens (forming the hippocampal fibers that connect the medial septal nucleus with the ipsilateral cingu-
commissure), and the glial sling (composed of pioneer corpus cal- late cortex develop in the septal leaves at the same time as the pioneer
losum axons bridging the interhemispheric fissure at the level of the callosal axons in the rat; they cross the glial sling and the corpus cal-
corticoseptal boundary). Later formed neocortical commissural axons losum along the way. Because the septum pellucidum develops in the
will use these commissural sites to cross the midline by a process called walls of the SMTM, it becomes circumscribed by the lamina rostralis,
fasciculation (by recognizing chemical signals on the axons that have genu, and body of the corpus callosum (Fig. 5-4F). The interhemispheric
already crossed, rather than signals from the developing brain, their space between the septal leaves becomes the cavum septi pellucidi (Fig.
migration is much less complex). As the number of neocortical axons 5-5C) (35,60); when the cavum extends posteriorly between the corpus
crossing at the more posterior hippocampal commissure and at the callosum and the hippocampal commissure, this extension is called the
more anterior corticoseptal boundary grows, these structures eventu- cavum Vergae. Initially thought to be a meningeal space (35), recent evi-
ally meet in what will become the posterior body of the corpus cal- dence suggests that it is the space that is left after the disappearance of
losum. Therefore, the anterior and the posterior callosum originally the glial sling (46), explaining why no meningeal epithelium is found on
are separated (40) but eventually unite to form a single structure. the medial aspect of the septal leaves in a premature infant with cavum
Because of the disproportionate growth of the frontal lobes in humans, septi pellucidi (37). The cavum becomes apparent about 20 weeks and
the hippocampal commissure, although initially in the lamina reu- usually disappears within 3 months after birth, its lumen becoming pro-
niens, is pushed posteriorly (stretching the forniceal columns) and gressively effaced from the back to the front (61,62).
becomes located under the splenium (35). This apparent front-to-back
progression is generally interpreted as a front-to-back growth of the Spectrum of Commissural Abnormalities
corpus callosum but is in fact the front-to-back relative displacement If the normal developmental process is disturbed, the development
of the hippocampal commissure (and of the splenial axons attached of all three of the cerebral commissures or any combination of the
to it) that results from the back-to-front growth of the frontal lobes cerebral commissures may be affected; this results in a spectrum of
and the corresponding accumulation of crossing axons in the anterior commissural anomalies (Fig. 5-7). The corpus callosum itself may be
callosal segment. Therefore, while the initial pioneer axons are begin- completely absent (agenetic) or partially formed (hypogenetic). As
ning to cross in the region of the interventricular foramina of Monro discussed in section “Anatomy and Embryology,” the corpus callo-
[35]), the glial “sling” is just beginning to form in the anterior body, sum originates in two independent portions, with axons crossing by
while the SMTM is forming in the posterior body (56,57). The genu, way of a glial sling anteriorly and the hippocampal commissure pos-
lamina rostralis, and body appear to form in rapid succession and all teriorly. The hippocampal commissure forms at week 11, before the
are present at week 15 (Fig. 5-6) (53,58); the hippocampal commis- anterior callosum at week 13; later callosal growth (through fascicu-
sure develops earlier, but the splenium (using hippocampal commis- lation) is mostly anterior prenatally, pushing the hippocampal com-
sure axons as guides for fasciculation to cross the midline) does not missure and associated posterior callosum posteriorly. Knowledge
become prominent until weeks 18 to 19 (53). So although the anterior of this sequence of development helps to differentiate the variants
corpus callosum (including the lamina rostralis) appears after the hip- of commissural malformation: (a) complete tricommissural agen-
pocampal commissure, it enlarges and becomes grossly visible first; the esis (global failure of the midline crossing processes, Fig. 5-8); (b)

FIG. 5-6. Early fetal development of the commissures. A and B. Midline sagittal T1 images of anatomic specimens estimated at 13 to 14 weeks. The corpus
callosum is short (white arrows) and the splenium has not developed yet although the hippocampal commissure is apparent. C. In an 18-week specimen,
the splenium can be identified, and the anterior corpus callosum has grown posteriorly as the frontal lobes expand, pushing the hippocampal commissure
and attached splenium dorsally.

Barkovich_Chap05.indd 373 5/6/2011 8:58:38 PM


374 Pediatric Neuroimaging

ant post

cc
s
s hc
A. a-commissural B. normal anatomy

Probst Probst FIG. 5-7. Schematic representations of various commissural and sep-
tal disorders. A. With complete absence of commissures, the leaves of
s hc
the septum pellucidum (s) border the medial edge of the limbic lobe.
C. global commissural agenesis D. isolated callosal agenesis B. During normal development, the anterior corpus callosum (blue, left)
at the corticoseptal boundary, the hippocampal commissure (violet)
and the splenium (blue, right) between the fornices, cross the midline.
ant post
C. In classic commissural agenesis, the commissural fibers do not cross
the midline. Callosal axons run parasagittally within the pellucidal leaves
(forming Probst bundles, blue ellipses) instead and fornices (violet ovals)
are not connected. D. In isolated callosal agenesis, the hippocampal com-
cc missure (hc, violet line between fornices) is present. E. In isolated agenesis
cc
of the hippocampal commissure, the corpus callosum (cc) is present, but
s hc fornices are not connected. F. In septooptic dysplasia, both callosal and
hippocampal commissures are present but the leaves of the septum pel-
E. agenesis of hippocampal commissure F. septo-optic dysplasia
lucidum are missing.

complete agenesis of corpus callosum, which almost always involves callosum to grow and push the hippocampal commissure poste-
both the callosal and the hippocampal commissures (Fig. 5-9); (c) riorly (Fig. 5-11); or (e) failure of the posterior callosum to grow
posterior agenesis, which may relate to a failure of the hippocampal (Fig. 5-12). Rarely, the hippocampal commissure may form in the
commissure (and therefore of the posterior callosum) to form (Fig. absence of a corpus callosum (Fig. 5-13); more rarely still, only an
5-10); (d) insufficient commissuration, with failure of the anterior intermediate segment is missing between anterior callosal axons and

FIG. 5-8. Complete (tricommissural) agenesis. A. Midline sagittal T1-weighted image shows absence of the anterior, callosal
and hippocampal commissures, with a high-riding third ventricular roof, radial pattern of the medial hemispheric sulci, and
normal posterior fossa contents. B. Coronal T2-weighted image shows widely spaced lateral ventricles bordered medially by
heavily myelinated Probst bundles (white arrows) in the septum pellucidum (resulting in the crescentic appearance of the
ventricular lumen). Note the abnormally shaped hippocampi, caused by extension of the temporal horns into the parahip-
pocampal gyri (black arrows) where the white matter tract (ventral cingulum) is hypoplastic or absent.

Barkovich_Chap05.indd 374 5/6/2011 8:58:39 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 375

FIG. 5-8. (Continued) C. Axial T2 image shows a wide interhemispheric fissure with prominent ventricular
atria (colpocephaly, black arrows), likely because of the poorly developed posterior white matter. D. Axial
T2-weighted image shows the parallel bodies of the lateral ventricles, separated from the interhemispheric
fissure by Probst bundles (b) and the medial hemispheric cortex.

a well formed splenium/hippocampal commissure (63,64). Callosal subsequent crossing of anterior callosal axons; also, absence of septal
anomalies in classic holoprosencephaly are atypical, as the splenium cortex precludes formation of a distinct fornix (hippocamposeptal
may be present without a normal genu or body, or the callosal body fibers). Yet, on the posterior cortical margin along the single ven-
and splenium may be present in the absence of a genu (63,65). In tricle, forniceal fibers are present, allowing development of a hip-
the middle interhemispheric (MIH) variant of holoprosencephaly, pocampal commissure, and the callosal fibers of the posterior part
the genu and splenium may be present without a normal callosal of the hemispheres may fasciculate along them to form a splenium,
body (66). Examples are shown in the section “Holoprosencephaly” or a splenium and body depending on the severity of the failure of
of this chapter. In classic semilobar or lobar holoprosencephalies, the interhemispheric fissure formation (64,67). In the interhemispheric
frontal lobe is undivided, and no midline corticoseptal boundary can variant of holoprosencephaly, the presence of the posterior corpus
develop to allow formation of an interhemispheric glial sling and callosum/hippocampal commissure may have the same explanation;
the anterior corpus callosum may be present because the septal area
developed normally with a normal anterior corticoseptal boundary,
a normal glial sling, indusium and glial wedge and, therefore, nor-
mal anterior callosum; the posterior body does not form because the
interhemispheric continuity has impaired formation of the corti-
coseptal boundary and prevented the eventual fusion of the anterior
and posterior callosal segments (38).
Specific callosal changes may be observed when the region of brain
that sends axons through a specific region of the corpus is destroyed
(e.g., porencephalies and schizencephalies). Those patients who have
had corpus callosotomies for seizure surgery (68) or transcallosal sur-
gical approaches to the lateral or third ventricles (69) have callosal
defects that may mimic congenital callosal defects if the surgical his-
tory is not known.
As stated above, the anterior commissure and hippocampal com-
missure are typically affected when the corpus callosum develops
abnormally (70). Most commonly, the anterior commissure will be
small and the hippocampal commissure absent in the setting of callosal
agenesis (64,70). Occasionally, however, the hippocampal commissure
will be enlarged in patients with hypogenesis or agenesis of the corpus
callosum (57): the enlarged commissure may mimic a section of cal-
losum on midline sagittal images; analysis of coronal images will show
that this commissure connects the fornices, not the cerebral hemi-
FIG. 5-9. Classic “callosal” agenesis. Midline sagittal T1 image shows a spheres (Fig. 5-13). An enlarged anterior commissure may be seen in
normal-looking anterior commissure. The appearance otherwise is similar to callosal agenesis, but this was rarely observed in large reported series
that of Figure 5-8A; both callosal and hippocampal commissures are missing. (64,71,72) and appears to be quite unusual.

Barkovich_Chap05.indd 375 5/6/2011 8:58:40 PM


376 Pediatric Neuroimaging

FIG. 5-10. Callosal hypogenesis (partial agenesis) in three patients; sigmoid bundle and tractography. A–C. Classic Probst bundles. Midline sagittal
T1-weighted image (A) shows a short segment of corpus callosum (white arrows) above the anterior commissure, in front of the foramen of Monro;
the lamina rostralis, the fornix, and the hippocampal commissure are not seen. Coronal T2-weighted image (B) shows an anterior callosal remnant
(black arrows) in the location of the genu, while a more posterior image (C) shows the typical appearance of commissural agenesis, with Probst bundles,
abnormal hippocampi, and medial extension of temporal horns. D and E. Sigmoid bundle. Midline sagittal T1-weighted image (D) shows a small cal-
losal remnant (white arrow) and an anterior commissure (white arrowhead). Axial T2-weighted image (E) shows a “sigmoid bundle”: a bundle of axons
starting in the right forceps minor (small white arrows), crossing the midline (black arrow, this is the callosal “remnant” seen in D), and extending into
the contralateral hemisphere in the location of a Probst bundle (large white arrow). White matter injury is present in the left hemisphere. F–H. Homo-
topic and heterotopic bundles identified by diffusion tensor imaging. Sagittal T1-weighte d image (F) shows callosal hypogenesis with small inferior
genu, absent rostrum, posterior body, and splenium. Fiber tracking of callosal axons superimposed on a color FA map shows homotopic bundles in the
forceps minor (blue), central cerebrum (orange), parietal lobe (purple), and forceps major (green). The yellow, heterotopic “sigmoid” bundle, crossing
from the left frontal lobe to the right occipital lobe, is seen with the other crossing fibers in (G) and is isolated in (H). (Figures F, G, and H are courtesy
Michael Wahl, San Francisco.)

Barkovich_Chap05.indd 376 5/6/2011 8:58:41 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 377

FIG. 5-10. (Continued)

FIG. 5-11. “Classic” posterior callosal hypogenesis (partial agenesis). A. Midline sagittal T1-weighted image shows a short,
but otherwise complete commissural plate. The anterior commissure is tiny or absent. B. Coronal T2-weighted image
through the amygdalae shows incomplete fusion of the septum pellucidum leaves. In this case, the posterior callosum is miss-
ing, but the fornix is abutting the undersurface of the posterior callosum (in A), which appears to be a normal splenium. This
malformation might result from a failure of the anterior callosum to grow normally and to displace the hippocampal com-
missure and splenium posteriorly. Note the small septum pellucidum between the fornix and the corpus callosum in A.

Barkovich_Chap05.indd 377 5/6/2011 8:58:43 PM


378 Pediatric Neuroimaging

FIG. 5-12. Callosal tapering, with mild posterior callosal hypogenesis. On this
midline sagittal T1-weighted image, the anterior commissure and anterior cal-
losum look normal, with a normal junction of the fornix suggesting a normal
hippocampal commissure. The splenium is less well developed suggesting a pos-
terior callosal commissural defect. As is commonly seen with splenium hypopla-
sia, the rostrum is absent.

Associated Anomalies and Syndromes More typically, affected patients present with seizures, macrocephaly,
The formation of the corpus callosum and its precursors occurs delayed development, mental retardation, or hypothalamic dysfunc-
between about 8 weeks and 20 weeks of gestational age (35,73). This tion (Fig. 5-16) (30,33,64,76,77). Those who are cognitively and neu-
corresponds to the period of the bulk of neuronal migration in the rologically normal often suffer from behavioral and neuropsychiatric
forebrain, and most of the cerebrum and cerebellum also form at the problems, leading to learning difficulties (78), sleep disorders (79),
same time. Therefore, in addition to anomalies of the other telenceph- language and social communication disorders (80), and visuospatial
alic commissures, anomalies of the corpus callosum are often associ- attention deficits (81).
ated with other anomalies of the cerebrum and cerebellum, such as Callosal anomalies are a part of many syndrome complexes
the Chiari II malformation, cerebellar hypoplasias, the Dandy-Walker (74,77,82–84); a 2010 search on OMIM (Online Mendelian Inheritance
spectrum (Fig. 5-14), malformations of cortical development (Fig. in Man) retrieves 187 syndromes in which “corpus callosum agenesis”
5-15), cephaloceles, hypothalamic malformations, and midline facial is a significant feature. The most frequently mentioned of these is the
anomalies (57,74,75). Although it is reported that isolated callosal agen- Aicardi syndrome (82). Aicardi syndrome is an X-linked dominant dis-
esis may be asymptomatic, in the experience of most neurologists and order consisting of infantile spasms, multiple cerebral malformations
neuroradiologists, incidental detection of callosal agenesis is very rare. (most commonly callosal agenesis, gray matter heterotopia, and poly-

FIG. 5-13. Callosal agenesis with hippocampal commissure mimicking a splenium. A. Midline sagittal T1-weighted image shows
apparent formation of the splenium in the absence of the anterior callosum. B. Coronal T1-weighted image shows that the commissure
connects the fornices and, therefore, is a hippocampal commissure, not a splenium (Reprinted from Barkovich AJ. Apparent atypical
callosal dysgenesis: analysis of MR findings in six cases and their relationship to holoprosencephaly. Am J Neuroradiol 1990;11:333–340,
with permission.).

Barkovich_Chap05.indd 378 5/6/2011 8:58:45 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 379

rarely callosal hypoplasia, periventricular nodular and subcortical het-


erotopia, PMG, posterior fossa and other extraparenchymal cysts, cer-
ebellar hypoplasia, choroid plexus papillomas, and microphthalamia.
Ophthalmologic exam reveals characteristic chorioretinal lacunae,
which result from retinal dysplasia, and ocular colobomata. Myelination
may be delayed (82,88–90). Callosal agenesis is also the most common
brain anomaly seen in the fetal alcohol syndrome, although patients
with this disorder more typically have normal neuroimaging studies;
the diagnosis is established by history and recognition of characteris-
tic facial and neurologic features. Patients with severe facial anomalies,
such as midline craniofacial dysgenesis (see section on “Cephaloceles
and Other Calvarial and Skull Base Defects”) or large midline clefts
have a high incidence of callosal anomalies. Other commissural abnor-
malities seen on neuroimaging studies include ventriculomegaly and
cavum septi pellucidi et Vergae (91).
The septum pellucidum may also be too small (surface area dispro-
portionately small in relation to the corpus callosum) with apparently
normal fornices. In hemimegalencephaly (HME), the septum may be
too thick and eccentric toward the dysmorphic hemisphere. Rarely,
the septum may be abnormally thick as an isolated anomaly; possible
FIG. 5-14. Commissural agenesis with posterior fossa cyst. Midline sagit- causes include the presence of too many axons, an aberrant white mat-
tal T2-weighted image shows that the anterior commissure (black arrow) is ter fascicle in the midline, or the persistence of residual cellular mate-
present, but not the hippocampal or the callosal commissures. The poste- rial, possibly glial or neuronal, in the lumen of the cavum. This finding
rior fossa is large with high insertion of the tentorium, hypoplastic rotated has not been associated with any clinical condition (92).
vermis, small pons, and a huge cistern magna. A complete list of syndromes with commissural anomalies is
beyond the scope of this text. Table 5-1 includes a listing of some of
the most common associated syndromes. Many of these syndromes are
microgyria [PMG]), chorioretinopathy, and an abnormal EEG pattern discussed in this chapter or other portions of this book.
(hypsarrhythmia). The syndrome occurs almost exclusively in females
(affected patients must have two X chromosomes, so patients with Anatomic Sequelae of Callosal Anomalies
Klinefelter syndrome [47XXY] can also have it [85]) with no family his- The imaging findings in patients with callosal anomalies vary with the
tory of ophthalmologic or neurologic disease. The mutation results in a cause of the callosal anomaly. If the problem is lack of formation or
spontaneous balanced translocation of the X chromosome. Prevalence destruction of the callosal axons, the result is a marked reduction of
rate is 2 to 15 per 100,000 girls (86). Affected patients typically have cerebral white matter; in such cases, the commissures are often abnor-
severe global developmental delay and early-onset, medically refrac- mal, with portions either too thin or completely missing (Fig. 5-3).
tory epilepsy (86,87). Intracranial anomalies (Fig. 5-17) include callosal When the problem is an abnormality within the interhemispheric
agenesis or hypogenesis (often associated with interhemispheric cysts), fissure, with an inability of the axons to cross the midline, the axons

FIG. 5-15. Commissural agenesis with polymicrogyria. A. Midline sagittal T1-weighted image shows that the anterior
commissure is present, but not the hippocampal or callosal commissures; a persistent craniopharyngeal canal (white
arrows) is present. B. Axial T2-weighted image shows bilateral frontal polymicrogyria.

Barkovich_Chap05.indd 379 5/6/2011 8:58:46 PM


380 Pediatric Neuroimaging

FIG. 5-16. Posterior tapering of the corpus callosum with hypothalamic


hamartoma, pituitary stalk agenesis, and posterior pituitary ectopia. The cor-
pus callosum is short with posterior tapering (large white arrows), but the junc-
tion with the fornix is normal; this suggests global callosal hypoplasia. Note the
hypothalamic hamartoma (small white arrow) associated with the absence of
the anterior commissure and the ectopic location of the hyperintense posterior
pituitary lobe (white arrowhead).

that would normally course into the contralateral hemisphere through the medial borders of the lateral ventricles, giving the ventricles a
the corpus instead turn at the interhemispheric fissure and run parallel crescentic shape, which is most pronounced frontally (Figs. 5-8B and
to that fissure, forming the U-shaped longitudinal callosal bundles of 5-19) (35,56,57). In either case, the third ventricle is located higher
Probst (Figs. 5-8B and 5-18) that form intrahemispheric rather than than normal, between the lateral ventricles, and the foramen of Monro
interhemispheric connections (98). These callosal fibers are best con- tend to be enlarged. When the callosal body is absent, the bodies of
sidered heterotopic; they connect different areas within one hemisphere the lateral ventricles are straight and parallel, away from the midline
rather than symmetrical areas of opposite hemispheres, resulting in a and separated from it by the medial walls of the hemispheres (Fig.
significant variant of normal functional anatomy. The Probst bundles 5-8D). Rarely, hypertrophy of the hippocampal or anterior commis-
lie lateral to the cingulate gyri and medial to the medial walls of the sure accompanies callosal agenesis or hypogenesis (Fig. 5-13) (63). The
lateral ventricles; their inferomedial borders merge with rudimentary formation of the corpus callosum seems to be associated with medial
fornices. Because of their location, the bundles of Probst invaginate and upward rotation of the cingulate gyrus with consequent forma-

FIG. 5-17. Aicardi syndrome. A. Midline sagittal T1-weighted image shows hypogenetic corpus callosum with a large interhemispheric cyst
and cerebellar vermian hypoplasia. B. Axial T1-weighted image shows the large interhemispheric cyst, multiple subependymal/subcortical
heterotopia and dysplastic cortex in both hemispheres.

Barkovich_Chap05.indd 380 5/6/2011 8:58:47 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 381

TABLE 5-1 A Partial List of Syndromes


Associated with
Commissural Anomalies
Aicardi syndrome
Apert syndrome
Chiari II malformation
CRASH syndrome (Chapter 8)
Dandy-Walker malformation
Fetal alcohol syndrome
Frontonasal dysplasia (93)
Inversion-duplication of short arm of chromosome 8
Median cleft face syndrome
Morning glory syndrome (usually with sphenoidal cephalocele)
Mowat-Wilson syndrome (94) FIG. 5-18. Formation of the parasagittal bundles (of Probst). Because of
the lack of induction by the midline glia, the commissural axons fail to cross
Neu-Laxova syndrome (95) the midline; instead when they reach the corticoseptal boundary they turn
Nonketotic hyperglycinemia (Chapter 3) and course parallel to the interhemispheric fissure within the septal leaves,
indenting the medial walls of the lateral ventricles. Broken lines represent
Pyruvate dehydrogenase deficiency (Chapter 3) fibers of the corpus callosum in the normal brain. Solid lines represent the
Oro-facial-digital syndromes fibers that fail to cross in callosal agenesis. (Modified from Rakic P, Yakovlev
PI. Development of the corpus callosum and cavum septae in man. J Comp
Rubinstein Taybi syndrome (96) Neurol 1968;132:45–72, with permission.).
Shapiro syndrome (hypothalamic-pituitary dwarfism and
paroxysmal hypothermia) (97)
Walker-Warburg syndrome enlarge, with the temporal horns extending inferomedially into the
parahippocampal gyri, where the hypoplastic ventral cingulum would
normally be found (Figs. 5-8B and 5-10C) (100–102). This ventricular
configuration is known as colpocephaly.
tion of the cingulate sulcus. When the corpus callosum does not form,
the cingulate gyri do not rotate and are small due to hypoplasia of the Imaging of Callosal Anomalies
cingulum (99,100); thus, the cingulate sulci remain unformed (57) and Agenesis of the corpus callosum can be detected on images obtained
the medial hemisphere sulci radiate all the way to the third ventricle in the axial plane, although the sagittal and coronal planes better
(Fig. 5-8A). In addition, the medial hemispheric sulci have a rather dis- demonstrate the associated anatomic deformities (Fig. 5-10). The
organized appearance, probably secondary to disorganization of the characteristic lateral convexity of the frontal horns, parallel lateral
white matter tracts in and near the midline (Figs. 5-8 to 5-11). This ventricles, colpocephaly, and upward extension of the third ventricle
abnormal appearance of the medial hemispheric sulci is one of the into the interhemispheric fissure between the lateral ventricles can all
hallmarks of absence of the corpus callosum; this finding is especially be identified in the axial plane (Fig. 5-8). Milder callosal anomalies,
helpful when evaluating newborns, in whom the corpus is thin and however, are difficult to identify with axial images. MRI, ultrasound,
often difficult to see (see Chapter 2). and modern multidetector CT all allow images to be easily obtained
The corpus callosum is the most tightly packed bundle of axons in or reformatted into the coronal and sagittal planes, but the tissue
in the brain. This compactness makes the corpus a very firm structure discrimination and the spatial definition of MRI are much superior
and gives the adjacent lateral ventricles their shape; the firmness of the to those of other modalities. In addition, DTI and tractography may
corpus also helps the ventricles to maintain their normal size, espe- allow a better understanding of the abnormal intrahemispheric and
cially posteriorly. The firm caudate heads and lentiform nuclei keep interhemispheric connectivity (Fig. 5-10G and H). The severity of cal-
the size of the frontal horns relatively small, even in the absence of losal hypogenesis or dysgenesis is assessed well on the midline sagit-
the corpus callosum (although, when the genu is absent, the frontal tal images (Figs. 5-8 to 5-12). As mentioned earlier, when the corpus
horns are convex laterally instead of concave, as in the normal case) is hypogenetic, the posterior portions and the inferior genu and ros-
(Figs. 5-8B and 5-10C). Posteriorly, however, only loose white matter trum are usually absent. Therefore, a hypogenetic corpus callosum
surrounds the ventricles when the callosal splenium is absent. More- may include the posterior genu; posterior genu and anterior body;
over, multiple white matter tracts (including the dorsal cingulum, genu and entire body; or entire genu, body, and hypoplastic splenium.
within the cingulate sulcus, and the ventral cingulum, within the para- Other times, the hypogenetic corpus callosum may be mistaken for
hippocampal gyrus) are often hypoplastic or absent when the corpus is hypoplastic (Fig. 5-20); a disproportionately small or absent splenium
hypogenetic, including those coursing around the ventricular trigone helps to make the diagnosis of hypogenesis. Other MR findings in the
and temporal horn (64,99). In callosal hypogenesis, therefore, the tem- absence of a corpus callosum include lack of inward folding of the cin-
poral horns, trigones, and occipital horns of the lateral ventricles often gulate gyri (with the medial hemispheric sulci extending all the way

Barkovich_Chap05.indd 381 5/6/2011 8:58:48 PM


382 Pediatric Neuroimaging

Diffusion tractography has added to our knowledge about white


matter pathways in the setting of callosal anomalies. In patients with
complete callosal agenesis, the axons running through the Probst bun-
dles connect anterior regions of the hemisphere with more posterior
regions, with the axons exhibiting a topographic organization (98).
In patients with hypogenesis of the corpus callosum, axons running
through the callosal remnant course not only to the expected areas but
also to all regions of the brain, both symmetrically (homotopic con-
nectivity) and asymmetrically (heterotopic connectivity) via so-called
sigmoid bundles (Fig. 5-10) (98,99).

Callosal Agenesis with Interhemispheric Cyst


Agenesis of the corpus callosum with interhemispheric cyst is a special
condition that may have a different cause than other types of callosal
agenesis. ACC with IHC can be divided into two major groups, one
in which the interhemispheric cyst is a diverticulum of the ventricu-
lar system and thus communicates with the ventricles (Type 1) and
one in which multiple, cysts are present that do not communicate with
FIG. 5-19. Schematic drawing illustrating the findings in callosal agen- the ventricles (Type 2, see Table 5-2) (103). Type 1 cysts are isointense
esis (compare with Fig. 5-8B). The lateral ventricles have a crescentic shape to CSF (Fig. 5-21), whereas Type 2 cysts are usually slightly hyperin-
secondary to the medially located bundles of Probst. The third ventricle tense to CSF on T1-weighted images and isointense to hyperintense
expands upward between the lateral ventricles into the interhemispheric on T2-weighted images (Figs. 5-22 and 5-23). Type 2 ACC with IHC is
fissure. The cingulate gyri remain everted and the cingulate sulci do not often associated with subcortical heterotopia or PMG and the Type 2b
form. (Reprinted from Barkovich AJ. Apparent atypical callosal dysgenesis: cysts are frequently found in Aicardi syndrome. Pavone et al. (104) have
analysis of MR findings in six cases and their relationship to holoprosen-
shown that large portions of the corpus callosum may be present and
cephaly. Am J Neuroradiol 1990;11:333–340, with permission.)
that affected patients frequently have severe neurological and develop-
mental disorders. Of note, as with all types of cysts, the cysts associated
to the third ventricle), crescent-shaped lateral ventricles (caused by with callosal agenesis may not develop or become apparent on imaging
an impression upon the medial walls of the ventricles by the medially until late in gestation (105,106). As such cysts are frequently associated
positioned bundles of Probst), incomplete rotation of the hippocampal with macrocephaly or frank hydrocephalus at birth, repeat sonographic
formation in the medial temporal lobes, inferomedial extension of the examination before delivery is recommended (105,106).
temporal horns of the lateral ventricles (due to hypoplasia of the ven- Type 2 cysts are often multiloculated. Under these circumstances,
tral cingulum [100]), and extension of the roof of the third ventricle it is important to establish which of the loculations communicate with
into the interhemispheric fissure (Figs. 5-9 to 5-11). one another and with the ventricles, in order to properly plan which
compartments should be shunted and in what order. A CT cystogram
or ventriculogram is performed after nonionic iodinated contrast is
introduced into the cyst(s) to establish which of the CSF collections
communicate.
Patients with Type 1 interhemispheric cysts are most commonly
boys who present with macrocephaly or cranial deformity. Patients
with Type 1a and Type 2d cysts have the best prognosis in our experi-
ence, while those with Types 1c, 2b, and 2c have the worst prognoses.
Some studies indicate that developmental delay and focal neurologic
deficits are common in Type 1 cysts, but are uncommon and less severe
in Type 2 cysts (64,107); however, this has not been verified in a pro-
spective series. Seizures develop in less than half of affected patients.
Shunting or fenestration is usually necessary in Type 2 cysts; if the
cysts are adequately decompressed, many of these patients may ulti-
mately have normal neurologic exams and normal school performance
(64,70), although epilepsy may become a problem.

Diagnosis of Callosal Agenesis/Dysgenesis in the Fetus


Fetal ultrasound is performed in virtually every pregnancy in indus-
trialized countries, and the diagnosis of a major malformation such as
a commissural agenesis is usually suggested on the basis of this exam;
in particular, ventriculomegaly (especially bilateral colpocephaly, with
FIG. 5-20. Hypogenesis mimicking hypoplasia. All the commissures are
posterior dilatation of the lateral ventricles) and absence of a normal
small, but present. The round commissure (white arrowhead) above the cavum septi pellucidi suggests the diagnosis, especially if associated with
posterior third ventricle is the hippocampal commissure. Although this a large interhemispheric fissure or interhemispheric cysts (108,109).
might easily be mistaken for callosal hypoplasia, absence of the inferior However, ultrasonography may fail to demonstrate the malformation
genu and hypoplasia of the splenium suggest that this is hypogenesis. itself, either because the surrounding tissues obscure the fetal head or

Barkovich_Chap05.indd 382 5/6/2011 8:58:49 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 383

TABLE 5-2 Classification of Callosal Agenesis with Interhemispheric Cyst


Cyst Type Sex Age, Reason of Presentation Cyst Characteristics Associated Anomalies
Type 1 Communicate with Ventricles
1a M Neonate, macrocephaly Isointense to CSF. Hydrocephalus.
Unilocular.
Communicates with ventricles.
1b M>F Neonate, macrocephaly Isointense to CSF. Thalamic fusion without
Unilocular. subcortical heterotopia.
Communicates with third ventricle.
1c M Neonate, seizures, Isointense to CSF. Small ipsilateral cerebral
microcephaly Unilocular. hemisphere.
Communicates with lateral and third ventricle.
Type 2 No communication with ventricle
2a M Neonate, macrocephaly Isointense to CSF. Hydrocephalus.
Multilocular.
No communication with ventricles.
2b F Infant, neurodevelopmental Multiloculated. Subependymal heterotopia.
disorders (severe), seizures, No communication with ventricles. Polymicrogyria.
micro/macrocephaly Hyperdense on CT, hyperintense on T1MR. Deficient falx cerebri.
2c M Childhood seizures, delay Isointense to CSF. Subcortical heterotopia.
Multilocular.
No communication with ventricles.
2d ? Macrocephaly Arachnoid cyst. None.
Isointense to CSF.
Unilocular.
No communication with ventricles.

Adapted from Barkovich AJ, Simon EM, Walsh CA. Callosal agenesis with cyst: a better understanding and new classification. Neurology 2001;56:220–227,
with permission.

because the malformation is mild (e.g., in mild callosal hypogenesis). (rather than hypoplastic or atrophic) on the midline sagittal cut alone
If possible, endovaginal ultrasonography may significantly improve (Figs. 5-24 and 5-25). Interhemispheric cysts are easily recognized in all
the diagnostic efficacy, but depending on the obliquity of the head, it three planes (Fig. 5-26), whereas malformations of the posterior fossa
may be limited by the small range of lateral motion of the probe (109). (brainstem, cerebellum) are better identified and characterized on sag-
Also, ultrasound may fail to demonstrate associated brain abnormali- ittal and axial images. Other commonly associated abnormalities better
ties that significantly worsen the prognosis. At many institutions, fetal shown by MRI include delayed sulcation and/or shallow sylvian fissure,
MRI is used to confirm the diagnosis of callosal agenesis/hypogenesis gray matter heterotopia, PMG, lissencephaly (with thick cortex), schi-
and to look for associated malformations because the diagnostic yield zencephaly, and choroid plexus cysts (32,108–111). The remainder of
is typically much superior (32,108–110). Fetal MRI may also be useful the fetus should be examined, if possible, for extraneural abnormalities
to diagnose commissural agenesis in fetuses referred for nonspecific of the skeleton, heart, and trunk.
ventriculomegaly or to rule out brain anomalies in fetuses where the
cavum septi pellucidi could not be identified on ultrasound. How- Malformations of Cerebral Cortical Development
ever, if abnormalities are mild and are missed on ultrasonography, no As a result of a greater awareness of their presence and of steadily
fetal MRI will be performed, probably explaining the lower incidence improving imaging techniques, malformations of cerebral cortical
of partial relative to complete agenesis in prenatal, as compared with development are being discovered with increasingly greater frequency
postnatal, series of commissural anomalies (32). on imaging examinations of children with developmental delay or
MRI is the optimal modality to assess the fetal brain parenchyma partial epilepsy. Several studies have now shown that malformations
after 18 to 20 weeks gestational age. Any assessment obviously should of cortical development are the cause of 23% to 26% of intractable
be with full knowledge of normal morphologic and size milestones epilepsies in children and young adults (112–120), and 15% of lesions
for the gestational age (see Chapter 2). Commissure malformations resected for intractable epilepsy (121). This number emphasizes the
in young fetuses are most easily seen on coronal images and are eas- point that cortical malformations must be ruled out in essentially every
ily inferred from the ventricular morphology on the axial cuts; it may pediatric patient with developmental delay or epilepsy. Moreover, it
be more difficult to ascertain that the corpus callosum is really absent is becoming increasingly clear that many malformations of cortical

Barkovich_Chap05.indd 383 5/6/2011 8:58:50 PM


384 Pediatric Neuroimaging

FIG. 5-21. Two examples of callosal hypogenesis and Type 1 interhemispheric cyst. A. Sagittal T1-weighted image shows absence of the
commissures and a large interhemispheric cyst (black arrows) in a microcephalic patient; note the associated malformations of the brainstem
and vermis. B. Coronal T1-weighted image shows that the cyst is continuous with the third and lateral ventricles and straddles the falx; no
septum pellucidum or Probst bundles can be identified. C. Sagittal T1-weighted image in a different patient reveals an enlarged head with a
small meningocele (white arrow) at the vertex. Only the genu of the corpus callosum is present. A large CSF-intensity region is seen rostral and
dorsal to the genu. The cerebellum and tentorium cerebelli are pushed down by the cyst. D. Axial T1-weighted image shows that the cyst is in
communication with the enlarged lateral ventricles.

development are caused by chromosomal mutations (122–132); thus, wall, between the developing third and lateral ventricles, known as the
the presence of cortical malformations is important for proper coun- subpallium germinal zone, ventral germinal zone, or the ganglionic
seling of parents of affected children. eminences (Fig. 5-27). The cells proliferating here will form the dien-
cephalon, the basal ganglia, and the GABAergic (neurons that make
Embryology of the Cerebral Cortex gamma-aminobutyric acid (153) as their neurotransmitter) interneu-
An understanding of the normal development of the cerebral cortex rons of the cerebral cortex, as discussed more completely in the fol-
is essential to understanding malformations of cortical development. lowing paragraph (133,134). Cells that will become the glutamatergic
Initially, after initial formation of the cerebral hemispheres from the (neurons that make glutamate as their neurotransmitter) neurons
developing prosencephalon, each cerebral hemisphere consists of of the cerebral cortex and glial cells are generated in the walls of the
a thick basal (ventral) portion, called the subpallium, and a thinner developing lateral ventricles, in proliferative zones called the pallial or
dorsal portion (the pallium, which will become the cerebral mantle). dorsal germinal zone (Fig. 5-28). Cell divisions begin in the dorsal ger-
The subpallium initially appears as bumps on the basal ventricular minal zones during the seventh week of gestation. During the eighth

Barkovich_Chap05.indd 384 5/6/2011 8:58:50 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 385

FIG. 5-22. Callosal agenesis with Type 2a interhemispheric cyst. A. Sagittal


T1-weighted image shows absence of the corpus callosum with a large midline CSF
collection inferiorly displacing the interthalamic adhesion (white arrows). B. Coronal
T1-weighted image shows the interhemispheric fluid collection extending on both
sides of the falx cerebri. The lateral ventricles (open white arrows) and third ventricle
(solid white arrow) are displaced by the interhemispheric cyst. C. Axial T2-weighted
image shows that the collection in fact is composed of multiple loculations.

gestational week, the first young neurons begin to migrate radially bodies act to guide migrating neurons (Fig. 5-29) when the hemispheres
outward from the germinal matrix in the walls of the lateral ventricles, enlarge. As different transcription factors start to be expressed, other
ultimately forming the cerebral cortex (27,29,135). Movement of neu- types of neurogenic progenitors form, called intermediate precursor
rons from the dorsal germinal zone to their final location is initially a cells (IPCs), neuroepithelial cells, and short neural precursors (27,137).
simple process. Cells in the germinal zone elongate, with the nucleus IPCs migrate to a transient SVZ between the ventricular zone and the
moving to the end of the cell that is farthest removed from the ventricu- intermediate zone (the developing white matter), where they divide
lar surface. Contact with the ventricular surface is then terminated and symmetrically into a pair of neurons that subsequently migrate to the
the remainder of the cell joins the nucleus at some distance from the developing cortex (29,138–142). Neuroepithelial cells and short neural
ventricular surface (136). Under the influence of certain genes, some precursors, in contrast, divide asymmetrically at the ventricular surface
neuroepithelial cells are induced to become specialized radial glial cells to produce only a single neuron or single IPC.
(RGCs) that span the entire thickness of the hemisphere from the ven- RGCs tend to be grouped in fascicles that consist of four to ten
tricular surface to the pia (Fig. 5-28) (27). The RGCs have two func- cells, all of which guide the migrating neurons. The fascicles supply
tions: (a) they are stem cells, which divide asymmetrically to produce necessary metabolites to the migrating neurons; they also organize the
daughter cells (neurons and glia), and (b) their elongated fiber-like vertical lamination of the developing neocortical plate (143). Neuronal

Barkovich_Chap05.indd 385 5/6/2011 8:58:51 PM


386 Pediatric Neuroimaging

FIG. 5-23. Type 2c callosal agenesis with interhemispheric cyst. A. Sagittal T1-weighted image shows absence of the corpus callosum
with a dorsal interhemispheric fluid collection of CSF intensity and a second collection (white arrows) that is hyperintense compared
to CSF. B. Axial proton density image shows several loculations between the hemispheres. The posterior one is hyperintense compared
to CSF. C. Postcontrast T1-weighted image shows enhancement of the walls of the posterior interhemispheric loculation. D. Axial
T1-weighted image shows subcortical heterotopia (black arrows) in both hemispheres.

migration along RGCs seems to be dependent upon (a) recognition to attach to the fascicle, and migration to the proper cortical layer and
of the glial cell by the neuron, (b) attachment of the neuron to the final position in the cortex (137,146–152). As migration nears comple-
glial cell, and (c) calcium entry into neuron; blocking calcium chan- tion, many of the RGCs begin to produce astrocytes, while others may
nels or blocking N-methyl-D-aspartate receptors inhibits cell migration transform into astrocytes (27).
(144,145). Recent studies of neuronal migration indicate that a diverse In contrast to the generation of glutamatergic neurons in the dor-
family of genes and transcription factors cooperate in orchestrating the sal ventricular zones, GABAergic neurons seem to be generated pre-
multistage, interrelated events involved in cortex development: control dominantly in the subpallium ventricular zones (the medial [MGE]
of mode of cell proliferation, fate determination, establishment of and lateral [LGE] ganglionic eminences and the preoptic area [POA],
polarity within the young neuron, detachment from the local substrate Fig. 5-27) (133,134). These zones have been divided based on the

Barkovich_Chap05.indd 386 5/6/2011 8:58:52 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 387

FIG. 5-24. Commissural agenesis in a 24-week GA fetus. A. Coronal image shows the characteristic appearance of commis-
sural agenesis. Note the bundles of Probst (black arrows) in the medial walls of the lateral ventricles, between the dark signal
layers of both the germinal matrix and medial cortex. B. On this midline sagittal image, the absence of corpus callosum is
difficult to confirm; complementary axial and coronal images are necessary to make the diagnosis in young fetuses.

FIG. 5-25. Commissural hypogenesis in a 24-week GA


fetus. A. Coronal image shows a rudiment of corpus cal-
losum bridging the hemispheres, but the general morphol-
ogy is similar to that in Figure 5-24A. B. On this sagittal
image, the corpus callosum is poorly visualized, just as in
Figure 5-24B. C. Axial image shows the typical appearance
of commissural hypogenesis with a wide interhemispheric
fissure and colpocephaly; the callosal rudiment (black
arrow) is well shown anteriorly.

Barkovich_Chap05.indd 387 5/6/2011 8:58:54 PM


388 Pediatric Neuroimaging

FIG. 5-26. Commissural agenesis with interhemispheric cyst in a 29-week GA fetus. A. Midline sagittal image shows the roof of the
third ventricle is pushed down by a midline cyst that appears separate from the third ventricle, suggesting a type 2 cyst. B. Axial image
shows wide separation of the hemispheres by a multiloculated interhemispheric CSF collection that is centered at the level of the
foramen of Monro; no commissure is seen, and the lateral ventricles have the characteristic morphology of posterior colpocephaly.
The right cerebral hemisphere seems dysmorphic, suggesting a Type 2c ACC with cyst.

expression patterns of several transcription factors, suggesting that POA progenitors produce neurons that migrate to the POA, amygdala,
specific features of the interneurons may be programmed very early globus pallidus, and cortex (158–163). It appears that the subtype
during the developmental process (28,154–157). Progenitors in the identity of the interneuron can be predicted by its time and place of
ganglionic eminences produce postmitotic neurons with distinct origin (154,156,159,161,164). POA-derived neurons destined for the
migratory trajectories and fates: LGE progenitors produce striatal cerebral cortex initially migrate to the developing cortical layer 1 and
projection neurons and olfactory bulb interneurons, whereas MGE then migrate inward to deeper cortical layers (161,165,166) whereas
progenitors produce cortical and hippocampal interneurons, and the cortical interneurons from the MGE migrate tangentially to the SVZ
of the dorsal pallium germinal zone (133,167–173) from where they
attach to a radial glial cell, along which they migrate radially outward
to the developing cortex (27,29,174). Other GABAergic cells are gener-
ated in the SVZ of the dorsal germinal zones (152, 170) but (as some
GABAergic neurons migrate through the SVZ) it is not entirely certain
whether the dividing cells originated in the dorsal ventricular zone, the
dorsal SVZ, or the ganglionic eminence (27). The distinct molecular
characteristics of neurons arising from different germinal zones may
also serve distinct roles in modulating cortical activity (133,164). Loss
of these neurons, either by lack of generation or abnormal migration,
causes significant neurologic dysfunction (175).
The cortical layer in which a neuron will eventually reside can be
predicted by the known sequence of neuronal development and migra-
tion (147,176). The level at which the migrating neuron disengages
from the radial glial cell seems to be determined by short range inter-
actions with other cells during the last mitotic cycle while the neuron
FIG. 5-27. This schematic demonstrates the ganglionic eminences, the is still in the germinal zone (177,178). This disengagement requires
germinal zones of the subpallium (thick basal [ventral] portion of the secretion of a glycoprotein (called reelin) by neurons in layer 1 (179).
developing cerebrum) where GABAergic neurons (those that secrete Reelin seems to be involved in the final stages of neuronal migration,
gamma aminobutyric acid—GABA—as a neurotransmitter) are generated, as well as disengagement of the migrating neurons from the RGCs
and the migration of those neurons. Neurons generated in the ventricular (180–182). In general, neurons migrate from the germinal zone to the
zone (VZ) and subventricular zone (SVZ) of the medial (MGE) and lateral
cortex in an “inside out” sequence. Those destined for the transient
(LGE) ganglionic eminences migrate locally to form the globus pallidus
(GP), the striatum (Str) and migrate tangentially to become GABAergic
deepest cortical layer (layer 7, also known as the “subplate” [183–187])
interneurons in the cerebral cortex (CTX). Neurons generated in the pre- migrate early, followed by those destined for layer 6, layer 5, layer 4,
optic area (POA) contribute to the POA, the amygdala and the globus palli- layer 3, and, finally, layer 2. The exception to the “inside out” rule of
dus, as well as some cortical interneurons. Also labeled is the thinner dorsal neuronal migration is that those neurons destined for the molecular
portion (the pallium), which will become the cerebral mantle. layer (layer 1) seem to be the first to arrive in the cortex (136,188,189).

Barkovich_Chap05.indd 388 5/6/2011 8:58:55 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 389

FIG. 5-28. This schematic demonstrates the dorsal (pallial) germinal


zone, where glutamatergic cerebral cortical neurons (those that secrete glu-
tamate as a neurotransmitter) are generated, and the migration of those
neurons. Some of the cells generated in the ventricular and subventricular
zones become radial glial fibers (rgf), which act as guides along which neu-
rons migrate radially to the developing cerebral cortical plate.

After their arrival in the cortex, neurons become arranged in discrete


horizontal layers and establish synaptic contacts with local and dis-
tant neurons in a process known as cortical organization (7,190,191).
The subplate (transient layer 7 of the cortex, which nearly disappears FIG. 5-29. Illustration of the relationship of the migrating neurons to
by term) is believed to play a large role in this organizational process fibers of the radial glial cells. A migrating neuron is seen ascending the radial
(183–187,192–194). glial fiber. Abnormal development of the neuron, damage to the radial glial
Regional specification of the cerebral cortex (remember that dif- fiber, or alteration of the molecules on the surface of either the migrating
ferent regions of the cortex have different layering patterns and differ- neuron or the radial glial cell will cause an arrest of cell migration.
ent functions) is thought to be determined by a combination of genetic
factors and connections with other portions of the nervous system normal gray matter. Abnormal T2 hyperintensity in the white matter
(152). It appears that early specification is the result of a cascade of is common in some of the malformations secondary to abnormal stem
genetic effects: morphogens and growth factors secreted by signalling cell development and in dystroglycanopathies; these findings will be
centers in the embryonic forebrain regulate expression of transcrip- discussed in the appropriate sections.
tion factor gradients across the developing cerebrum. The expression
of these transcription factors then impacts the expression of others, Classification of Malformations of Cerebral
and so on, until the final cellular phenotypes develop. This subject is Cortical Development
currently under active investigation (150–152). Malformations of cortical development are divided into three cat-
egories based upon the step at which cortical development was likely
Causes of Malformations of Cortical Development first disturbed: (a) stem cell proliferation or apoptosis, (b) neuronal
Any abnormality that inhibits neuronal or glial proliferation, neuronal migration, and (c) late migration and cortical organization (Table 5-3)
migration, or subsequent cortical organization can result in a malfor- (7). We will present the malformations in this order. Developmental
mation of cortical development. These abnormalities may include (a) anomalies of the cerebral neocortex occur in a number of different
gene mutations that interfere with cell proliferation, radial glial fascicle syndromes, many of which are listed with appropriate references in
development, the ability of the neurons to migrate, or the ability to Table 5-4 (199). The details of most of these syndromes will not be
disengage within the developing cortex and subsequently grow neu- elaborated upon in this chapter, as they are more appropriately dis-
rites and form synapses (7,195); (b) destructive events, such as infec- cussed in a genetics text. Only a few well-known, common, or particu-
tions or ischemia that damage the germinal matrix, the radial glial larly instructive syndromes will be elaborated upon.
fibers, the molecular layer, or the overlying “pial limiting membrane”
(191,196,197); or (c) the presence of exogenous toxins, such as drugs Special Imaging Techniques for Cortical Malformations
or alcohol from ingestion of toxic substances, or endogenous toxins Proper imaging technique and a high index of suspicion are crucial for
from metabolic disorders, such as pyruvate dehydrogenase deficiency the identification of cortical malformations. MRI is the imaging study
or nonketotic hyperglycinemia (see Chapter 3), that may interfere with of choice, as its high contrast resolution allows a much better analy-
one or more of the steps of cortical development (6,198). In malfor- sis of the cerebral cortex than any other neuroimaging technique. CT
mations secondary to abnormal migration and those secondary to misses the abnormality in more than 30% of affected patients (279).
abnormal late migration and cortical organization, the neurons tend Our protocol includes (a) T1-weighted volumetric three-dimensional
to be histologically normal. Therefore, the cortex usually has normal Fourier transformed (3DFT) spoiled gradient-echo sequences using
signal intensity on imaging studies; that is, their signal is isointense to 1.0 mm partition size (and reformatting in three orthogonal planes),

Barkovich_Chap05.indd 389 5/6/2011 8:58:56 PM


390 Pediatric Neuroimaging

TABLE 5-3 Malformations of Cortical Development


I. Malformations due to abnormal neuronal and glial proliferation C. Heterotopia
or apoptosis 1. Subependymal (periventricular)
A. Decreased proliferation/increased apoptosis or increased pro- 2. Subcortical (other than band heterotopia)
liferation/decreased apoptosis—abnormalities of brain size 3. Marginal glioneuronal
1. Microcephaly with normal to thin cortex III. Malformations due to abnormal cortical organization (including
2. Microlissencephaly (Extreme microcephaly with thick late neuronal migration)
cortex) A. Polymicrogyria and schizencephaly
3. Microcephaly with extensive polymicrogyria 1. Bilateral polymicrogyria syndromes
4. Macrocephalies 2. Schizencephaly (polymicrogyria with clefts)
B. Abnormal Proliferation (abnormal cell types) 3. Polymicrogyria or schizencephaly as part of multiple
1. Non-Neoplastic congenital anomaly/mental retardation syndromes
a. Cortical hamartomas of tuberous sclerosis B. FCD Types I, III
b. FCD Type II C. Microdysgenesis
c. HME IV. Malformations of cortical development, not otherwise classified
2. Neoplastic (associated with disordered cortex) A. Malformations secondary to inborn errors of metabolism
a. DNET 1. Mitochondrial and pyruvate metabolic disorders
b. Ganglioglioma 2. Peroxisomal disorders
c. Gangliocytoma B. Other unclassified malformations
II. Malformations due to abnormal neuronal migration 1. Sublobar dysplasia
A. Lissencephaly/subcortical band heterotopia spectrum 2. Others
B. Cobblestone complex/pial limiting membrane anomalies/con-
genital muscular dystrophy syndromes

TABLE 5-4 Syndromes Featuring a Neocortical Neuronal Migration Disorder


with Extracerebral Lesions
McKusick Other Brain Extracerebral Pattern of
Entity Number Cortical Dysplasia Anomalies Symptoms Inheritance References
Chromosome Anomalies
Deletion 1p36.3 — Polymicrogyria Atrophy, leukoen- Clinodactyly, mid- Chrom (200)
cephalopathy, thin face hypoplasia,
corpus callosum flat nasal bridge,
cardiac defects
Inversion 2p12–q14 — Pachygyria Holoprosencephaly, Trigonocephaly, Chrom (201)
microcephaly ptosis, coloboma,
hearing loss (see
Baraitser-Winter
syndrome)
Duplication 3q — Polymicrogyria, — Hypertrichosis Chrom (202)
microcephaly, hyp-
oplasia of olfactory
bulbs
Deletion 4p– — Heterotopia, Microcephaly Prominent Chrom (1183,1184)
microgyria glabella, down-
turned mouth,
preauricular pit,
cardiac defects
Deletion 4q — Heterotopia, Microcephaly, wide Flat malae, promi- Chrom (203)
pachygyria ventricles nent lower lip,
Fallot tetralogy

(Continued)

Barkovich_Chap05.indd 390 5/6/2011 8:58:58 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 391

TABLE 5-4 Syndromes Featuring a Neocortical Neuronal Migration Disorder


with Extracerebral Lesions (Continued)
McKusick Other Brain Extracerebral Pattern of
Entity Number Cortical Dysplasia Anomalies Symptoms Inheritance References
Trisomy 9p — Band heterotopia Corpus callosum Enophthalmia, full Chrom (204,205)
agenesis, cerebellar nasal tip, down-
hypoplasia, wide turned mouth,
ventricles brachytelepha-
langy
Trisomy 13 — Heterotopia Holoprosencephaly, Polydactyly, Chrom (206)
microcephaly, cer- scalp defects,
ebellar hypoplasia microphthalmia,
cleft lip/palate
Miller-Dieker 247200 Lissencephaly Type I Corpus callosum Brachycephaly, fur- Chrom (207)
syndrome (del agenesis, midline rowed forehead,
17p13.3) calcifications short upturned
nose, long thick
upper lip
Ring 17 — Heterotopia Wide Vventricles, Broad face, Chrom (208)
peripheral prominent chin,
neuropathy full lips
Trisomy 18 — Heterotopia Corpus callosum Prominent occiput, Chrom (1185,1186)
agenesis micrognathia,
clenched fingers,
cardiac defects
Trisomy 19 mosaic — Heterotopia Microcephaly Growth retardation, Chrom (1187,1188)
hypertelorism, mosaicism
pointed short
nose, small
mouth, limb
anomalies
Trisomy 21 — Heterotopia Microcephaly, Brachycephaly, short Chrom (1189,1190)
Alzheimer plaques stature, cardiac
defects, short
broad hands
Deletion 22q11 — Polymicrogyria, Microcephaly, cer- Cleft palate, Chrom (209–213)
heterotopia ebellar hypoplasia, prominent nose,
cysts dysplastic ears,
cardiac defects
69, XXX — Heterotopia Macrocephaly, wide Growth retarda- Chrom (1191–1193)
ventricles, corpus tion, asymmetry,
callosum agenesis syndactyly
Dysmorphic syndromes
Cornelia de Lange 122470 Heterotopia Microcephaly, cer- Hirsutism, Unknown (214)
syndrome ebellar hypoplasia, synophrys, long
mental retardation philtrum, thin
upper lip, upper
limb deficiencies
(Continued)

Barkovich_Chap05.indd 391 5/6/2011 8:58:58 PM


392 Pediatric Neuroimaging

TABLE 5-4 Syndromes Featuring a Neocortical Neuronal Migration Disorder


with Extracerebral Lesions (Continued)
McKusick Other Brain Extracerebral Pattern of
Entity Number Cortical Dysplasia Anomalies Symptoms Inheritance References
Sotos syndrome 117550 Neuronal heterotopia Large ventricles, Macrocephaly, over- Unknown (215)
prominent trigone; growth syndrome
less often: persis-
tent midline cava,
macrocephaly,
open operculum,
absent corpus cal-
losum
Metabolic disorders
(1) Peroxisomal disorders
Zellweger syndrome 170955 Pachygyria, mainly Wide ventricles, Typical facial dys- AR (1194–1196)
perirolandic and germinolytic morphism, hepa-
occipital; polymi- cysts, delayed tosplenomegaly,
crogyria, mainly myelination, liver dysfunction,
frontal and perisyl- muscle hypotonia, failure to thrive,
vian distribution seizures retinopathy,
on MRI sensory deafness,
± cataract
Neonatal adreno- 202370 Polymicrogyria may Progressive Hepatosplenom- AR (216,217)
leukodystrophy be present leukodystrophy, egaly, failure to
muscle hypotonia, thrive, retinopa-
seizures thy, sensory deaf-
ness, ± cataract
Peroxisomal bifunc- 261515 Polymicrogyria Muscle hypotonia, Mild dysmor- AR (218,219)
tional protein Seizures phism, enlarged
deficiency fontanelles
(2) Mitochondrial disorders
PDHC E1a 312170 Polymicrogyria, Delayed myelination, Mental retardation, X-linked, (1197,1198)
deficiency subependymal atrophy-link gyral spastic prob-
heterotopia changes, olivary quadriparesis ably fatal
dysplasia in male
embryos
Familial respiratory — Polymicrogyria, Leigh syndrome Lactic acidosis AR? (220)
chain disease with leptomeningeal
cerebral calcifica- heterotopia,
tions and neocor- clustered
tical dysplasia heterotopia
Voltage-dependent 604492 Single case with Hydrocephalus Unknown (221)
anion channel perisylvian cortical
deficiency dysplasia
Glutaric aciduria 231680 Leptomeningeal Renal cysts AR (222)
Type II heterotopia
Fumaric aciduria 136850 Polymicrogyria Uncovered sylvian Fumaric aciduria, AR (223,224)
fossae, hypoplasia external dysmor-
of cerebral white phism
matter

(Continued)

Barkovich_Chap05.indd 392 5/6/2011 8:58:58 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 393

TABLE 5-4 Syndromes Featuring a Neocortical Neuronal Migration Disorder


with Extracerebral Lesions (Continued)
McKusick Other Brain Extracerebral Pattern of
Entity Number Cortical Dysplasia Anomalies Symptoms Inheritance References
(3) Organic acidurias
3-hydroxyisobutyric, 236795 Polymicrogyria Cerebral calcifica- External dysmor- AR (225)
acidemia tions, cerebellar phism
hypoplasia
D-2-hydroxy-glutaric Occipital agyria in Delayed gyral and D-2-hydroxyglutaric AR (226)
aciduria minority of cases opercula develop- aciduria, elevated
ment, subependy- GABA in CSF,
mal cysts cardiomyopathy
(4) Disorders of the cholesterol biosynthesis
Smith-Lemli-Opitz 268670 Polymicrogyria (rare, Microcephaly, delayed Failure to thrive, AR (1199–1202)
syndrome not yet confirmed myelination cleft palate, 2/3
in cases with con- syndactyly, heart
firmed biochemical malformations,
abnormality) typical facial
dysmorphism,
increased 7-dehy-
drocholesterol in
serum
Transcriptional regulation
Rett syndrome/ 312750 Nodular heterotopia, Neonatal encephalop- XL-D (227)
MECP2 mutation perisylvian cortical athy with postural
dysplasia abnormality
Genetic brain disruptions
Pseudo-TORCH 251290 Polymicrogyria Micrencephaly, Small for gestational AR (228)
(infection-like) intracranial cal- age, corneal
syndrome cifications, small clouding, microg-
brainstem nathia, throm-
bocytopenia, bile
duct hypoplasia
Early embryonic brain malformations
Encephalocele — Heterotopia Unknown (229)
Holoprosencephaly 236100 Leptomeningeal het- Variable (230,231)
erotopia
Craniotelencephalic 218670 Polymicrogyria, Absent olfactory Trigonocephaly, pan- AR (232)
dysplasia periventricular nerves, anterior craniosynostosis,
heterotopia, focal encephalocele, microphthalamia,
lissencephaly optic nerve hyp- small nose, preau-
oplasia, aqueductal ricular pit
stenosis, cysts
in frontal lobes,
absent septum pel-
lucidum
Meckel-Gruber 249000 Microgyria Posterior encephalo- Sloping forehead, AR (1203–1205)
syndrome cele, molar-tooth polydactyly;
hindbrain mal- polycystic kidneys,
formation, corpus hepatic fibrosis,
callosum agenesis, cleft lip/palate
holoprosencephaly
(Continued)

Barkovich_Chap05.indd 393 5/6/2011 8:58:58 PM


394 Pediatric Neuroimaging

TABLE 5-4 Syndromes Featuring a Neocortical Neuronal Migration Disorder


with Extracerebral Lesions (Continued)
McKusick Other Brain Extracerebral Pattern of
Entity Number Cortical Dysplasia Anomalies Symptoms Inheritance References
Joubert syndrome many Polymicrogyria Hypoplastic cerebellar Polydactyly, Variable (233–235)
and related vermis, Absent SCP retinal dystrophy,
disorders decussation nephronophthisis
Multi-Organ syndrome
(1) Neurocutaneous disorders
Neurofibromatosis 162200 Polymicrogyria, Tumors, macroceph- Café au lait spots, AD (236,237)
Type I heterotopia aly, foci of long T2 Lisch nodules,
on MRI neurofibromas
Linear nevus seba- 601359 Ipsilateral pachygyria, Wide ventricles, por- Linear verrucous AD See Chapter 6
ceous, epidermal glial hyperplasia, encephaly, mental lesions, ichthyosis
nevus hemimegalencephaly retardation hystrix, café-au-
lait spots, heman-
gioma
Tuberous sclerosis 1911100 Heterotopia, hemi- Subependymal nod- Hypopigmentation, AD (238–241)
megalencephaly ules, tumors angiofibroma,
shagreen patches,
renal angioli-
poma, dental pits
Hypomelanosis of Ito 146150 Heterotopia, polymi- Macrocephaly Linear depigmenta- Chrom (242,243)
crogyria tion, asymmetry, mosaicism
cataract
Encephalo-cranio- 176920 Heterotopia Porencephaly, mac- Abnormal pigmen- Unknown (244)
cutaneous rocephaly, wide tation, epibulbar
lipomatosis ventricles, cerebral dermoid, facial
atrophy asymmetry
Oculo-cerebro- 164180 Polymicrogyria, Corpus callosum Orbital cysts, Unknown (245–247)
cutaneous syn- heterotopia agenesis, cerebellar microphthalmia,
drome (Delleman hypoplasia focal skin defects
syndrome)
Incontinentia 308300 Heterotopia, Microcephaly Whorled hyper- XL-D (248,249)
pigmenti polymicrogyria pigmentation,
eye anomalies,
hypodontia
Sturge-Weber 185300 Polymicrogyria asso- Leptomeninges Glaucoma/buphthal- Unknown (250)
syndrome ciated with vascular angiomatosis, mos, vascular skin
impairment perivascular calci- changes in trigem-
fications, cortical inal distribution
necrosis
Klippel-Trénaunay 149000 Hemimegalencephaly Polymicrogyria Cutaneous heman- Unknown (251,252)
syndrome giomas, enlarged
bones
Neu-Laxova 256520 Polymicrogyria, Micrencephaly, Small for gestational AR (95,253,254)
syndrome lissencephaly corpus callosum age, ichthyosis,
agenesis, cerebellar proptosis, hyp-
hypoplasia oplastic eyelids,
short neck,
arthrogryposis
(Continued)

Barkovich_Chap05.indd 394 5/6/2011 8:58:59 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 395

TABLE 5-4 Syndromes Featuring a Neocortical Neuronal Migration Disorder


with Extracerebral Lesions (Continued)
McKusick Other Brain Extracerebral Pattern of
Entity Number Cortical Dysplasia Anomalies Symptoms Inheritance References
(2) Connective tissue disorders
Ehlers-Danlos syn- many Heterotopia Abnormal vascular- Hypermobility, Unknown (255,256)
drome, Many types ization emphysema,
aneurysms Val-
salva
(3) Neuromuscular disorders
Merosin-negative 156225 Occipital agyria; Cerebral white matter Muscular dystrophy AD (257–260)
congenital muscu- leptomeningeal affected
lar dystrophy heterotopia in one
case report
Myotonic dystrophy 160900 Polymicrogyria Macrocephaly, Myotonia, cataract, AD (261)
progressive atro- frontal balding,
phy, peripheral hypogonadism,
neuropathy, white ECG anomalies
matter lesions
Dystroglycanopathies 236670, Cobblestone cortex, Dysmyelination, Retinal dysplasia, AR (257,260,262)
(Walker-Warburg, 253800, subpial heterotopia cerebellar dysplasia, persistent hyper-
Fukuyama CMD, 253280 ventral pontine plastic primary
MEB disease) hypoplasia, col- vitreous
licular fusion
(4) Skeletal dysplasias
Thanatophoric dys- 187600 Polymicrogyria Megalencephaly, Short limb dwarfism, AD (1206,1207)
plasia mesial temporal narrow thorax,
lobe anomalies cloverleaf skull
Fontaine-Farriaus — Periventricular het- Generalized cranio- Unknown (263,264)
craniosynostosis erotopia synostosis, facial
dysmorphism,
brachydactyly and
anonychia
Osteogenesis imper- 166210 Agyria, glial hyper- Perivenous microcal- Fractures, deforma- AD (265)
fecta Type II plasia cifications, white tions
matter lesions
(5) Oculo-cerebral syndromes
Aicardi syndrome 304050 Heterotopia, Corpus callo- Chorioretinopathy, XL (85,266,267)
polymicrogyria sum agenesis, hemivertebrae
glioependymal
cysts, infantile
spasms
MICRO syndrome 600118 Polymicrogyria Corpus callosum Microphthalmia, AR (268–270)
agenesis (incon- cataract, optic
stant) atrophy, motor
neuropathy
(6) Reduction defects
Adams-Oliver 100300 Polymicrogyria Thin ventricular Aplasia cutis con- AD (1208,1209)
syndrome white matter, genita of the skull,
patchy gliosis terminal trans-
verse limb defects,
constriction
(Continued)

Barkovich_Chap05.indd 395 5/6/2011 8:58:59 PM


396 Pediatric Neuroimaging

TABLE 5-4 Syndromes Featuring a Neocortical Neuronal Migration Disorder


with Extracerebral Lesions (Continued)
McKusick Other Brain Extracerebral Pattern of
Entity Number Cortical Dysplasia Anomalies Symptoms Inheritance References
(7) Urogenital abnormalities
Galloway Mowat 251300 Leptomeningeal het- Microcephaly, cer- Nephrotic syndrome AR (1210–1213)
syndrome erotopia, indistinct ebellar granular
cortical layering, atrophy, absence
polymicrogyria, of dentate gyrus,
periventricular myelin deficiency
heterotopia
Oligohydramnios — Abnormal lamination Contractures, facial Variable (271)
sequence (Porter of cerebral cortex, dysmorphism,
syndrome) white matter het- hypoplastic
erotopia, meningeal kidneys
and molecular
zone neuronal-glial
ectopias
(8) OFD syndromes
Oro-facial-digital 311200 Polymicrogyria, Corpus callo- Lobulated tongue, XL-D (272,273)
syndrome I heterotopia sum agenesis, multiple frenula,
wide ventricles, syndactyly
Dandy-Walker
cyst, midline glio-
ependymal cysts
Oro-facial-digital 277170 Polymicrogyria, Molar tooth mid- Hexadactyly, cleft AR (274,275)
syndrome VI heterotopia hindbrain mal- lip/palate, congen-
(Varadi-Papp formation, tuber ital heart disease,
syndrome) cinereum hamar- bifid toes
toma
(9) Other monogenic disorders
Baraitser-Winter 2433310 Classical lissencephaly Microcephaly Trigonocephaly, AD? (201,276,277)
syndrome (prob- ptosis, coloboma,
ably identical hearing loss,
to Ramer-Lin mental retarda-
syndrome) tion; 2p12-q14
inversion in some
patients
X-linked 307000 Polymicrogyria, Corpus callosum Contractures, men- XL-R (278)
hydrocephalus pachygyria agenesis, hypoplas- tal retardation,
tic medullary pyra- spasticity
mids, aqueductal
stenosis
AD, autosomal dominant; AR, autosomal recessive; XL-D, X-linked dominant; XL-R, X-linked recessive; Chrom, chromosomal.
(Adapted from Reference 199).

Barkovich_Chap05.indd 396 5/6/2011 8:58:59 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 397

(b) T2-weighted 3D (volumetric) Fast Spin Echo (3DFSE) volumetric uses 99mTc HMPAO or 123I iodoamphetamine. PET is more sensitive for
acquisition (using 1 mm partition size and multiplanar reformation) or temporal lobe than for extratemporal epilepsy (295). However, the sen-
2 to 3 mm spin-echo or fast spin-echo sequences in two planes (usually sitivity and specificity of 18F-FDG PET for extratemporal lesions can be
axial and coronal), and (c) 3D FLAIR using 1 mm partition size and improved dramatically (50% to >90%) when quantitative analysis with
multiplanar reformation or 2 to 3 mm FLAIR sequences in coronal and a region of interest template (284) or coregistration with MRI (293) is
axial planes in all children with developmental delay or epilepsy. Param- used (Fig. 5-31). 11C flumazenil PET shows changes in GABAA/benzo-
eters (repetition time, echo time, inversion time) should be adjusted to diazepine receptor binding; decreased 11C flumazenil binding has been
optimize contrast between gray matter and white matter; this varies with observed in patients with FCD (285). In one study, abnormalities were
the age of the patient (see Chapter 1). The FLAIR sequence is important, found on 11C flumazenil PET in 72% of patients with partial epilepsy
as subtle white matter abnormalities may give important clues to the and normal conventional MRI studies (296,297); however, these results
presence of the cortical malformation (especially in focal cortical dys- have not been reproduced (297). Another compound, 11C methionine,
plasias (FCDs) [280,281]). The use of parallel imaging, with an 8- or has also been shown to be useful in the detection of small cortical dys-
16-channel coil, allows high signal/noise on thin imaging sections in a plasias by PET (298,299). Increased uptake is seen in the dysplasia. The
clinically reasonable imaging time. Even small/subtle lesions are usually mechanism by which the 11C methionine is concentrated in the area of
detectable using this technique, especially when performed at 3 T. the dysplasia is not known. As with 18FDG PET, 11C methionine PET is
When children have extratemporal partial epilepsies that do not fit nonspecific, and can be concentrated by tumors and abscesses as well
into the classification of benign epilepsies of childhood (282), it is espe- as dysplasias. Ictal SPECT may be sensitive to extratemporal foci of
cially important to search for malformations of cortical development cortical dysplasia, especially in the frontal lobes (289,300,301), but is
(112,282,283). It is essential to know the seizure semiology and the used less as PET becomes more available. The timing of radionuclide
location of any abnormal electrical activity on EEG when interpreting injection is critical in ictal SPECT because variable and evolving pat-
such studies, as the abnormalities can be extremely subtle and are easily terns of perfusion may make localization much more difficult if the
missed if the general area (hemisphere or quadrant) of affected brain isotope is not administered as closely as possible to the time of seizure
is not known. If no MRI abnormality can be identified in spite of this onset. Postictal studies are not reliable in localizing extratemporal foci
knowledge, the cause is usually a small FCD. Diffusion tensor imaging (301). Even better is digital subtraction of ictal and interictal SPECT
(DTI), SPECT, positron emission tomography (PET), and magneto- studies followed by colocalization of the difference image with MRI
encephalography (MEG) may be helpful to find these small and subtle (302,303), which has high interrater reliability and a high rate of local-
lesions; PET, MEG, and SPECT should be coregistered to MRI images ization of the epileptogenic focus. Overall, PET and SPECT are use-
(281,284–294). If MEG is available, it has the advantage of not requir- ful adjunctive examinations but must be correlated with MRI, which
ing radioactive tracers; however, it requires a large team of scientists, remains the most useful neuroimaging exam in the work-up of epi-
technologists, and psychologists, as well as neurologists, neurosur- lepsy (281,293,304).
geons, and neuroradiologists to make it work (292). Subtle anatomic Some evidence has been presented that proton MR spectroscopic
lesions can sometimes be detected in the regions of abnormal electrical imaging may be useful in localization of epileptogenic foci (305–307);
activity (Fig. 5-30) (290) and sometimes the actual focus of ictal onset this technique is most useful in the temporal lobe, particularly in
can be directly identified (291,292). PET studies are usually performed hippocampal sclerosis. Woermann et al. (307) have shown abnormal
with 18F-fluorodeoxyglucose (18F-FDG) or 11C flumazenil, while SPECT metabolite levels in nearly 90% of patients with malformations of

FIG. 5-30. Use of MSI in detecting focal cortical dysplasia. Axial (A) and sagittal (B) images of volumetric
T1-weighted volumetric data set show subtle focal blurring of the cortical white matter junction (black arrows).
This would be impossible to detect without the “spikes” (white triangles), which represent abnormal focal activity
detected by MEG and coregistered to the MR image. Histology after resection showed focal cortical dysplasia.

Barkovich_Chap05.indd 397 5/6/2011 8:58:59 PM


398 Pediatric Neuroimaging

FIG. 5-31. Use of 18FDG PET in imaging of malforma-


tions of cortical development. Note reduced uptake in
the left frontal lobe (black arrows in A) on this PET scan
coregistered with a T1-weighted MR image. MRI veri-
fies the abnormality, showing impaired myelination in
the subcortical white matter (white arrows in B and C).
Histology after resection showed focal cortical dysplasia
Type IIb. PET shows decreased uptake of 18FDG in epi-
leptogenic lesions on interictal studies; it is most useful
when coregistered with MRI.

cortical development; however, the abnormalities were inconsistent, increase in the apparent diffusion coefficient) and decreased anisotropy
with the metabolite levels varying among patients. In addition, the compared with the same area of the brain in age matched controls
spectroscopic abnormalities have been far more widespread than the (294,311,312); these findings may reflect the changes in the brain that
lesion (308,309), suggesting that the MRS findings reflect secondary cause of seizures, may be a result of the seizures, or both. The changes in
changes rather than the primary epileptogenic focus. Li et al. (310) have diffusivity of the white matter may help to determine the extent of the
shown that the NAA/Cr ratio is reduced in malformations secondary dysplasia better than signal alterations on standard imaging and, in this
to abnormal stem cell formation (in which the neurons are dysplastic way, may aid surgical resection (294). Although neither the diffusion
or immature), is variably normal or reduced in heterotopic gray matter changes nor the spectroscopic changes are specific for malformations
(in which neurons are of variable maturity and have variably reduced of cortical development, they may at times serve the same function
synapses), and is normal in PMG (in which the neurons are mature). as PET and SPECT in localizing the region of abnormality such that
This suggests that the low NAA values present in some cortical malfor- reinvestigation with MRI might better locate the lesion.
mations result from dysplastic or immature neurons in the malforma- Overall, MRI remains the cornerstone of the imaging evaluation of
tion; however, these results have not been reproduced. malformations of cortical development (280,313–315). Even if the epi-
Finally, DTI has been used to detect cortical malformations that leptogenic focus is identified by another technique, the focus is always
result in epilepsy. Studies have shown that the white matter underly- coregistered with an MR image before any intervention is planned.
ing the cortical malformation has increased diffusivity (manifest as Therefore, this section deals primarily with MRI.

Barkovich_Chap05.indd 398 5/6/2011 8:58:59 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 399

Malformations Secondary to Abnormal Stem Cell Proliferation of the fetus (Fig. 5-33), as well as on postnatal studies. Although the
or Apoptosis work of classifying microcephalies is in its infancy, ultimately the
Microcephalies Microcephalies are divided into primary micro- imaging analysis of them will depend on these characteristics. It is
cephalies, which usually have a genetic cause and are the result of the hope of the authors that a better system of classifying these dis-
either impaired proliferation of neuronal precursors or impaired orders by imaging characteristics will be developed before the next
apoptosis (316–319), and secondary microcephalies, which result edition of this book.
from late prenatal, perinatal, or postnatal injury (the latter are not
described in this chapter). It is likely that many of the causative genes Microcephaly with Simplified Gyral Pattern and Microlissencephaly.
are involved in DNA repair or cell cycle regulation and, thus, regula- Microcephaly with simplified gyral (MSG) pattern is the term that has
tion of cell proliferation (320); some have already been identified, been proposed to describe malformations in which children are born
while others will likely be found soon, as the interest in this process with head circumferences of 3 or more standard deviations below the
is rapidly increasing (318,319,321–323). norm and their imaging studies show too few gyri and abnormally
Currently, genes have been identified for very few microcepha- shallow sulci (<1/2 the depth of normal sulci) (324,325). This is the
lies and the patterns seen on imaging studies show a wide variation only classification that has been based on imaging characteristics.
(Table 5-5). Using purely morphologic characteristics, microcepha- Primary and secondary sulci are formed but tertiary sulci are not.
lies can be segregated into five major categories: gyral patterns, corti- The volume of white matter in the cerebral hemispheres is reduced.
cal thickness, presence or absence of heterotopia, callosal anomalies, A number of genes have been identified that, when mutated, result
and relative size of the forebrain, hindbrain, and ocular structures. in MSG pattern (see Table 5-5). Unfortunately, simplified gyral pat-
The gyral pattern can be normal, simplified (sulci are too few and terns seem very common in microcephaly; therefore, it is likely that
too shallow but cortical thickness is normal, Fig. 5-32), microgyric simplified sulcation is a more general consequence of diminished
(Fig. 5-33), or pachygyric (sulci are too few and too shallow with cell proliferation (and consequent diminished white matter volume)
thick cortex, Fig. 5-34). Heterotopia may be present (Figs. 5-32 and and not one that will allow segregation of specific genetic disorders.
5-35) or absent. The corpus callosum can be absent, hypoplastic, or However, it is likely that microcephalic patients with thickened cor-
hypogenetic (see earlier section on callosal anomalies and Figs. 5-34 tex in addition to simplified sulcation (a malformation that we call
and 5-35). The forebrain can be large compared with the hindbrain microlissencephaly) have different pathoembryogenesis and clearly
(Fig. 5-34), commensurate in size with the hindbrain, or small com- different disorders. These patients are separated into Group 6, while
pared with the hindbrain (Fig. 5-33). Similarly, the ocular globes can patients with MSG are in the other groups and can be separated by
be commensurate with cerebral size or large (Fig. 5-35). It is important differences in associated anomalies of the brain or other viscera. MSG
to note that most of these characteristics can be identified on MRIs and microlissencephaly are both heterogeneous groups, but affected

TABLE 5-5 Genetic Causes of Microcephaly with Known Imaging Findings


Gene Chromosome OMIM No. Protein Inheritance Description
MCPH1 8p23 607117 Microcephalin AR Simplified gyral pattern
Microcephaly, PVNH
Good clinical function
ASPM 1q31 608716 Spindle-like microcephaly protein AR Simplified gyral pattern
Microcephaly, PVNH, HCC
ARFGEF2 20q13.13 608097 ARFGEF2 AR Mild microcephaly
Notable PVNH
Callosal hypoplasia
Myelin delay
CDK5RAP2 9q34 608201 CDK5 regulatory subunit-associated protein 2 AR Simplified gyral pattern
CENPJ 13q12.2 609279 Centromeric protein J AR Simplified gyral pattern
SLC25A19 17q25.3 607196 Mitochondrial deoxy-nucleotide carrier AR Simplified sulcation, early death,
2-ketoglutaric aciduria
EIF2AK3 2p12 226980 eIF2 akinase 3 AR Simplified sulcation in
Wolcott-Ralston Syndrome
PNKP 19q13.33 605610 Polynucleotide kinase 3’ phosphatase AR Normal sulcation.
Proportional cerebrum, cerebel-
lum. Decreased WMV
AR, autosomal recessive; PVNH, periventricular nodular heterotopia; HCC, hypogenesis of the corpus callosum; WMV, white matter volume.

Barkovich_Chap05.indd 399 5/6/2011 8:59:01 PM


400 Pediatric Neuroimaging

FIG. 5-32. Microcephaly with simplified gyral pattern,


moderate and severe. A and B. Moderately simplified gyral
pattern. Sagittal T1- (A) and axial T2- (B) weighted images
show too few gyri, shallow sulci (about 1/2 of normal), nor-
mal myelination, and a normal thickness of cerebral cortex.
C–E. Extremely simplified gyral pattern with hypoplastic
corpus callosum, proportional cerebellum, and hetero-
topic gray matter. Sagittal T1-weighted image (C) shows
extreme microcephaly with a very thin corpus callosum
(white arrows). Cerebellum and brainstem are proportion-
ate to the cerebrum. Axial T2-weighted image (D) shows an
extremely simplified gyral pattern with too few, too shallow
sulci. Coronal T1-weighted image (E) shows periventricu-
lar nodular heterotopia (white arrows) protruding into the
ventricular trigones.

Barkovich_Chap05.indd 400 5/6/2011 8:59:01 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 401

FIG. 5-33. Microcephaly with polymicrogyria in a 23-week fetus with biparietal diameter more than 3 standard deviations
below mean for gestational age. Axial (A) and coronal (B) SSFSE images show extensive polymicrogyria of the cerebrum (the
cortical surface should be smooth at this age, see Chapter 2). Note that the cerebellum is much larger than the cerebrum on
the coronal image (B).

patients in both are almost certainly microcephalic because of reduced normal myelination, and a normal-appearing cerebral cortex (normal
proliferation of neurons or glia. cortical thickness and normal appearing cortical-white matter junction,
Fig. 5-32A and B). Almost all patients in this group show progressively
Group 1. Group 1 includes patients who are products of normal preg- more severe developmental delay as they grow and reach school age.
nancies, have normal deliveries, normal neonatal courses, and normal One cause of MSG Group 1 appears to be the Nijmegen breakage
neonatal exams, notable only for their very small head sizes. Poor feeding syndrome, an autosomal recessive disorder belonging to the family of
and poor weight gain may characterize the neonatal courses. All manifest DNA repair disorders (326). Affected patients also have immunodefi-
corticospinal tract signs during the first year of life, ranging in severity ciency, X-ray hypersensitivity, and predisposition to cancer.
from mild spasticity to multiple beats of clonus and bilateral Babinski
signs. The imaging studies of all patients in this group show a simpli- Group 2. The patients in group 2 are commonly products of breech
fied gyral pattern with too few gyri, shallow sulci (about 1/2 of normal), presentations and manifest abnormal neonatal reflexes and limb

FIG. 5-34. Microcephaly with absent corpus callosum, frontal pachygyria, and pontocerebellar hypoplasia. Sagittal T2-weighted
image (A) shows callosal agenesis and small pons and cerebellum compared with cerebrum. Axial T2-weighted image (B) shows
pachygyria of both frontal lobes (black arrows). Note the abnormal enlargement and shape of the lateral ventricles.

Barkovich_Chap05.indd 401 5/6/2011 8:59:02 PM


402 Pediatric Neuroimaging

FIG. 5-35. Microcephaly with thin corpus callosum, dis-


proportionately large eyes, and slightly disproportionately
large cerebellum. A. Sagittal T1-weighted image shows
microcephaly with very thin corpus callosum; the brainstem
and cerebellum are slightly large compared with cerebrum.
B. Axial T2-weighted image shows disproportionately
large ocular globes; they are as big as basal ganglia and
thalami combined. Gyral pattern is simplified. C. Coronal
T2-weighted image shows the cerebellum to be dispropor-
tionately enlarged compared with cerebrum. Heterotopia
(white arrows) protrude into the ventricular trigones.

spasticity that begin in the neonatal period. Affected patients have atresia, hypertonicity, cortical thumbs, microphthalmia, optokinetic
difficulty suckling and recurrent vomiting and, therefore, manifest poor nystagmus, and abnormal neonatal reflexes, and neonatal seizures.
weight gain. They develop generalized tonic seizures during the first These patients are sometimes described as having the “apple peel syn-
few days of life. MRI shows a very simplified gyral pattern with too few drome” (OMIM 243605) (329–331). MR scans are identical to group 1
gyri, shallow sulci (about 1/3 of normal), and delayed myelination. The (Fig. 5-32A and B), showing too few gyri, shallow sulci, and age-appro-
cerebral cortical thickness and gray-white junction appear normal. priate myelination; the cerebral cortex has a normal appearance.

Group 3. Patients in group 3 are typically born at term after uncom- Group 5. Patients in group 5 are profoundly microcephalic (4–5 stan-
plicated deliveries. They have abnormal neonatal courses, manifest- dard deviations below normal) and profoundly abnormal from the
ing absence of neonatal reflexes, spasticity, seizures, and poor feeding. time of birth. All are profoundly hypotonic, have abnormal neonatal
MR scans in these patients show fewer gyri and shallower sulci than reflexes, and develop myoclonic seizures during the first week of life.
groups’ 1 and 2, as well as periventricular nodular heterotopia (PVNH) Suck and swallow are weak, requiring gastrostomy for feeding. All show
(Fig. 5-32C–E); they may have arachnoid cysts, and have normal minimal neonatal development. MRI of these patients show large sub-
myelination. MCPH1 and ASPM mutations may belong in this group arachnoid spaces surrounding extremely small brains with impaired
(322,327,328). Cortical thickness and gray matter-white matter junc- myelination, reduced volume of white matter, and very simplified
tion appear normal. gyral patterns with no more than about 5 gyri in each hemisphere and
sulci less than one-third the depth of normal. The cerebral cortex may
Group 4. Patients in group 4 have significant prenatal or neonatal appear abnormally thin. Patients are severely debilitated and die within
problems, such as polyhydramnios, congenital arthrogryposis, jejunal the first months of life.

Barkovich_Chap05.indd 402 5/6/2011 8:59:04 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 403

Group 6. Patients in group 6 have findings of complete or nearly Chapter 11), ischemia, inborn errors of metabolism, and prenatal
complete agyria, with thickened cortex (Fig. 5-36). Many have asso- exposure to toxins or radiation, before classifying patients as having
ciated agenesis or hypogenesis of the corpus callosum and cerebellar microlissencephaly.
hypoplasia. Some have PVNH. Most are hypotonic at birth, develop
seizures in the first week of life, and achieve few developmental mile- Focal Cortical Dysplasia with Dysplastic Neurons (Focal Trans-
stones. One disorder in this group, sometimes called Amish lethal mantle Cortical Dysplasia, Focal Cortical Dysplasia Type II)
microcephaly, is caused by mutations of the SLC25A19 gene at 17q25.3, First described by Taylor et al. (338) in 1971, FCDs are the single most
which encodes a nuclear mitochondrial deoxynucleotide carrier (332). important cause of medically refractory focal epilepsy in childhood.
Affected patients are profoundly microcephalic at birth with moderate FCD is the third most common histologic diagnosis (after hippocam-
micrognathia. Neurologic exam is mostly normal at birth, but feeding pal sclerosis and epilepsy-associated tumors) in children who undergo
becomes more difficult after the first 2 months and irritability increases epilepsy surgery, found nearly 15% to 20% of children who have
markedly; deterioration increases until death, usually during the first surgery for intractable epilepsy (280,339–341); the majority of these
year (332). Imaging shows profoundly small, smooth cerebrum and are FCD Type II (121). FCDs appear to be mainly sporadic, with few
cerebellum with small brainstem. The corpus callosum is typically familial cases reported. The wide spectrum of these disorders has only
hypogenetic. A few PVNH may be present. recently begun to be recognized. Although previous classifications of
FCDs have been proposed (342), they suffered from poor interobserver
Group 7. Group 7, also called leukodysplasia, microcephaly, and and intraobserver reproducibility among pathologists (343) and have,
cerebral malformation, has been recently described and is linked to therefore, been revised. They are currently divided into four groups
chromosome 2p16 (333). Affected patients are severely microcephalic (344). Mild malformations of cortical development (mMCD) are char-
(head circumference 3 or more standard deviations below normal) at acterized by normal cortical architecture and abundant ectopic neurons
birth, show severe failure of postnatal brain growth, develop intrac- located in or adjacent to cortical layer 1 or in the subcortical white mat-
table seizures during the neonatal period, do not show developmental ter. These are currently believed to result from late prenatal, perinatal,
progression, and die within the first 3 years of life (333). Imaging shows or early postnatal (as yet undefined) processes. Type 1 FCDs are associ-
a cortex of normal thickness, simplified gyral pattern, lack of opercula- ated with architectural disturbances of the radial arrangement (FCD
tion, markedly reduced volume of white matter with callosal hypopla- Type Ia) or tangential arrangement (FCD Type Ib) of cortical neurons
sia and ventriculomegaly, pontine hypoplasia, and cystic lesions in the or both (FCD Type Ic); these are also suspected to result from late pre-
subcortical white matter of the temporal lobes (Fig. 5-37) (333). natal, perinatal, or early postnatal disturbances. When changes of FCD
Many of the patients with MSG were formerly classified as having Type I are associated with other pathology, they are diagnosed as FCD
“microcephalia vera” (334,335), or “radial microbrain” (190,336,337). Type III (IIIa with hippocampal sclerosis, IIIb with glial or glioneu-
However, the exact meaning of those terms was rather murky and ronal tumors, IIIc with vascular malformation, IIId with scarring from
they were defined differently in different publications. This classifi- traumatic injury, porencephaly, or infection). Type II FCDs have more
cation better organizes these profoundly microcephalic patients and pronounced architectural and cytoarchitectural disturbances such as
helps in understanding the underlying developmental abnormalities dysmorphic neurons (FCD Type IIa) and balloon cells (FCD Type IIb)
(324). As the causes and prognoses of secondary microcephalies are (344); they are also characterized by overabundance of GABAergic
quite different, it is important to rule out known causes of profound neurons (personal communication, Dr. Roberto Spreafico). Type II
microcephaly, such as prenatal infection (TORCH infections, see FCD is thought to be due to earlier, prenatal, disturbances to cortical

FIG. 5-36. Microlissencephaly. Sagittal (A) and axial (B) images show a profoundly microcephalic brain (compare size of brain to size of face) that is
completely smooth, with a thick cortex. The corpus callosum is hypogenetic (small splenium, absent inferior genu and rostrum). Microlissencephaly is
probably better classified as a lissencephaly rather than a type of microcephaly.

Barkovich_Chap05.indd 403 5/6/2011 8:59:04 PM


404 Pediatric Neuroimaging

FIG. 5-37. Leukodysplasia, microcephaly, and cerebral malformation (Micro-


cephaly with simplified gyral pattern group 7). Sagittal T1-weighted image
(A) shows thin corpus callosum (white arrows) and small cerebellum. Axial
T2-weighted images (B and C) show simplified gyral pattern with normal corti-
cal thickness, very large subarachnoid spaces, reduced white matter, ventriculom-
egaly, and lack of operculation.

development, possibly genetic (7,280). Because they are thought to Almost all affected patients have epilepsy, which typically becomes
arise from an early disturbance, Type II FCDs are discussed in this sec- clinically apparent during the first decade of life, sometimes as early
tion. mMCD and FCD Types I and III are discussed in a later section, as the first few days (120,280,315,345). Compared with FCD Type I
“Malformations Secondary to Abnormal Cortical Late Migration and and Type III, patients with Type II FCD come to medical attention
Cortical Organization.” at younger ages, have higher seizure frequencies, and are much more
FCD Type II is an important cause of epilepsy, both in children commonly extratemporal in location; seizure outcome after surgery is
and in adults (7,280,315,340,342). This malformation is referred to by generally better (280,315). The seizure type seems to vary with patient
many names, including Taylor type of FCD, focal transmantle corti- age (120). The epilepsy of most patients is highly refractory to medica-
cal dysplasia (345), and forme fruste tuberous sclerosis (because many tion secondary to the high intrinsic epileptogenicity of these lesions
FCD II lesions are histologically and radiologically identical to corti- (117,351), which seems to be related primarily to the dysplastic, large
cal hamartomas of patients with tuberous sclerosis (68,346) and some neurons within the lesions (339). Therefore, many of these patients
show a loss of heterozygosity at the TSC1 [347] or the TSC2 gene locus are helped by surgical resection of the dysplasia if the epileptogenic
[348,349]). The term FCD Type II is preferred and should be used. As focus can be found. The presence or absence of other neurologic signs
with tuberous sclerosis, studies of molecular markers of the dysplastic and symptoms of affected patients depends upon the size and location
neurons and balloon cells suggest that FCD Type II derive from RGCs of the malformation. When larger regions of cortex or eloquent areas
(350). Because of the overlap with tuberous sclerosis, patients with are involved, patients may have neurologic signs/symptoms. When
radiologic or histologic findings of this malformation should be seen only small regions of cortex are involved, as is often the case, affected
by a geneticist or child neurologist in order to be screened (skin, kid- patients may have normal neurologic examinations (280,345). EEG is
neys, heart, etc.) for tuberous sclerosis (346). characterized by total absence of background activity and a distinctive

Barkovich_Chap05.indd 404 5/6/2011 8:59:05 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 405

pattern of repetitive, high amplitude, fast spikes followed by high histologic diagnosis of these disorders (343). Recent work suggests that
amplitude slow waves interspersed with relatively flat periods. MRI findings in FCD Type II are characteristic (no consistent differ-
On histologic exam, the normal six-layered appearance of the ence has been found yet between Type IIa and Type IIb, so both are
cerebral cortex is disturbed. In addition, abnormal cells are seen in the discussed together) (280,345,359). When the involved region of brain
cerebral cortex and in the underlying white matter; these cells typically is large, the cortical gyral pattern may be abnormal, with expanded gyri
include large neurons, dysplastic neurons, atypical glia, and balloon and, occasionally, irregular sulci (Fig. 5-38). Abnormal signal intensity
cells, admixed with normal neurons (342,344,352,353). Glial prolifera- (typically hyperintense on FLAIR and T2-weighted images) is seen in
tion and reduced myelination are seen in the affected white matter, and subcortical white matter; the cortical-white matter junction typically
may contribute to the T2 prolongation that is nearly always seen in the appears sharp on both T1- and T2-weighted images (Fig. 5-31), but is
subcortical white matter in FCD IIb (352,354). The affected regions of often indistinct on FLAIR images. In some patients, a linear or curvilin-
cortex have an increased number of excitatory neurons and a decreased ear focus of abnormal signal intensity extends from the cortical-white
number of inhibitory neurons, along with impaired GABA-mediated matter junction to the ventricular surface (Fig. 5-39); this is referred to
neuronal inhibition, probably explaining the intrinsic epileptogenicity as the transmantle sign and appears to be specific for FCD Type II when
of the affected cortex (355–358). Developmental lineage studies sug- seen (280,315). The signal intensity of this focus varies with the age of
gest that the abnormal cells derive from the neocortical (dorsal) ven- the patient. In neonates and young infants, it is bright on T1-weighted
tricular and SVZs, possibly from RGCs, and are likely glutamatergic images and somewhat dark on T2-weighted images (Fig. 5-40); this
(excitatory transmitters) (350). may be caused by intrinsically abnormal tissue or stimulation of myeli-
Publications describing neuroimaging studies of patients FCD nation by the electrical activity of the seizures (360). Between the ages
Types I and II are confusing, largely because there is inconsistency in of about 12 and 30 months, while the brain is actively myelinating,

FIG. 5-38. Large to medium FCD IIb. Sagittal T1 (A), axial FLAIR
(B), and coronal T2 (C) images show a large cortical and subcor-
tical lesion (white arrows) in the orbital surface of the left frontal
lobe and underlying white matter. The cortex is normal thickness
or thin, with blurring of parts of the cortical-white matter junction.
The affected white matter (white arrows) has transmantle extension
(from pia to ependyma) and shows hypointense T1 and hyperin-
tense T2/FLAIR signal, reflecting hypomyelination.

Barkovich_Chap05.indd 405 5/6/2011 8:59:06 PM


406 Pediatric Neuroimaging

FIG. 5-39. Axial (A) and coronal (B) T2-weighted images in a child with medially refractory partial epilepsy show a hyperin-
tense “transmantle sign” (white arrows) extending from the bottom of a right frontal sulcus to the superolateral margin of the
lateral ventricle. This is a specific sign for FCD IIb.

dysplasias may be difficult to detect by MRI using any standard long-standing dysplasias may erode the inner table of the calvarium
sequences; therefore, if EEG studies continue to show focal epilepto- (Fig. 5-41) like cortical tubers of tuberous sclerosis (see Chapter 6).
genesis, the T1 hyperintensity of myelinated white matter should be On occasion, T2 hyperintensity is seen extending for several millime-
suppressed with a magnetization transfer pulse (361) or a repeat MRI ters along the cortical-white matter junction on one or both sides of
should be performed after age 3 years. After the white matter myeli- the tract from the cortex to the ventricle; this may be caused by the
nates, the tract is seen as hyperintensity on T2-weighted and, especially, malformation but is more likely caused by the seizures it produces.
FLAIR images, particularly when the image is parallel to the track (Fig. The cortical-white matter junction may be blurred on high resolution
5-39) (362). This track of T2 hyperintensity correlates well with the T1-weighted images (Fig. 5-41), presumably due to incomplete myeli-
location of the balloon cells (362,363), but the abnormal signal is more nation or astrogliosis in the subcortical white matter. The acquisition
likely due to gliosis and hypomyelination of the white matter. The of thin section, high resolution images is extremely useful in detecting
tract is difficult to see on T1-weighted images unless a magnetization the smaller, subtler foci (345,364–366).
transfer pulse is used to suppress the hyperintensity of the white mat- A group of patients with calcified FCD Type II lesions have been
ter (361). Some of the affected cortex may also be T2 hyperintense and reported (349). These look like calcified cortical tubers of tuberous

FIG. 5-40. FCD IIb in young infant. Coronal T1- (A) and T2- (B) weighted images show abnormal signal (white arrows) radiating
to the superolateral margin of the lateral ventricle from the depth of the affected sulcus. Note that in the unmyelinated brain, the
abnormal signal is hyperintense on T1-weighted images and hypointense on T2-weighted images.

Barkovich_Chap05.indd 406 5/6/2011 8:59:07 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 407

FIG. 5-41. FCD II resembling a tuber of tuberous sclerosis. Axial T1 (A)


and FLAIR (B) images and coronal T2 image (C) shows a focally expanded
gyrus (arrows) in the left temporal lobe, causing remodeling of the overlying
calvarium. The dysplasia (cortex and underlying white matter) is hypoin-
tense in (A) and hyperintense in (B) and (C). The cortical-white matter
junction is more indistinct in (A) and (B) than in (C).

sclerosis (see Chapter 6), with subcortical calcification (very hyperatten- proliferation, migration, and organization within the affected hemi-
uating in CT, hypointense on T1- and T2-weighted MR images) and vol- sphere (368–372). The many different radiologic (369,370,373) and
ume loss, resulting in enlarged subarachnoid spaces around the lesion. pathologic (371,374,375) appearances suggest that HME is a heteroge-
Our experience with diffusion imaging in FCD Type II is similar neous malformation, with several different disorders (likely with sev-
to that in the literature (294). The white matter under the lesion has eral causes) included under a single heading. The brain can be affected
increased diffusivity and decreased fractional anisotropy. It is not clear in isolation, in association with cutaneous abnormalities (about 1/3
how much of this effect is the result of the lesion itself and how much is [376]), or with hemihypertrophy of part or all of the ipsilateral half
the result of recurrent seizure activity and its effect on the tissue. of the body; overall, about half of affected patients have other associ-
The few reports of proton MRS in transmantle dysplasias suggest ated anomalies (377). Rarely, involvement of the cerebral hemisphere is
that there is a significant reduction of the NAA/Cr ratio compared with associated with enlargement and dysplasia of the ipsilateral cerebellar
normal controls and with normal areas in the affected brain (305,310). hemisphere and brainstem, a condition referred to as total HME (378).
The choline peak is normal to slightly increased. On short echo spec- All cases seem to be sporadic and, of interest, only one pair of monozy-
tra (TE = 20–30 milliseconds), myo-inositol is increased. Our limited gotic twins with HME has been reported (379). Affected children typi-
experience with perfusion imaging suggests that these lesions have cally have macrocephaly at birth and in early infancy; this may be the
normal or reduced blood volume compared to normal white matter. reason for initial referral for imaging, even though affected patients do
not have clinical signs of increased intracranial pressure. More com-
Hemimegalencephaly (Unilateral Megalencephaly) HME, first monly, affected infants come to attention because of an intractable sei-
described in 1835 (367), is the name given to a hamartomatous over- zure disorder that begins at a very early age (usually before the first
growth of all or part of a cerebral hemisphere with defects in neuronal birthday), hemiplegia, and severe developmental delay (369–371,374).

Barkovich_Chap05.indd 407 5/6/2011 8:59:08 PM


408 Pediatric Neuroimaging

Earlier onset of epilepsy seems to correlate with more severe motor quite characteristic; it is enlarged, usually in proportion to the enlarge-
impairment (376). Seizures tend to be more severe when the mega- ment of the affected hemisphere, with the frontal horn being abnor-
lencephaly is localized to the frontal lobe (380). A high incidence of mally straight and pointing superiorly and anteriorly (Figs. 5-43 and
HME seems to occur in patients with the epidermal nevus syndrome 5-44) (369). In occasional patients, however, the ipsilateral ventricle
(see Chapter 6 [376,381–383]). Other associated conditions include is small (373). Rarely, the affected portion of the brain has a bizarre,
Proteus syndrome (sometimes considered a form of epidermal nevus hamartomatous appearance (Fig. 5-45) (369,370); in this situation, the
syndrome [384]) (385,386), unilateral hypomelanosis of Ito (376,387), characteristic enlargement of the affected brain and ipsilateral ventricle
neurofibromatosis Type I (388), Klippel-Trenaunay-Weber syndrome allows diagnosis. In a minority of cases, the cerebellar hemisphere and
(251), and tuberous sclerosis (241,389). No difference has been found brainstem ipsilateral to the affected cerebral hemisphere are enlarged
between the syndromic and nonsyndromic types of HME (378). (Fig. 5-44) (378).
Pathologically, HME is a disorder reflecting abnormalities in pro- It is important to recognize that the radiologic appearance of
liferation, migration, maturation, and differentiation of neurons (375); the HME brain may change over time. Wolpert et al. (391) reported
thus, many suspect this disorder to be the result of abnormal stem a patient in whom the affected hemisphere atrophied during the first
cell lineage, differentiation, and proliferation (7,372). Affected hemi- year of life and was smaller than the contralateral (normal) hemi-
spheres contain areas of pachygyria, PMG, and heterotopia, as well as sphere when imaged at age 1 year. An analogous change was seen on
dyslamination, dysplastic neurons, immature neurons, balloon cells, SPECT imaging, where the tracer uptake in the affected hemisphere
hypomyelination, and astrogliosis of the hemispheric white matter diminished over time (392). We have seen reduction in hemispheric
(371,374,375,379). Interestingly, studies show an increased number size of affected patients after episodes of status epilepticus. Salamon
of glial cells on the affected side compared to the contralateral side et al. (379) reported that the “normal” hemisphere is often smaller than
(390); it is not known whether this increase is developmental or reac- age-matched controls and suggest that it is not normal at all; indeed,
tive. Reports conflict on the number of cortical neurons in the affected they report that children with small “normal” hemispheres have poorer
hemisphere (379,390). neurodevelopmental outcomes after hemispherectomy.
On CT and MRI, part or all of a hemisphere may be affected; the Proton MR spectroscopy in HME shows reduction of the gluta-
frontal and occipital lobes are involved in nearly equal frequency in mate and NAA peaks in white matter. Reduced NAA has also been
localized megalencephaly (Fig. 5-42) (380). The involved region may reported in cortical tubers of tuberous sclerosis (see Chapter 6) and in
be moderately to markedly enlarged. Although the appearance of the cortical dysplasias with balloon cells (305,310) and probably relates to
large hemisphere varies considerably, the most typical appearance the immaturity of the involved neurons.
shows the cortex to have an abnormal gyral pattern with broad gyri, The affected portion of the hemisphere has reduced metabolic
shallow sulci, blurring of the cortical-white matter junction, and cor- activity, and 18FDG PET scans show markedly reduced glucose uptake
tical thickening (Fig. 5-43); however, the gyral pattern may appear in affected regions (Fig. 5-43) (393). Anatomic hemispherectomy,
grossly normal or may be frankly agyric or polymicrogyric (Fig. 5-44) functional hemispherectomy, or hemidecortication may, therefore,
(369,370). The white matter is of abnormally low signal on CT and be indicated if the seizure disorder is intractable and the contralateral
often shows heterogeneous signal intensity (some T2 hyperintense hemisphere is normal (394,395). However, hemispherectomy is con-
and some isointense to myelinated white matter) on MR studies, rep- traindicated if the contralateral hemisphere has cortical malforma-
resenting heterotopia and dysplastic neurons and glia (Figs. 5-43 and tions; therefore, careful assessment of the contralateral hemisphere is
5-44). The configuration of the ipsilateral lateral ventricle is usually crucial in affected patients.

FIG. 5-42. Localized megalencephaly. Axial (A) and coronal (B) T1-weighted images show enlargement of the left
occipital lobe (black arrows) with broad gyri, shallow sulci, and poor differentiation of cortex and underlying white mat-
ter. The coronal image better shows abnormal white matter signal intensity and poor cortical white matter differentiation
in the megalencephalic region (arrows).

Barkovich_Chap05.indd 408 5/6/2011 8:59:09 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 409

FIG. 5-43. Typical hemimegalencephaly. Axial T2-weighted (A) and FLAIR (B) images in an epileptic teenager show an
enlarged left hemisphere with fewer, broader gyri than the normal right hemisphere, and abnormally hyperintense white
matter. The cortical-white matter junction is blurred, particularly in the posterior aspect of the hemisphere (white arrows).
Coronal T2-weighted image (C) shows uplifting of the left frontal horn (white arrows) as well as abnormal white matter and
blurring of the cortical-white matter junction in the medial left convexity. Axial image from a PET study (D) shows reduced
uptake of 18F-FDG in the affected left hemisphere, most notably in the posterior hemisphere (white arrows).

Malformations Secondary to Abnormal Neuronal Migration to abnormal neuronal migration are those due to overmigration of
Malformations with Too Few or Too Shallow Sulci Smooth brains neurons resulting from abnormal pial basement membrane formation
can result from many causes, including congenital infection (especially (cobblestone malformations) and those due to diffuse arrest of neu-
cytomegalovirus, see Chapter 11) and diminished cell proliferation ronal migration (lissencephalies).
(“Microcephaly with Simplified Gyral Pattern and Microlissencephaly,”
see previous section), in addition to abnormal neuronal migration. Malformations Due to Abnormal Pial Basement Membrane
Careful attention to factors such as brain size (more than 3 standard Formation
deviations below normal at birth suggests MSG pattern), cortical thick- Background: Why do we find CNS abnormalities in patients with
ness (diminished thickness should suggest in utero infection, dimin- Congenital Muscular Dystrophies? Malformations secondary to
ished cell proliferation), and smoothness of the cortical-white matter abnormal pial basement membrane formation are a group of disor-
junction (irregularity of the junction indicates PMG from mutation ders that are typically characterized by anomalies of the brain, eyes,
or midgestational injury) are essential to proper classification. The and muscles. They were initially discovered as a result of the observa-
most important malformations with too few or too shallow sulci due tion that many children with congenital muscular dystrophies (CMD)

Barkovich_Chap05.indd 409 5/6/2011 8:59:10 PM


410 Pediatric Neuroimaging

FIG. 5-44. Hemimegalencephaly with more severe anomaly. Axial T2-weighted images (A and B) show polymi-
crogyria (black arrows) in the left frontal lobe and subcortical heterotopia (white arrows in B) in the white matter
more posteriorly. The ipsilateral ventricle (v) is large. Axial diffusivity image (C) shows minimally reduced diffu-
sivity in the affected hemisphere. Coronal T2-weighted image (D) shows enlarged, uplifted lateral ventricle (black
arrows), enlarged ipsilateral cerebellar hemisphere (white arrows), and abnormally hypointense white matter.

had associated anomalies of the brain and ocular globes; the first such understood. It seems that abnormalities of molecules involved in these
diseases described were Fukuyama congenital muscular dystrophy linkages result in a lack of connection of the RGCs to the pial base-
(FCMD) (396,397), muscle-eye-brain (MEB) disease (398,399), and ment membrane in addition to the development of abnormal gaps in
the Walker-Warburg syndrome (WWS) (400,401). It was subsequently that membrane. These defects result in a lack of normal detachment of
discovered that the muscular abnormalities in these patients were the migrating glutamatergic neurons from RGCs and, consequently, lack
result of abnormal linkage of the muscle fibers to the muscle basal of normal laminar organization of the cortex; some neurons detach
lamina; without proper linkage, the muscles were unable to contract early and remain in the subcortical white matter while others overmi-
properly (402–404). Subsequent studies showed that similar linkages grate through the gaps in the pial limiting membrane into the subpial
are important in the development of the cerebral and cerebellar corti- and subarachnoid spaces (402–405). Once the molecules involved in
ces (where the same molecules link RGCs to the basement membrane the linkage mechanisms were discovered, neuroscientists began to look
of the developing cortex), as well as the retina (linkage of glial guide for mutations of genes encoding these molecules; many such muta-
cells to the retinal limiting membrane) (402–404). The molecular tions were found in patients with this spectrum of disease. Responsible
mechanisms shared by these different linkages were the “missing link” genes discovered so far in humans include POMT1, POMT2, FCMD,
in the connection between muscle, eye, and brain disease. The pre- FKRP, LARGE, POMGnT1, and GPR56 (125,405–412). Zic2 and Foxc1
cise mechanisms that are impaired in these processes are not yet fully have been implicated in such defects in animal models (413,414).

Barkovich_Chap05.indd 410 5/6/2011 8:59:11 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 411

FIG. 5-45. Hemimegalencephaly with bizarre, hamartomatous enlargement of the left hemisphere. The cortex is thick and
convoluted. Myelination appears abnormally advanced.

Mutations of genes coding for other proteins involved in the linkage of in the geographic distribution of mutations; POMT1 appears to be
the radial glia to the limiting membranes include laminin and merosin more commonly mutated in Middle Eastern WWP populations, while
(a form of laminin), mutations of which cause merosin-deficient FCMD mutations seem to be common in European and American
CMD (see Chapter 3) (415–421); CMD with leukoencephalopathy and WWP patients, particularly in Ashkenazi Jewish families (125).
occipital agyria (258,422,423); CMD with severe mental retardation, On imaging studies, patients with WWP have a thickened cortex
microcephaly, abnormal white matter, partial merosin deficiency, and with only a few shallow sulci and a distinctive cortical appearance,
cerebellar hypoplasia (424); and CMD with partial merosin deficiency, ocular anomalies (which may be unilateral or bilateral), hydrocepha-
mental retardation, dysmyelination, and cerebellar cysts (425). lus, callosal hypoplasia, and profound hypomyelination (Fig. 5-46)
A number of studies have shown that several of the four major (260,431). An occipital cephalocele is present in about 10% (Fig. 5-47).
phenotypes of congenital muscular dystrophy with malformations of The cerebral cortex (Fig. 5-46) is composed of multiple small bundles
cortical development—WWS, FCMD, MEB, or limb-girdle muscular of disorganized cortical neurons that project through the pia on the
dystrophy (which has no CNS involvement)—can be produced by outer cortical surface and into the underlying white matter on the
mutations of any of the genes involved in linkage of the RGCs to the inner cortical margin (190); this appearance has been called a “cobble-
pial basement membrane (406–411,426) with those mutations having stone” cortex (432). Small nodules of gray matter may be seen a few
the most severe effect on protein function causing WWS and those millimeters deep to the projections from the inner cortical margin (Fig.
with the least severe effect causing classic FCMD or limb-girdle muscu- 5-46). The bundles of neurons are separated by fibroglial vascular tis-
lar dystrophy. This should be kept in mind as these major phenotypes sue that extends radially from the cerebral white matter through the
and their imaging findings are described below. cerebral cortex into the subarachnoid space (which is obliterated by
this tissue, resulting in hydrocephalus). Few, if any, normal sulci are
Walker-Warburg Phenotype The Walker-Warburg phenotype seen in the cortex. Characteristics of the cortex may be difficult to
(WWP) is the most severe in this group of malformations. Affected appreciate in the hydrocephalic neonate, whose cortex is compressed
patients are obviously abnormal at birth, manifesting hypotonia that by the enlarged ventricular system (Fig. 5-48); therefore, careful obser-
is usually profound and unchanging, in addition to ocular abnormali- vation is required to make the diagnosis before the ventricles have been
ties (retinal dysplasia, colobomas, persistent hypoplastic primary vitre- decompressed. Characteristic brainstem anomalies include pontine
ous, congenital glaucoma or microphthalmos, optic nerve hypoplasia hypoplasia, an enlarged quadrigeminal plate with fusion of the supe-
[427]) and progressive macrocephaly due to congenital hydrocepha- rior and inferior colliculi, and a distinctive “kink” in the dorsal pons;
lus; they may have posterior cephaloceles and testicular defects as well a second “kink” may be present at the ventral cervicomedullary junc-
(400,401,428–430). Most patients have complete lack of psychomotor tion (Figs. 5-46 and 5-47). The cerebellum is small and dysmorphic
development and die in the first year of life secondary to recurrent aspi- with abnormal foliation; the vermis is more severely affected than the
ration and respiratory illnesses. As discussed above, this phenotype has hemispheres (Figs. 5-46 to 5-48, often referred to—incorrectly—as
been seen in mutations of multiple genes (406–411). While the WWP “cerebellar polymicrogyria”). Orbital imaging findings include ocular
is seen in all ethnic groups, a striking difference has been observed asymmetry, secondary to congenital microphthalmos or congenital

Barkovich_Chap05.indd 411 5/6/2011 8:59:12 PM


412 Pediatric Neuroimaging

FIG. 5-46. Walker-Warburg phenotype. A. Sagittal T1-weighted image shows ventriculomegaly with extremely stretched
corpus callosum. The brainstem is characterized by kinking at the pons (small white arrowhead) and cervicomedullary
junction (large white arrowhead), a huge quadrigeminal plate (white arrows) and a very small cerebellar vermis. B. Axial
T2-weighted image shows the very small cerebellar hemispheres (white arrows) and a midline pontine cleft (black arrow).
C and D. Axial T2-weighted images show the appearance of the cobblestone cortex. Image from this patient (C) shows a
smooth outer cortical surface with nodules of gray matter protruding radially inward. Images from a different patient (D)
beautifully show radially oriented bands of neurons (black arrows), separated by strands of gliotic white matter.

glaucoma, with vitreal and subretinal hemorrhages due to retinal dys- Fukuyama Congenital Muscular Dystrophy Phenotype FMCD is
plasia or sublenticular hemorrhage due to persistent hyperplastic pri- the most common congenital muscular dystrophy in Japan (396,397).
mary vitreous (PHPV) (Fig. 5-49). See later section of this chapter for It is an autosomal recessive disorder, with the vast majority of Japa-
a discussion and descriptions of ocular anomalies. Rarely, a coloboma nese cases (with the classic phenotype) being caused by an ancient ret-
is identified. No correlation has been noted between the MR appear- rotransposon insertion in the 3’ noncoding region of the FKTN gene at
ance of the brain or orbits and the mutated gene (125). chromosome 9q31–33 (434,435). However, other mutations of FKTN
The WWP can be detected by fetal MRI during the mid second can cause the very severe WWP (426,436) and the milder limb-girdle
trimester (Fig. 5-50) by observation of the very small cerebellum, the muscular dystrophy phenotype (437); thus, as stated above, clinical
characteristic brainstem, the very large tectum, the ventriculomegaly, phenotype is dependent on the effect of the mutation on the function
and (if imaged between 20 and 24 weeks, when a distinctive pattern of of the protein in the linkage of RGCs to the pial limiting membrane and
cerebral lamination is normally seen (see Chapter 2) (433), the absence the structural integrity of the membrane (409). Patients with FCMD
of the normal cerebral lamination. The ocular findings (microphthal- phenotype present with hypotonia and severe developmental delay;
mia, colobomas) are confirmatory, if detected. seizures develop during the first year of life in about half of affected

Barkovich_Chap05.indd 412 5/6/2011 8:59:13 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 413

FIG. 5-47. Walker-Warburg phenotype with occipital cephalocele. Sagittal T1-weighted image (A) shows hydrocephalus
with the pontine (small white arrowhead) and cervicomedullary junction (large white arrowhead) kinks, a very small vermis,
and a small cephalocele (white arrow) protruding through the occipital bone. Coronal T2-weighted image (B) nicely shows
the cobblestone cortex and an extremely small cerebellum.

individuals. Analysis of serum shows elevated creatine kinase level; Neuroimaging with MR reflects the gross pathologic findings
this elevation typically leads to a muscle biopsy, which shows changes (438,439,441–443). Within the cerebral cortex, three types of abnormal-
consistent with muscular dystrophy (396,397,438,439). Patients with ity are seen pathologically (441). Of these, two are detected by imag-
FCMD may have ocular abnormalities (in particular dysplasias of ing: unlayered PMG, which is seen primarily in the frontal lobes, and
the retina that lead to myopia, nystagmus, chorioretinal degeneration cobblestone cortex, which is largely temporo-occipital (439). The fron-
[440]), but they are less clinically severe than those in MEB and WWP. tal PMG is seen as irregularity of the cortical surface and cortical-white
Hydrocephalus is less common than in patients with WWP. Anomalies matter junction (Fig. 5-51). The temporo-occipital cobblestone cortex
of the corpus callosum and cephaloceles are uncommon. is thick with a smooth outer surface (Fig. 5-51) but a slightly irregular

FIG. 5-48. Walker-Warburg phenotype in a neonate. Sagittal T1-weighted image (A) shows massive hydrocephalus. Note that the pons is
very thin and the quadrigeminal plate is enlarged with fused colliculi. Axial T2-weighted image (B) shows massive ventriculomegaly. The
characteristic “cobblestone” cortex is seen, although it is more difficult to appreciate in the setting of severe hydrocephalus.

Barkovich_Chap05.indd 413 5/6/2011 8:59:14 PM


414 Pediatric Neuroimaging

FIG. 5-49. Ocular anomalies in Walker-Warburg phenotype include intravitreal hemorrhage (A), subretinal hemorrhage (B), and
persistent hyperplastic primary vitreous (C and D).

inner surface. As in many patients with WWP, partly contiguous nodules in Finland and caused by mutations of POMGnT1 (444), which
of gray matter can be identified deep to the inner surface of the cortex participates in O-mannosyl glycan synthesis. It turns out that MEB
(Fig. 5-51). Most patients with FCMD have dysmorphic cerebellar cor- can also be caused by mutations in a number of different genes, the
tex, which is manifest on imaging studies as dysmorphic folia with sub- molecular products of which are involved in that pathway, includ-
cortical cysts. These cysts tend to be located in the dorsal midportion of ing POMGnT1 (445), POMT1 (408), LARGE (446), and FKRP (406).
the cerebellar hemisphere, particularly in the superior semilunar lobule Affected patients are hypotonic and have impaired vision (manifest as
(Fig. 5-51) (438). On histology, the cysts are found to have leptomenin- impaired visual fixation) from birth; epilepsy is common and mental
geal tissue within them and a molecular layer of nearly normal cerebellar retardation is usually severe (399). Spasticity typically begins to develop
tissue lining their walls, suggesting that they may have formed from sub- after age 5 years (399).
arachnoid spaces that were engulfed by fusion of neurons and glia that On pathologic examination, the cerebral cortex of patients with
migrated beyond the cerebellar pial limiting membrane (438,443). The MEB shows abnormal convolutions with a granular (cobblestone) sur-
other major finding on imaging studies of FCMD is delayed myelination. face to the cerebral cortex. A limited area of agyria may be seen over the
The abnormal white matter is hypodense on CT and shows abnormal T1 occipital convexity. Light microscopy shows resemblance to the cor-
hypointensity and T2 hyperintensity on MRI. Of interest, when myeli- tex in WWP and FCMD in that irregular bundles of myelinated axons
nation does occur, it begins in the subcortical white matter (Fig. 5-51) penetrate the cortex from the white matter and gliovascular strands
and extends centrally (439) in a pattern completely opposite to that of cut through the cortex from the pia, thereby separating the cortex into
normal development (in which periventricular and deep white matter is irregular neuronal clusters (432). No horizontal lamination or verti-
myelinated first and subcortical white matter last, see Chapter 2). cal columns of cortical neurons can be identified. Similar gliovascular
FMCD is difficult to detect by fetal MRI. In our experience, the strands penetrate the cerebellar cortex, which is dysplastic and shows
abnormality can be suspected in the early third trimester when a delay cysts similar to those in Fukuyama CMD (432).
in sulcation is associated with cerebellar cysts. If the cysts cannot be Neuroimaging of patients with MEB reveals diffusely abnormal
detected, the delay in sulcation itself is nonspecific. cerebral cortex, which resembles PMG; it is thickened with decreased
number and depth of sulci, with most severe involvement in the frontal
Muscle-Eye-Brain Phenotype The MEB phenotype was originally lobes (Figs. 5-52 and 5-53). Myelination is delayed (Fig. 5-52B and C)
thought to be a specific disorder (398,399), found predominantly and, as in FCMD, it begins in the subcortical, instead of deep, white

Barkovich_Chap05.indd 414 5/6/2011 8:59:15 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 415

FIG. 5-50. Fetal MRI of Walker-Warburg phenotype. A and


B. Sagittal images at 22 (A) and 30 (B) weeks gestation show the
large tectum (black arrows), pontine kink (small black arrow-
head), and cervicomedullary kink (large black arrowhead). The
cerebellar vermis is very small at both stages. C. Axial image
shows marked ventriculomegaly with a thin cerebral mantle.

matter (Fig. 5-52C). Patches of T2 hyperintensity may remain within of ocular anomalies (colobomas, microphthalmia) or cerebellar cysts
the cerebral hemispheres (Figs. 5-52D and 5-53B) even after myelina- confirms the diagnosis.
tion is completed. The brainstem is characterized by a very hypoplas-
tic pons with a vertical anterior midline cleft (Fig. 5-53A) and a large, Bilateral Frontoparietal Polymicrogyria The syndrome called bilat-
abnormally rounded midbrain tectum (Fig. 5-52A). The ventricles are eral frontoparietal polymicrogyria (BFPP) is an autosomal recessive
typically large, the Sylvian fissures are wide, and the cerebellum is hyp- syndrome that is found primarily in families from central Asia and the
oplastic with dysmorphic folial pattern and cortical/subcortical cysts Middle East, although families have been found in the western hemi-
(Figs. 5-52C and 5-53A) (447). Hydrocephalus may be present and sphere, as well (128). It has been well described and the causative gene
require shunting, the septum pellucidum may be absent and the cor- (GPR56) has been identified at 16q12.2–21 (128,448). In the cerebrum,
pus callosum may be dysmorphic or hypoplastic; it is not clear whether the gene product appears to be important for attachment of the distal
the latter findings are developmental or due to hydrocephalus. Thus, end of RGCs to the pial limiting membrane; when the gene is knocked
the imaging appearance can be considered intermediate between the out in animal models, gaps appear in the membrane, allowing over-
WWP and FCMD phenotype. migration of neurons (405). In the cerebellum, expression of GPR56
The diagnosis of MEB phenotype can be suggested on fetal MRI if appears to be important in the adhesion of migrating granule cells to
a fetus is found to have a large tectum and small pons with ventral cleft extracellular matrix molecules of the pial basement membrane in the
in association with delayed or abnormal sulcation (Fig. 5-54). A finding rostral cerebellum (449). These pathophysiologic mechanisms are very

Barkovich_Chap05.indd 415 5/6/2011 8:59:16 PM


416 Pediatric Neuroimaging

FIG. 5-51. Fukuyama congenital muscular dystrophy in an 11-month-old girl. A. Sagittal T1-weighted image shows hypoplas-
tic pons, hypoplastic vermis, and fused colliculi. B. Axial T2-weighted image shows multiple cerebellar cortical cysts. C. Axial
T1-weighted image shows lack of myelin in the cerebral white matter and absence of normal gyri, particularly in the temporal
and occipital lobes. D. Axial T2-weighted image at the same level as (C) shows irregularity of the cortical-white matter junction
(open black arrows) in the frontal lobes, suggestive of polymicrogyria, and temporal/parietal cobblestone lissencephaly. Note the
nodules of gray matter (solid black arrows) just central to the occipital cortex. E–G. Coronal T1-weighted images show myelina-
tion (arrows) beginning in the subcortical white matter with the deep and periventricular white matter still unmyelinated.

Barkovich_Chap05.indd 416 5/6/2011 8:59:17 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 417

FIG. 5-52. Myelinating MEB phenotype. Images (A–C) were acquired at age 18 months, image (D) was acquired at age 4 years.
A. Sagittal T1-weighted image shows hypoplastic pons, dysmorphic cerebellar vermis with cortical cysts, and fused midbrain colliculi.
B. Axial T2-weighted image shows abnormally shallow sulci in the frontal lobes and incomplete myelination. Note that the subcorti-
cal U fibers are affected in the frontal lobes, but spared in the parietal and occipital lobes. C. Coronal T1-weighted image shows that
the subcortical white matter is myelinating, but the deep white matter is not; this pattern is characteristic of congenital muscular
dystrophies. Note the cortical/subcortical cerebellar cysts (white arrows). D. Axial T2-weighted image shows progression of myelina-
tion. Note that several areas of abnormal patchy T2 hyperintensity (white arrows) remain.

similar to that described with other disorders described in this section; MRI in BFPP shows thickened, irregular frontal cortex; the medial
thus, it is not surprising that a cobblestone-like malformation devel- aspects of the frontal lobes are often involved, which helps differentia-
ops. The phenotype is similar to, but milder than, MEB. In view of the tion from true PMG. Abnormal foci of T2 prolongation are commonly
cobblestone-like pathophysiology, some argue that it is not correct to seen in the white matter immediately subjacent to the cortex. The cor-
call this disorder PMG; however, BFPP is the term currently being used pus callosum is large and shows abnormal contour. The cerebellum is
in the literature and it will be used here. Affected patients show a charac- small and dysmorphic, with an appearance similar to those of patients
teristic syndrome of developmental delay in both cognitive and motor with CMD, but only a few, small cysts are seen and those only occa-
spheres, disconjugate gaze (typically esotropia), refractory seizures sionally. The pons is typically small (128), but not as small as in MEB
(most commonly symptomatic generalized epilepsy), and ataxia (450). (Fig. 5-55).

Barkovich_Chap05.indd 417 5/6/2011 8:59:19 PM


418 Pediatric Neuroimaging

FIG. 5-53. MEB phenotype. Axial T2-weighted


images show midline pontine cleft (white arrow-
heads in A), cerebellar cysts (white arrows in A),
abnormal frontal gyri (white arrows in B) and foci
of abnormal myelination (white arrowheads in B).

FIG. 5-54. Thirty-two-week fetus with MEB pheno-


type. A. Sagittal image shows ventriculomegaly with large
tectum (black arrow), small pons, and small cerebellum.
Pontine kink is similar to that in Walker-Warburg syn-
drome phenotype. B. Axial image shows small cerebel-
lar hemispheres and small pons with midline cleft (white
arrow). C. Axial image shows marked ventriculomegaly
and abnormal cortex with multiple small gyri.

Barkovich_Chap05.indd 418 5/6/2011 8:59:20 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 419

FIG. 5-55. Bilateral frontoparietal


polymicrogyria due to GPR56 mutation.
Sagittal (A) and axial (B) T2-weighted
images show small pons, small dysmor-
phic cerebellum, and a cobblestone-like
cortex (white arrows in B), with abnor-
mal sulci and the cortex composed of
radially-oriented bundles of neurons.

Other Congenital Muscular Dystrophy Syndromes Other congenital therefore, distinct from MSG pattern, in which too few neurons are
muscular dystrophy syndromes have been described that do not fit well produced or too many undergo apoptosis (programmed cell death),
into the Walker-Warburg, FCMD, MEB, or BFPP phenotypes. These and from cobblestone malformation due to abnormal pial basement
include CMD with occipital agyria (see Chapter 3 and Fig. 3–17) (422), membrane formation, which are associated with overmigration of neu-
and those with cerebellar cysts and leukodystrophy without cortical rons. Patients with LIS1 mutations seem to have defects in outgrowth
malformations or hydrocephalus (Fig. 5-56) (425,451,452). From the of neuronal processes (the “arms” that extend outward from the neu-
point of view of the neuroimager, it is important to remember that ronal cell body and act as leading processes) and in transporting the cell
the findings of leukodystrophy, cobblestone cortex, cerebellar subcor- nucleus into the neuronal processes; as a result, the postmitotic neuron
tical cysts, small pons, and rounded quadrigeminal plate suggest the cannot navigate from the ventricular zone to the radial glial pathway
diagnosis of cobblestone malformation due to abnormal pial basement (456). Both GABAergic interneuron migration from the ganglionic
membrane formation. Additional clinical, genetic, and imaging char- eminences and glutamatergic neuron migration from the dorsal hemi-
acteristics will contribute to the final diagnosis. spheric germinal matrix seem to be affected in lissencephaly (457).

Lissencephaly (The Agyria-Pachygyria Complex) The term lis- Classic Lissencephaly/Band Heterotopia Spectrum Classic
sencephaly means “smooth brain” and refers to a paucity of gyral and lissencephaly is the term that describes the appearance seen with the
sulcal development on the surface of the brain. Agyria is defined as an first described and most common form of lissencephaly: a thick cortex
absence of gyri on the surface of the brain in association with a thick composed of (a) a thin molecular layer adjacent to the pia, (b) a thin
cortex and is synonymous with “complete lissencephaly,” whereas pachy- outer cortical layer, (c) a “cell-sparse zone” medial to the outer corti-
gyria is defined as the presence of a few broad, flat gyri with thickened cal layer, and (d) the thickest part of the cortex located deep (medial)
cortex and is used interchangeably with the term “incomplete lissen- to the cell-sparse zone (Fig. 5-57). Some patients with classic lissen-
cephaly.” These definitions adopt the concepts of Hennekam and Barth cephaly have a defect of the LIS1 (also called PAFAH1B1) gene at locus
that proper use of the terms agyria and pachygyria requires a thick cortex 17p13.3 (124,458,459). A subset of the group with LIS1 mutations also
and that these malformations result from an arrest of neuronal migra- has mutation of the adjacent YWHAE gene, resulting in characteris-
tion (199). A finding of broad gyri and shallow sulci in the absence of tic facial anomalies (bitemporal hollowing, prominent forehead, short
thickened cortex should be considered a simplified gyral pattern, result- nose with upturned nares, prominent upper lip, thin vermilion border
ing from either deficient cell production or impaired white matter devel- of the upper lip, small jaw); this group is classified as the Miller-Dieker
opment in utero (199). Of importance in distinguishing pachygyria from syndrome (460,461). Other patients with classic lissencephaly have
PMG (see section “Malformations Secondary To Abnormal Late Migra- mutations of the DCX gene at chromosome Xq22.3-q23 (X-linked lis-
tion and Cortical Organization”) is the observation that the sulci seen in sencephaly [462]); these patients are typically hemizygous boys whose
pachygyria (and in simplified gyral patterns) are normal, early forming mothers have band heterotopia (122,463,464) (affected female siblings
sulci that can be identified and named by those experienced in neuro- also have band heterotopia). A third gene, TUBA1A at chromosome
anatomy; the sulci in PMG are abnormal and do not correspond to those 12q12, accounts for approximately 7% of classic lissencephaly with a
described in neuroanatomy texts. In addition, the cortex of pachygyria four-layer cortex and cell-sparse zone (465); the diagnosis can be sug-
has smooth inner and outer surfaces, whereas that of PMG will be seen gested if an anomalous corpus callosum (absent rostrum, abnormally
to have irregularities due to the microgyri and microsulci (453). curved body, or vertical splenium), small cerebellum, rounded hip-
Both agyria and pachygyria are likely caused by abnormal regu- pocampi, or thin brainstem are identified (131,465,466).
lation of microtubule activities. Microtubules are involved in many It has been estimated that 40% to 75% of patients with classic lis-
key intracellular processes, such as mitotic spindle orientation, vesicle sencephaly have mutations of either LIS1 (up to 60%) or DCX (∼12%)
transport, and nuclear migration (454,455). Most genes known to cause (131,467–469), meaning that up to half of lissencephaly patients have
classic lissencephaly are known to participate in microtubule dynam- other mutations; this is not surprising in view of the many genes that
ics and abnormalities of this function almost certainly causes, in some are involved in normal neuronal migration. Irrespective of the causative
way, an arrest of normal neuronal migration (454). Lissencephalies are, gene, children with classic lissencephaly have similar neurological

Barkovich_Chap05.indd 419 5/6/2011 8:59:21 PM


420 Pediatric Neuroimaging

FIG. 5-56. Congenital muscular dystrophy due to FKRP mutation


in a 13-month-old child. Sagittal T1-weighted image (A) shows small
pons and small, vertical callosal splenium. Coronal T1-weighted
image (B) shows hyperintensity from early myelination in subcortical
(black arrows) but not deep white matter; note cerebellar cysts (white
arrows). Axial T2-weighted image (C) shows irregular, microgyric-
like cortex (black arrows) in both frontal lobes.

FIG. 5-57. Schematic showing cortical archi-


tecture in classic lissencephaly. In complete
lissencephaly (agyria), a large cell-sparse zone
(open black arrow) separates the molecular layer
(layer I) and an outer cortical layer (layers III, V,
VI in figure) from a thick deeper layer of dis-
organized neurons. In incomplete lissencephaly
(pachygyria), the outer cortical layer is thicker,
the cell-sparse zone (black arrow) thinner, and
the inner cortical layer smaller.

Barkovich_Chap05.indd 420 5/6/2011 8:59:22 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 421

syndromes, although the age of onset and severity of the clinical ber of neurons that are mutated and to the severity of the mutation
syndrome may vary depending upon the severity of the cortical mal- (126,464,469,478,480–483). Fewer neurons with mutations, secondary
formation (335,461,470,471). Those with complete classic lissenceph- to mosaicism or heterozygosity, or mutations that have (presumably)
aly are typically hypotonic at birth, gradually developing appendicular less severe effects on neuronal migration result in milder phenotypes
and oropharyngeal spasticity as their nervous system matures (470,472). (less severe pachygyria or band heterotopia) (469,475). In patients with
Children with incomplete lissencephaly have less severe motor abnor- LIS1 mutations, the areas of agyria are most frequently parietooccipital
malities and hypotonia (470). Infantile spasms are common in severely in location while the pachygyric areas are more common in the fron-
affected infants. The development of medically refractory epilepsy at a tal and temporal regions (470,472,484); indeed, about two thirds of
very early age, with increasingly complex seizure disorders over time, is patients with posterior predominant lissencephaly have mutations of
characteristic. Systemic anomalies, particularly those of the ears, eyes, LIS1 (471). Some patients have more severe pachygyria or agyria in the
heart, and kidney, are present in the more severely affected patients frontal lobes with less severe involvement of the occipital lobes; most of
(429,460,461,470,473). these patients seem to have mutations of DCX (485). Microscopically,
Mothers of boys with lissencephaly due to DCX mutations have the cerebral cortex in patients with LIS1 mutations is composed of a
the same mutation of the X chromosome as do their offspring but normal molecular layer, a thin outer layer composed of neurons that
have band heterotopia instead of lissencephaly (474), or may even have would normally be in cortical layers 5 and 6, a cell-sparse zone contain-
mental retardation or epilepsy with completely normal MR studies ing neurons that would normally be in layer 4, and a thick inner layer
(475,476). These patients seem to have either mild mutations or inac- composed of neurons that would normally be in cortical layers 2 and 3
tivation of a large percentage of the abnormal X chromosomes (475). (Fig. 5-57) (486). The inner layer of neurons is thought to represent
Many patients with band heterotopia have a significantly reduced num- neurons that were prematurely stopped during their migration to the
ber of sulci and gyri (477). As it has been shown that band heterotopia cortex. No radial or horizontal organization is seen in the cortex of
can be caused by mild mutations of DCX or LIS1 (478,479) or other patients with TUBA1A mutations; a cell-sparse zone may (466) or may
genes (204), they are probably better classified as a form of incomplete not (131,466) be identified, depending upon the location of the muta-
lissencephaly. They are therefore discussed in the “Lissencephaly (The tion and, perhaps, other factors.
Agyria-Pachygyria Complex)” section rather than the “Heterotopia” Imaging studies of patients with complete classic lissenceph-
section in this edition. aly (agyria) reveal a smooth brain surface with diminished white
Macroscopic examination of the brain shows that most patients in matter and shallow, vertically oriented, Sylvian fissures (Fig. 5-58)
this group have areas of both agyria and pachygyria, that is, incomplete (461,470,472,487,488). In patients with LIS1 and DCX mutations, a
lissencephaly. The severity of the gyral pattern is related to the num- thin outer cortical layer is separated from a thick deeper cortical layer

FIG. 5-58. Agyria in a neonate. Sagittal T1-


weighted image (A) shows no medial hemispheric
sulci other than the parietooccipital sulcus. Axial
(B) and coronal (C) T2-weighted images show
a smooth cerebral surface and the typical corti-
cal pattern with thin (dark) outer cortical layer,
relatively thick (bright) cell-sparse zone, and thick
(dark) inner cortical layer (black arrows). Vol-
ume of white matter is diminished. Note relative
enlargement of the ventricular trigones.

Barkovich_Chap05.indd 421 5/6/2011 8:59:24 PM


422 Pediatric Neuroimaging

by a zone of white matter (the cell-sparse zone, Fig. 5-58) that seems pachygyria also have a thickened cortex, but broad gyri and shallow
to myelinate normally as the child matures. The cerebrum has been sulci (Figs. 5-60 and 5-61) are present. Pachygyria can be distinguished
described as having a figure-of-eight appearance on axial images as a from PMG when thin section, high resolution images are obtained
result of the shallow, vertical Sylvian fissures. In cases of severe lissen- (453). In pachygyria, the cortical-white matter junction is smooth,
cephaly, sagittal images may show callosal hypogenesis with a charac- and, in some cases, a layer of normal white matter can be detected in
teristic vertical splenium. The ventricular trigones and occipital horns the cell-sparse zone (Figs. 5-60 and 5-61). In PMG, the cortical-white
are enlarged, mostly because of underdevelopment of the calcarine matter junction is always irregular (453) (see Fig. 5-62 and the sub-
sulci (Fig. 5-58) (472). The brainstem often appears small, probably sequent section “Polymicrogyria” under “Malformations Secondary to
because many of the corticospinal and corticobulbar tracts do not Abnormal Cortical Organization and Late Migration”). Pachygyria can
form. Absence of the corpus callosum or severe pontine/cerebellar be focal or diffuse. When focal (Fig. 5-61), it is almost always bilateral
hypoplasia should suggest a TUBA1A mutation (489,490). Agyria can and typically posterior in patients with LIS1 mutations. When diffuse
sometimes be detected by prenatal imaging (MRI or ultrasound) by (Fig. 5-60), it is often associated with regions of agyria and is typi-
the absence of gyri, persistence of a cell-sparse zone after 25 gestational cally more severe in the parietooccipital region of the brain (and least
weeks (Fig. 5-59) and the abnormal morphology of the sylvian fissures severe in the frontal and temporal lobes) in LIS1 mutations (485). The
(in the normal fetus, the base of the sylvian fissure, the insula, has a mid-frontal lobes are most severely affected in pachygyria secondary
“square” appearance with sharply angled margins, whereas the lissen- to DCX mutations (Fig. 5-63) (485). When patients have classic lissen-
cephalic patient has a shallow, rounded sylvian fissure [491]). cephaly with more severe involvement in the anterior cerebrum than
Incomplete lissencephaly, in which areas of pachygyria are present in the posterior cerebrum, a good family history should be obtained.
together with areas of agyria (Figs. 5-60 and 5-61) or areas of normal Band heterotopia may be found in some female family members if
brain, is much more common than complete lissencephaly. Areas of there is family history of seizures (Fig. 5-63D).

FIG. 5-59. Agyria in a 32-week fetus. Sagittal image (A)


shows a mature posterior fossa with large pons and cere-
bellar vermis. Axial (B) and coronal (C) image show that,
despite fetal maturity, the surface of the brain remains
smooth. A cell-sparse zone (white arrows) can be seen; nor-
mally, “layers” of the cerebral cortex are not seen after about
25 weeks. Also notice the persistent rounding of the sylvian
fissures (black arrowheads); the maturation of the insulae
normally cause “squaring” of the sylvian fissures by about
23 to 24 gestational weeks (see Chapter 2).

Barkovich_Chap05.indd 422 5/6/2011 8:59:24 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 423

FIG. 5-60. Diffuse pachygyria (chromosome 17-linked). A. Parasagittal T1-weighted


image shows thickened cortex with relatively few and large gyri and sulci. B. Axial
T1-weighted image shows that the pachygyria is more severe in the parietal and occipi-
tal lobes than in the frontal lobes. The sylvian fissures (curved black arrows) are abnor-
mally vertical. C. Axial T2-weighted image shows hyperintensity (small white arrows),
representing the cell-sparse zone, in the parietal lobe.

Lissencephaly with Cerebellar Hypoplasia. Some patients with DCX mutations (465). In my experience, undulation of the deep
classic lissencephaly have anomalies of the corpus callosum or cerebel- cortical layer (Fig. 5-64B) is also most common with TUBA1A muta-
lar hypoplasia (Fig. 5-64) (131,485,492). Patients with DCX mutations tions. Therefore, if both cerebellar and callosal anomalies are present
may have complete callosal agenesis or an abnormally shaped corpus in a patient with the appearance of classic lissencephaly, particularly if
with a flat callosal body and vertical splenium (493). Patients with the deep cortical layer is undulating, testing for TUBA1A mutations is
LIS1, DCX, and TUBA1A mutations all can have cerebellar hypoplasia warranted.
(131,485,492,494), but it is usually only mildly hypoplastic in LIS1 and
DCX mutations, likely resulting from the fact that the same ligands and Band heterotopia. Band heterotopia (477,495), also called “double
processes that are involved in neuronal migration in the cerebrum are cortex” (496,497), is the mildest form of classic lissencephaly. The layer
also at work in the cerebellum. Of the well-characterized lissenceph- of arrested neurons is thinner, the cortex is thicker, and the cortex has
alies, only TUBA1A lissencephalies consistently have anomalies of the a more normal gyral pattern than in pachygyria. Affected patients are
corpus callosum, cerebellum, and hippocampus (Fig. 5-64); TUBA1A almost exclusively female and may present at any age, although they
mutations comprise greater than 30% of lissencephalies with signifi- usually present in childhood with developmental delay of variable
cant cerebellar hypoplasia (465). Severe callosal hypoplasia/hypogen- severity and mixed seizure disorders (477,496–498). Several authors
esis is nearly always associated, along with a very hypoplastic brainstem have described patients who are normal except for relatively mild
(Fig. 5-64A). The cortex is pachygyric, but not as thick as in LIS1 or seizure disorders (463,477,496). Rarely males can have band heterotopia

Barkovich_Chap05.indd 423 5/6/2011 8:59:25 PM


424 Pediatric Neuroimaging

FIG. 5-62. Polymicrogyria. Note the irregular cortical-white matter junc-


tion (black arrows). Contrast with Figures 5-60 and 5-61.

thickness with shallow sulci; thickness of the outer cortex can be used
as a (rather arbitrary) marker to differentiate band heterotopia (thin in
lissencephaly, normal in band heterotopia). Band heterotopia may be
complete, surrounding the central white matter (Figs. 5-65 and 5-66),
or partial (463,503); when partial, the frontal lobes seem to be pref-
erentially involved in women (DCX mutations, Fig. 5-63D), whereas
the posterior brain regions are more commonly involved in men (LIS1
FIG. 5-61. Posterior pachygyria. A. Parasagittal T1-weighted image shows mutations) (463,498,503). Rarely, a second layer of heterotopia may
normal appearing frontal cortex with pachygyria (white arrows) in the be identified in the temporal lobe (477). A more severe anomaly of
parietal and occipital lobes. B. Coronal T1-weighted image shows a smooth the cortex is associated with a worse clinical prognosis for epilepsy.
cortical-white matter junction (open black arrows) and the cell-sparse zone Foci of T2 prolongation may be seen in the cerebral white matter; the
(solid black arrows). presence of these foci is associated with a poor motor outcome (477).
When imaged in a neonate, the band may be mistaken for myelinating
white matter; identification of the cell-sparse zone between the band
secondary to somatic mosaicism of LIS1 or hemizygous mutations for and the cortex (Fig. 5-67), in addition to the shallow sulci, will help to
DCX (477–479,499–501). One case report has revealed band hetero- make this differentiation easier. When studied by PET using 18F-FDG,
topia associated with trisomy 9p; mutation analysis of LIS1 and DCX band heterotopia are found to have a glucose uptake that is similar to
was negative (204). Other reports of patients with 9p duplication have (504,505) or greater than (506) normal cortex. This finding contrasts
been reported with epilepsy, mental retardation, and brain abnormali- with the hypometabolism found in cortical dysplasias and in most epi-
ties, suggesting that this is another cause of band heterotopia (498). In leptogenic foci. Morell et al. (507) have found epileptiform discharges
addition, we have seen a region of band heterotopia in a patient with emanating from the band, suggesting that it is the source of the seizure
Zellweger syndrome (see Chapter 3). Thus, the indications are that, activity in affected patients. Others have found increased blood flow to
although most cases are due to DCX mutations, mutations of many the portion of the band that underlies the motor cortex using BOLD
genes involved in neuronal migration can cause band heterotopia. His- imaging (508,509). This combination of findings suggests that the
tologic analysis shows that the subcortical bands contain heterotopic band may have substantial communications with the rest of the brain
neurons that are of small pyramidal shape without clear radial orienta- and may send out projection fibers to communicate with the body.
tion. The neurons may be arrayed into clusters, sheets, or wide bands.
The layer of heterotopic neurons is separated from the overlying cortex Variant Lissencephalies Variant lissencephaly is a term used to
and underlying lateral ventricles by normal white matter. Of interest, describe brain anomalies with thick cortex (not as thick as in classic lis-
the overlying cerebral cortex has normal cytoarchitecture (4,502). sencephaly) and reduced sulcation, but no cell-sparse zone (486). Four
On imaging, band heterotopia appear as homogeneous bands of different groups are currently in this category and it will likely grow.
gray matter that are situated between the lateral ventricles and the cere-
bral cortex and separated from both by a layer of normal-appearing X-linked Lissencephaly with Callosal Agenesis and Ambiguous
white matter (the white matter may be incompletely myelinated in Genitalia. X-linked lissencephaly with callosal agenesis and ambiguous
infants, Figs. 5-65 and 5-66). The overlying cortex typically has normal genitalia appears to be a separate type of lissencephaly (510,511) that

Barkovich_Chap05.indd 424 5/6/2011 8:59:27 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 425

FIG. 5-63. Members of family with X-linked lissencephaly. A and B. Sagittal T1-weighted (A) and axial T2-weighted (B) images show
pachygyria in the frontal lobes (white arrows), but more normal gyral pattern posteriorly. C. Axial T2-weighted image of a 7-month-old
boy, who is the nephew of the patient shown in (A) and (B). Note that the more normal gyri (white arrows) are in the posterior regions
of the brain and the more severe pachygyria/agyria is anterior. Note the cell-sparse zone (white arrowheads) in the agyric region. D. Axial
T2-weighted image of the mother of the boy in (C) and the sister of the man in (A) and (B) shows bilateral, right larger than left, anterior
band heterotopia (white arrows).

is caused by mutations of the ARX gene at Xp22.13 (512). The protein mental retardation with normal-appearing brains (514,515). All affected
product of ARX appears to function in the proliferation and develop- patients with X-linked lissencephaly and ambiguous genitalia to date
ment of cortical neurons (both interneurons and projection neurons) have been genotypic males, and all have had intractable neonatal onset
(513,514), as well as those of the thalamus, hippocampus, striatum, and epilepsy (typically myoclonic seizures), normal head size at birth with
olfactory bulbs. Of interest, mutations of this gene cause a wide variety severe microcephaly developing over the first few months of life, poor
of syndromes, from hydrocephalus with abnormal genitalia to mental temperature regulation, chronic diarrhea, and ambiguous or underde-
retardation with agenesis of the corpus callosum and abnormal genita- veloped genitalia (510,516,517). Related females may have mental retar-
lia, to infantile epileptic-dyskinetic encephalopathy, infantile spasms or dation and epilepsy and, often, agenesis of the corpus callosum (516).

Barkovich_Chap05.indd 425 5/6/2011 8:59:27 PM


426 Pediatric Neuroimaging

FIG. 5-64. Lissencephaly with callosal anomaly and cerebellar hypoplasia. Sagittal T1-weighted image (A) shows
severe pontocerebellar hypoplasia and absence of the corpus callosum. Axial T2-weighted image (B) shows a smooth
cortical surface with an undulating deep cortical layer (black arrows). The combination of callosal agenesis, pontocer-
ebellar hypoplasia, and undulating deep cortex should raise the possible diagnosis of TUBA1A mutation.

FIG. 5-65. Typical appearance of band


heterotopia. Parasagittal T1-weighted
image (A) shows the gray-matter-intensity
band (b) separated from the cortex by a
layer of partially myelinated white mat-
ter (m). Note that the cortex has nor-
mal thickness but shallow sulci. Axial
T2-weighted image (B) and coronal
T1-weighted image (C) show ventricu-
lomegaly and incomplete subcortical
myelination.

Barkovich_Chap05.indd 426 5/6/2011 8:59:29 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 427

The cerebral cortex, usually measuring about 6 to 7 mm in thickness,


is thicker than normal but is thin compared to that in lissencephaly
secondary to LIS1 or DCX mutations (which are usually about 15 mm
thick) (512). The corpus callosum is always completely absent and the
basal ganglia are either small and dysplastic (often cystic, Fig. 5-68) or
completely absent. The brainstem and cerebellum appear normal.

Lissencephaly Secondary to Mutations in the Reelin (Reln) Signal-


ing Pathway. Lissencephaly secondary to mutations of the reelin sig-
naling pathway results in severely affected children, who are hypotonic
at birth and showed marked delay in motor and cognitive milestones;
they do not sit or stand unsupported, nor do they develop language
(494,519,520). Generalized epilepsy begins at an early age; some
affected patients have congenital lymphedema (519). Reelin, the protein

FIG. 5-66. More severe band heterotopia. Coronal T2-weighted image


shows a thicker layer of heterotopia (b) with fewer, shallower sulci. This is
difficult to separate from pachygyria, but we use the normal thickness of
the outer cortical layer as a marker of band heterotopia.

On pathologic examination, brains of affected patients show


absence of the corpus callosum without Probst bundles; a trilayered
cortex (composed of a cellular molecular layer, a thin middle layer
composed of mainly pyramidal neurons, and a thick inner layer com-
posed of multipolar neuronal types); small, disorganized basal ganglia;
hypoplastic or absent olfactory bulbs and optic nerves; spongy, gliotic
white matter containing numerous heterotopic neurons; and dysplastic
hypothalamus lacking most of its normal nuclei (486,516,518).
Imaging studies show anterior pachygyria, with only a few, shallow
sulci, and posterior agyria (Fig. 5-68); no cell-sparse zone is identified.

FIG. 5-68. Neonate with X-linked lissencephaly with agenesis of the


corpus callosum and ambiguous genitalia due to ARX mutation. Sagittal
T1-weighted image (A) shows absence of the corpus callosum and abnor-
mal sulcation of the medial cerebral hemisphere. Axial T1-weighted image
(B) shows areas of pachygyria and agyria with moderately thick cortex. Note
that the cortex is less thickened than in lissencephaly secondary to XLIS or
FIG. 5-67. Band heterotopia in an infant. The band (b) is separated from DCX mutations. No cell-sparse zone is identified. Basal ganglia are nearly
the overlying cortex by an unmyelinated cell-sparse layer (c). absent, with enlarged perivascular spaces (white arrows) in their place.

Barkovich_Chap05.indd 427 5/6/2011 8:59:30 PM


428 Pediatric Neuroimaging

FIG. 5-69. Variant lissencephaly secondary to reelin (RELN) mutation. A. Sagittal T1-weighted image shows abnormal gyral
pattern of the medial hemisphere, a very small and unsulcated cerebellum and a small pons. The callosal splenium is incompletely
formed. B. Coronal T1-weighted image shows decreased sulcation of the cerebrum, moderately thickened cortex. Note that no
cell-sparse zone is identified.

product of the RELN gene (which maps to 7q22), is an extracellular at chromosome 19q13.2, cause similar impairments of neuronal
matrix protein that is critical for the normal disengagement of migrat- migration and organization and result in similar malformations (522).
ing neurons from RGCs in mice and, likely, in humans. In its absence, Mutation of VLDLR is seen in the disorder that has been called the
younger migrating neurons are unable to pass neurons generated ear- disequilibrium syndrome (520,523).
lier and migrate to the outer layers of the cortex (181,521). As a result, Imaging studies of children with RELN mutations shows a thick-
the cortex is disorganized, with layer 6 neurons closest to the molecu- ened cortex (measuring up to 1 cm in thickness) with too few sulci
lar layer (layer1), followed by layer 5, layer 4, etc. (Normally, layer 5 (Fig. 5-69); of interest, no cell-sparse zone is identified. Hippocampi are
migrates past layer 6, layer 4 migrates past layer 5, etc. See discussion incompletely rotated. The brainstem and cerebellum are extremely small.
at the beginning of the section “Malformations of Cortical Develop- The cerebellum is completely smooth, with no foliation (Fig. 5-69). The
ment.”) Mutations of other genes in the reelin signaling pathway, such appearance is extremely characteristic and is sometimes referred to as
as the very low density lipoprotein receptor (VLDLR) gene, localized to the Norman-Roberts type of lissencephaly (494). Patients with VLDLR
chromosome 9p24, and the apolipoprotein E receptor 2 gene (APOE2) mutations have the same finding, but less severe (Fig. 5-70) (522).

FIG. 5-70. Variant lissencephaly secondary to mutation of very low density lipoprotein receptor (VLDLR) gene.
Sagittal T1-weighted image (A) shows abnormally broad midline gyri with few midline sulci. The pons and cerebel-
lum are abnormally small and the cerebellum (white arrows) has insufficient foliation. Axial T2-weighted image
(B) shows insufficient sulcation but normal cortical thickness. (These images are courtesy of Dr. Robert Sevick,
Calgary.)

Barkovich_Chap05.indd 428 5/6/2011 8:59:31 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 429

Lissencephaly with Central Predominance. In some patients with section “Malformations Secondary to Abnormal Cortical Organiza-
lissencephaly, the most severe gyral anomalies are in the posterior tion and Late Migration.”
frontal region (524). This pattern has been described to occur in the
Baraitser-Winter syndrome (276), a disorder characterized by micro- Periventricular (Subependymal) Nodular Heterotopia Patients
cephaly, ocular anomalies (iris and retinal colobomas, ptosis, hyper- with PVNH can be separated into two large groups. Most patients have
telorism, epicanthic folds), ptosis, mental retardation, dysmorphic heterotopia that are asymmetric, few in number, and largely confined
features, and short stature (525). We have also seen this pattern in a to the trigones and the temporal and occipital horns (Fig. 5-71); these
family with DCX mutations. Imaging characteristically shows abnor- are rarely familial but may be associated with other brain anomalies
mal sulcation with simplified gyral pattern (too few sulci) and frank such as the Chiari II malformation, cerebellar hypoplasia, cephaloceles,
pachygyria (with cortical thickening) in the suprasylvian/paracentral ectopic posterior pituitary gland, PMG, frontonasal dysplasia, Donnai-
cortex. Thin subcortical band heterotopia have been reported poste- Barrow syndrome, fragile X syndrome, and agenesis of the corpus cal-
riorly in a few cases (525). Pathology seems to be similar to that of losum (129,195,529–532). A smaller number of patients have a large
lissencephaly from DCX mutations (486). number of heterotopic nodules that completely or nearly completely
line the walls of the lateral ventricles (Fig. 5-72). PVNH may be famil-
Heterotopia Gray matter heterotopia are collections of nerve cells in ial, with both X-linked and autosomal recessive patterns of inheritance
abnormal locations secondary to arrest of radial migration of neurons. (127,533,534). A number of causative genes have been identified (533),
(Note that the word heterotopia is plural; the singular form of the noun including the filamin-A (FLNA) gene at chromosome Xq28 (535–537)
is heterotopion). Heterotopia can be isolated or can be seen in associa- and the ARFGEF2 gene at chromosome 20q13 (associated with micro-
tion with other structural anomalies. While one might argue that all cephaly [127]). Other loci associated with PVNH include 1p36 (538),
anomalies secondary to abnormal neuronal migration are heterotopia 5q14.3–15 (132), and 7q11.23 (539). It is currently thought that PVNH
(in the sense that all are composed of normal nerve cells in abnormal are caused by loss of integrity of the neural ependymal at the ventricu-
locations), the term heterotopia is reserved for cases in which the ecto- lar wall, either due to physical trauma or disruption of intracellular
pic neurons are in nodular or laminar form and are located in an area vesicle trafficking due to mutations (540). The loss of integrity of the
other than the cortex. neural ependyma may disrupt the attachment of RGCs, thus disrupt-
Patients with heterotopic gray matter almost always come to ing migration.
medical attention as a result of a seizure disorder (526–528). For Whether sporadic or familial, children with isolated heterotopia
the purposes of clinical evaluation and prognostication, it is useful (i.e., no other brain or visceral anomalies) usually manifest mild clini-
to divide heterotopia into three groups: (a) periventricular (sub- cal symptoms, with normal development, normal motor function, and
ependymal) heterotopia; (b) focal subcortical heterotopia; and (c) onset of seizures during the first or second decade of life. Seizures are
leptomeningeal heterotopia (7). Leptomeningeal heterotopia is the typically mixed partial complex and tonic-clonic (129,528,536,537,541).
result of overmigration of neurons through gaps in the pial limiting Focal cortical dysplasia may be found in the overlying cortex, possibly
membrane; those with genetic causes were discussed in the previ- acquired due to proliferation of seizure activity via axons connecting
ous section concerning malformations resulting from gaps in the pial the epileptogenic heterotopia with the affected cortex (542). Girls with
limiting membrane. When isolated or due to in utero injury to the X-linked PVNH often have a large cisterna magna and may have a
pial liming membrane, they cannot be detected by imaging at this small cerebellar vermis (Fig. 5-72) (526,535,536). Boys with syndromic
time. Another disorder, referred to as band heterotopia (or, some- subependymal heterotopia may have associated cortical malformations
times, double cortex) was discussed in this section in previous edi- (Fig. 5-73), syndactyly, ear abnormalities, and severe mental retarda-
tions of this book. However, band heterotopia are better classified tion (535). Both girls and boys with FLNA mutations are more likely to
as a mild form of lissencephaly (7) and are discussed in the “Classic have congenital cardiovascular defects such as patent ductus arteriosus
Lissencephaly/Band Heterotopia Spectrum” section, earlier in this or cardiac valvular anomalies, and a propensity for premature stroke

FIG. 5-71. Periventricular nodular het-


erotopia confined to the walls of the tem-
poral horns, trigones, and occipital horns
of the lateral ventricles. Axial T1-weighted
image (A) and coronal T2-weighted image
(B) show nodules of heterotopic gray mat-
ter (white arrows) protruding into the
ventricular trigones. When confined to
temporal horns and trigones, PVNH can be
difficult to identify on axial images.

Barkovich_Chap05.indd 429 5/6/2011 8:59:32 PM


430 Pediatric Neuroimaging

FIG. 5-72. Diffuse periventricular nodular


heterotopia associated with FLNA mutation. Sagittal
T1-weighted image (A) shows small cerebellar vermis
(white arrows) with enlarged pericerebellar cisterns.
Axial T2-weighted image (B) shows continuous het-
erotopia (white arrows) along the ventricular bodies,
while coronal T2-weighted image (C) shows hetero-
topia (white arrows) extending into white matter lat-
eral to ventricular bodies and temporal horns.

(129,543). Girls or boys with periventricular heterotopia and other due to FLNA mutations vary in configuration depending on what
malformations (callosal agenesis, schizencephaly, PMG, subcortical part of the FLNA protein the mutation affects and how severely pro-
heterotopia, cephalocele, etc.) are more likely to suffer from significant tein function is impaired (533,543,547). Although most patients with
cognitive impairment; their seizures tend to develop earlier (during the mutation of this gene show diffuse PVNH lining the walls of the bod-
first decade) and are more difficult to control (541). ies of the lateral ventricles, some with local missense mutations may
On imaging studies, PVNH are smooth, round or ovoid masses show only a few bilateral peritrigonal PVNH (543). Familial heteroto-
(Figs. 5-71 to 5-73) that are isointense with gray matter on all imaging pia also have an increased incidence of associated mega cisterna magna
sequences. When ovoid, the long axis is parallel to the adjacent ventric- (543,548,549).
ular wall. They may appear exophytic, protruding into the adjacent lat- PVNH can be identified by fetal MRI and fetal brain sonography
eral ventricle (Fig. 5-71), and sometimes causing the ventricle to appear (550,551), appearing as nodules of gray matter intensity in the wall of
compressed or they may extend into adjacent white matter (Fig. 5-72). the lateral ventricle (Fig. 5-74). As in the postnatal diagnosis, they are
They are differentiated from the subependymal hamartomas of tuberous often associated with other anomalies such as mega cisterna magna,
sclerosis (see Chapter 6) by (a) their shape (hamartomas of tuberous cephalocele, or callosal anomalies. It may be difficult to differentiate
sclerosis are irregular and their long axis tends to be perpendicular to heterotopia from remaining areas of germinal matrix. In this regard,
the adjacent wall of the ventricle [544–546]), (b) their signal intensity it is important to remember that the germinal zone involutes early in
(subependymal hamartomas of tuberous sclerosis are usually isoin- the posterior portions of the ventricles; therefore, residual nodules in
tense to hypointense compared to mature white matter [239,546], not the walls of the trigones, a common location for PVNH (Fig. 5-74), are
isointense with gray matter), and (c) the fact that they do not enhance likely to be heterotopia. If uncertain, a follow-up MR examination in
after intravenous infusion of paramagnetic contrast (239,544). PVNH 2 to 3 weeks may be useful.

Barkovich_Chap05.indd 430 5/6/2011 8:59:33 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 431

PVNH are sometimes associated with ventriculomegaly and, as a


result, affected fetuses may be detected as abnormal on routine fetal
sonography. More and more, these fetuses are referred for fetal MRI to
look for the cause of the ventricular enlargement (550). The walls of
the lateral ventricles should be closely scrutinized on all such studies
to look for nodules of gray matter intensity. The presence of associated
mega cisterna magna increases the likelihood of finding PVNH.
Rarely, periventricular heterotopia are smooth and linear, con-
forming to the contour of the wall of the lateral ventricle (Fig. 5-75).
The significance of these heterotopia and their relationship to PVNH
(or to band heterotopia) are unclear.

Focal Subcortical Heterotopia Focal subcortical heterotopia is the


name given to large, swirling, curvilinear masses of mixed gray mat-
ter and white matter, extending from cortex to ventricle, and often
containing blood vessels and CSF (possibly from subarachnoid space)
(528,552). Patients with focal subcortical heterotopia have variable
motor and intellectual disturbances, depending upon the volume and
the effect on the overlying cortex. Children with bilateral, large, thick
subcortical heterotopia present with moderate to severe developmental
delay and motor dysfunction. Those with large unilateral heterotopia
have hemiplegia and less severe (if any) mental retardation; children
FIG. 5-73. X-linked periventricular heterotopia in a male. Axial T2-
weighted image shows heterotopia (small black arrows) completely lining
with small or thin unilateral subcortical heterotopia may have normal
the ventricles. Foci of dysmorphic cortex (large black arrows) are present. motor function and normal development (528,552). Almost all affected

FIG. 5-74. Fetal MRI of periventricular nodular hetero-


topia. Parasagittal (A), axial (B), and coronal (C) images
show the heterotopia (white arrows) as nodules of gray-
matter intensity protruding into the ventricular lumen.

Barkovich_Chap05.indd 431 5/6/2011 8:59:34 PM


432 Pediatric Neuroimaging

FIG. 5-75. Periventricular linear heterotopia. Coronal (A) and axial (B) images show a smooth, curvilinear layer of gray
matter (arrows) lining the walls of the lateral ventricles. The relationship of these linear heterotopia to nodular subependymal
heterotopia and to band heterotopia is unclear.

patients eventually develop epilepsy, usually during the first or second heterotopia may appear to exert mass effect on the adjacent ventricle
decades (528,552). Some early reports suggested that surgical resection or the interhemispheric fissure (Fig. 5-77) and, thus, may be mistaken
may be useful in patients with subcortical heterotopia and medically for tumors. Further scrutiny of the images, however, will show that the
refractory epilepsy (553). affected region of the hemisphere is small and that the apparent mass
On neuroimaging, focal subcortical heterotopia appear as large, effect is actually distortion of the hemisphere caused by the dysplasia.
somewhat heterogeneous masses that are isointense to cortical gray Thus, heterotopia can be differentiated from tumors, which will enlarge
matter on all sequences. They sometimes appear as multinodular gray the affected hemisphere and, other than compression from mass effect,
matter masses (Fig. 5-76) and other times appear to be composed of has normal overlying cortex. Other important features in distinguishing
swirling, curvilinear bands of gray matter (Fig. 5-77). The portion heterotopia from tumors are that heterotopia lack surrounding edema,
of the hemisphere that is affected is almost always small compared they remain isointense with gray matter on all imaging sequences and
with the contralateral side, and the overlying cortex is thin with shal- with all imaging modalities, and they do not enhance after adminis-
low sulci, often resembling PMG (Figs. 5-76 and 5-77). Subcortical tration of contrast agents (487,527,528). Associated brain anomalies

FIG. 5-76. Small area of subcortical heterotopia. Axial (A) and coronal (B) T1-weighted images show gray matter (black
arrows) extending from the wall of the trigone to areas of abnormal cortex (black arrowheads). The coronal image (B) shows
the multiple small nodules of gray matter that compose the lesion. Note that the affected portion of the hemisphere is small
and the overlying cortex has abnormal sulcation, both typical of subcortical heterotopia.

Barkovich_Chap05.indd 432 5/6/2011 8:59:35 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 433

FIG. 5-77. Large area of subcortical heterotopia. Axial (A) and coronal (B) T2-weighted images show large curvilinear,
swirling masses of gray matter (white arrows) extending from the ventricular surface to the cortex in the middle third
of the right cerebral hemisphere. The overlying cortex is thin and has areas of abnormally deep sulcation (white arrow-
heads). The affected hemisphere is small. (These images courtesy Dr. Gilbert Vezina, Washington, D.C.)

are common; callosal agenesis or hypogenesis is present in up to 70%, with an associated derangement of sulcation; in areas of PMG, no nor-
and the ipsilateral basal ganglia are dysmorphic in greater than 70% mal sulci are seen.
(552). Subcortical heterotopia will sometimes appear to contain ves- Patients with PMG may present with developmental delay, focal
sels or fluid, thereby mimicking tumors. Closer examination in these neurologic signs and symptoms, or epilepsy, depending upon the
cases will show that these are cerebrospinal fluid and vessels coursing portion(s) of brain involved (556). It may be associated with con-
inward from the cerebral cortex (528). The embryological mechanism genital infection (557,558) (also see Chapter 11), in utero ischemia
by which the CSF and vessels get into the center of the cerebral hemi- (559,560), or chromosomal mutations (many have been identified
sphere is unclear. [200,209,448,561–570]). No difference in neurologic manifestations
Occasionally, regions of heterotopia course radially through the has been detected in patients who have PMG due to different causes
cerebral mantle from the cortex to the ventricular surface (Fig. 5-78). (200,557,571,572). Affected patients may present at any age. The
These patients almost always come to medical attention because of severity of the clinical presentation depends upon the extent of corti-
epilepsy; occasionally, they have fixed neurological deficits as well, with cal involvement; bilateral involvement and involvement of more than
the type of deficit depending on the location of the lesion. These trans-
mantle heterotopia can be differentiated from closed lip schizencephaly
by the absence of a dimple at the ventricular surface, indicating the
absence of a cleft between two tightly apposed walls of a schizenceph-
alic cleft. The relationship of transmantle heterotopia to schizencephaly
and to transmantle dysplasias (FCD Type II) is being investigated.
No fetal diagnoses of subcortical heterotopia have been reported.
Marsh et al. (554) reported that proton spectroscopy of hetero-
topia shows elevated creatine and choline with normal NAA. Li et al.
(310) found that the NAA/Cr ratio is variable, ranging from normal to
low compared with age matched normal controls. In our limited expe-
rience, perfusion of heterotopia is similar to that of cerebral cortex.

Malformations Secondary to Abnormal Cortical Organization


and Late Migration
Polymicrogyria PMG is a malformation of cortical development in
which the process of normal cortical development is interrupted dur-
ing the late stages of neuronal migration and during the stages of corti-
cal organization. The result is the abnormal development of the deeper
layers of the cerebral cortex and the formation of multiple small gyri. FIG. 5-78. Transmantle heterotopia. Coronal 3D GRE image shows a linear
Thus, it is classified as a disorder of cortical organization (7). PMG has focus of gray matter (black arrows) extending from the cortex to the ventric-
a range of histologic appearances, all having in common a derange- ular surface. No dimple is seen at the ventricular surface to suggest the pres-
ment of the normal six-layered lamination of the cortex (190,336,555) ence of a cleft (which would be indicative of closed-lip schizencephaly).

Barkovich_Chap05.indd 433 5/6/2011 8:59:36 PM


434 Pediatric Neuroimaging

half of a single hemisphere are poor prognostic indicators, portending reason, it is important to realize that both of these cortical appearances
moderate to severe developmental delay and significant motor dys- can be seen in patients with PMG.
function (572). The malformation may be focal, multifocal, or dif- Regions of PMG can be flat and congruent to the arc of normal
fuse; it may be unilateral, bilateral, and asymmetrical, or bilateral and cortex (Fig. 5-79), or may extend centripetally, with the cortex appear-
symmetrical. The Sylvian fissure is affected in 60% to 80% of cases ing as if it were buckled or folded inward (Figs. 5-82 and 5-83). The
(556,573); however, any area, including the frontal, occipital, and infolding of cortex may be small or large; the character of the cortex
temporal lobes, can be involved (190,571–575). Associated brain mal- in these regions of infolding is similar to that in superficial PMG, with
formations are common and include corpus callosum agenesis and bumpy, irregular inner and outer cortical surfaces. PMG may be unilat-
hypogenesis, cerebellar hypoplasia (9), PVNH (531), and subcortical eral (∼40%) or bilateral (∼60%). As stated above, the cortex s urround-
heterotopia (576). Affected patients may be microcephalic, normo- ing the sylvian fissures is involved in up to 60% to 80% of cases, with
cephalic, or macrocephalic (200). the frontal lobe being most commonly involved (∼70%), followed by
parietal (63%), temporal (38%), and occipital (7%) lobes (575). The
Imaging of Polymicrogyria PMG has a variable appearance on striate cortex, cingulate gyrus, hippocampus, and gyrus rectus are typi-
MRI studies (577–579). The pattern depends partially upon the plane cally spared (575). Rarely, in less than 5% of affected patients, poly-
of imaging, the thickness of the slices and the stage of myelination microgyric cortex will be calcified; this is likely dystrophic in nature,
of the brain at the time of the imaging study, but the variability also related to prenatal injury or infection or, possibly, to repeated seizure
reflects different morphologies of PMG (579). The most common activity. Finally, it is important to realize that anomalous venous drain-
appearance is a slightly thick cortex with cortical surface an irregu- age is common in areas of dysplastic cortex (580), seen in up to 51%
larly bumpy outer and inner surface; high resolution images can some- (575). Large vessels are especially common in regions where there is a
times resolve individual gyri in these cases (Fig. 5-79B). However, the large infolding of thickened cortex (Fig. 5-83). Such large vessels, when
outer surface may be paradoxically smooth because the outer cortical seen in association with abnormal, thickened cortex, should not be
(molecular) layer fuses over the microsulci (Fig. 5-79A). The affected mistaken for vascular malformations. Angiography is not indicated.
cortex may appear thick and coarse (Fig. 5-79) or fine and delicate On fetal MRI, PMG can be detected by the appearance of shal-
(Figs. 5-80 and 5-81). When the microgyri are small, PMG may be low sulci earlier than expected (Fig. 5-84, and see list of timing of nor-
missed or misdiagnosed on routine spin-echo images. Images with mal sulcal development in Chapter 2) and in unexpected locations. In
thin sections and optimal gray matter-white matter contrast (we use our experience, this is the earliest finding of PMG; a previous injury
volume 3DFT spoiled gradient acquisition [T1 weighted] and volume may have been detected at the site. Later in development, many small
3DFT fast spin-echo [T2-weighted] images, both with ≤1.5 mm parti- gyri and sulci may be seen (Fig. 5-84). In bilateral PMG syndromes,
tion size) are obtained. Evaluation in three planes is often necessary appearance of normal sulci is delayed and shallow abnormal sulci may
to detect irregularities of the gray matter-white matter junction, which develop. The normal “squaring” of the sylvian fissure that normally
are often the only evidence of dysmorphic brain (Fig. 5-81) (453). The develops between about 21 and 24 gestational weeks (see Chapter 2)
volume acquisitions can be displayed as 3D images and can be utilized does not occur if the sylvian region is affected by the PMG.
for stereotactic localization, aiding in surgical therapy, if appropriate.
The degree of myelination also has an effect on appearance. In unmy- Syndromes Associated with Polymicrogyria
elinated regions, the polymicrogyric cortex looks thin (2–3 mm) and Bilateral Perisylvian Polymicrogyria. Several specific syndromes
bumpy, while in myelinated areas it looks thicker (5–8 mm) and rela- are associated with cerebral PMG. Kuzniecky et al. first described the
tively smooth (577). The reason proposed for this is that a 4 to 5 mm syndrome of bilateral perisylvian PMG (also called congenital bilat-
layer of gliotic white matter runs through the polymicrogyric cortex, eral perisylvian syndrome [581]), which is the most common PMG
blending in with white matter in the unmyelinated brain and blend- syndrome. This syndrome may be sporadic or familial (564,582).
ing in with cortex after myelination (Fig. 5-82) (577). Whatever the The inheritance patterns appear to be heterogeneous, suggesting that

FIG. 5-79. Coarse focal polymicrogyria. Axial


T2-weighted images show left posterior perisylvian
polymicrogyria extending from the insular cortex
posteriorly into the temporal operculum (white
arrows). Note how the high resolution images allow
resolution of some individual microgyri (white
arrowheads in B), but some microgyri are so closely
apposed that the cortex appears paradoxically
smooth (black arrowheads in A).

Barkovich_Chap05.indd 434 5/6/2011 8:59:37 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 435

FIG. 5-80. Delicate focal polymicrogyria. A. Parasagittal


T1-weighted image shows thin, delicate PMG throughout the
lateral left frontal lobe. B and C. Axial T1-weighted (B) and
coronal T2-weighted (C) images show the delicate irregular-
ity of the individual gyri (white arrows) over the left frontal
convexity.

FIG. 5-81. Thin section axial T2-weighted image shows delicate polymicro-
gyria (arrows) localized to the right circular sulcus of an infant.

Barkovich_Chap05.indd 435 5/6/2011 8:59:38 PM


436 Pediatric Neuroimaging

FIG. 5-82. Evolution of MR appearance of polymicrogyria. A. Axial T2-weighted image at age 2 months. Polymicrogyria is seen in much of the right hemi-
sphere. In the right parietal infolding of cortex (white arrow), the undulation of the cortex can be seen. B. Axial T2-weighted image at age 10 months shows
increased myelination of the brain. The right parietal infolding (white arrow) of cortex now shows a linear hyperintensity near the cortical-white matter
junction. The undulation of the cortex is no longer apparent. C. Axial T2-weighted image at age 3 years shows the infolding (white arrow) to be composed
of apparently thickened cortex without obvious undulations.

FIG. 5-83. Infolding of polymicrogyria with anomalous venous


drainage. A. Parasagittal T1-weighted image shows infolding of
cortex (black arrows) in left posterior frontal lobe. B and C. Axial
precontrast and postcontrast T1-weighted images show large
venous structures (white arrows) draining from the abnormal
sulcus (black arrows).

Barkovich_Chap05.indd 436 5/6/2011 8:59:39 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 437

FIG. 5-84. Fetal MRI of polymicrogyria. Coronal image (A) at 23 gestational weeks shows small gyri (white arrows) in the right
frontal lobe; the anterior frontal lobe should be agyric at this age. Axial image (B) of another fetus at 31 weeks shows microgyri (white
arrows) over the left frontal convexity. Compare to normal gyri in the contralateral hemisphere.

mutations of several different genes can cause this malformation MRI of bilateral perisylvian PMG typically has a coarse PMG
(582); three locations (Xq21.33-q23 [SRPX2], 22q11.2, and Xq28) have appearance, but the appearance of this malformation varies consider-
been identified (200,211,562,568). Sporadic cases with severe involve- ably with severity. Bilateral perisylvian PMG can be graded according to
ment may present with a syndrome of developmental pseudobulbar severity (with grade 1 the most severe and grade 4 the mildest) (587):
palsy (oropharyngeal dysfunction and dysarthria, 100%), epilepsy
(80%–90%), mental retardation (50%–80%), and, sometimes, congeni- Grade 1, with perisylvian PMG extending to the frontal or occipital
tal arthrogryposis (581,583–585). Less severely affected patients present pole (Fig. 5-85);
in infancy or early childhood with developmental delay (60%), poor pal- Grade 2, with PMG extending beyond the perisylvian region but not
atal function (40%), hypotonia (30%), arthrogryposis (30%), or motor to either pole (Fig. 5-86);
deficits (25%) (584,586); seizures (many clinical types) are present in Grade 3, with PMG of the perisylvian region only (Fig. 5-87);
40% to 60% (584,586). Studies of familial cases of congenital bilateral Grade 4, with PMG restricted to the posterior perisylvian regions.
perisylvian PMG show a lower incidence of these clinical manifestations
(582,587), possibly because patients with minimal symptoms are more Other Bilateral Polymicrogyria Syndromes. Other syndromes of
readily identified and examined. Developmental reading disorder and bilateral symmetrical PMG have been described (578). Several groups
reading impairment without severe motor or cognitive handicap may have described bilateral symmetrical frontal polymicrogyria (Fig. 5-88)
be the cause of seeking medical attention in this group (588). (589). These patients typically present with spastic quadriparesis and

FIG. 5-85. Grade 1 bilateral perisyl-


vian polymicrogyria. Sagittal (A) and
axial (B) images show course poly-
microgyria centered in both sylvian
fissures and extending to the occipital
poles.

Barkovich_Chap05.indd 437 5/6/2011 8:59:41 PM


438 Pediatric Neuroimaging

FIG. 5-86. Grade 2 bilateral perisylvian polymicrogyria. Axial T2-weighted images (A and B) show extensive
coarse polymicrogyria centered in both sylvian fissures but sparing the frontal and occipital poles.

epilepsy. Guerrini et al. (574,590) have described patients with bilateral as a malformation secondary to abnormal pial basement membrane
medial parietaloccipital polymicrogyria (Fig. 5-89). The authors have formation (405) and has been discussed in that section.
seen cases of bilateral lateral parietal PMG and many with combinations
of the above-mentioned patterns; thus, it appears that any region of cor- Epilepsy Syndromes Associated with Polymicrogyria. Epilepsy
tex may be involved by bilateral, symmetrical PMG (578). A syndrome syndromes can also be associated with PMG (571,572,574,583,592,593),
of congenital hemiplegia and epilepsy has been described in patients including Aicardi syndrome, which was discussed in section “Anom-
with large areas of unilateral PMG (591). These patients typically pres- alies of the Cerebral Commissures” on callosal anomalies, oculo-
ent in infancy with delayed motor development. A familial syndrome cerebral-cutaneous syndrome (Delleman syndrome [246,247], which
of unilateral polymicrogyria has also been described (130). In contra- has frontal predominant PMG, PVNH, enlarged lateral ventricles or
distinction to the coarse pattern of PMG in bilateral perisylvian PMG, hydrocephalus, agenesis of the corpus callosum sometimes associ-
bilateral frontal PMG and bilateral parasagittal parietooccipital PMG ated with interhemispheric cysts, a large, dysplastic midbrain tectum,
have more delicate PMG patterns. A different imaging appearance is absent cerebellar vermis, and small cerebellar hemisphere), DiGeorge
seen in BFPP secondary to GPR56 mutations, which is better classified syndrome (also called the 22q11.2 deletion syndrome in which several

FIG. 5-87. Grade 3 bilateral perisylvian polymicrogyria. Axial T1-weighted images show the polymicrogyria
restricted to the sylvian fissures and immediate suprasylvian areas.

Barkovich_Chap05.indd 438 5/6/2011 8:59:42 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 439

FIG. 5-88. Bilateral frontal polymicrogyria.


Axial T1-weighted images show delicate poly-
microgyria (white arrows) extending through
the entirety of the frontal lobes.

genes are deleted resulting in cardiac, renal, and parathyroid anomalies Megalencephaly Syndromes with Polymicrogyria. A syndrome of
as well as right > left PMG [211]), and Warburg Micro syndrome megalencephaly with polymicrogyria and hydrocephalus has been
(microcephaly, microcornea, congenital cataract, mental retardation, described. Affected children present at birth or in early infancy with
optic atrophy, and hypogenitalism with hypotonia, callosal hypo- or macrocephaly and hypotonia, and show delayed motor and cogni-
agenesis, and frontoparietal PMG [268–270]). It is interesting to note tive development. Epilepsy, postaxial polydactyly, cutis marmorata,
that the epileptogenic focus is typically not within the polymicrogyric midface capillary malformation, and coarse facial features are fre-
cortex itself, but in the cortex adjacent to the PMG, known as the para- quently present (595–599). Imaging (Fig. 5-90) reveals ventriculo-
microgyral zone (594). Studies in laboratory animals show increases in megaly (from progressive hydrocephalus) and PMG, which is most
postsynaptic glutamate (excitatory) receptors and decreases in GABAA severe in the perisylvian region (typically Grade 1 or 2). Progres-
(inhibitory) receptors in the paramicrogyric cortex (594), factors that sive tonsillar herniation often accompanies the progressive hydro-
likely promote epileptogenesis. In addition, the polymicrogyric cortex cephalus (600); therefore, sequential MR studies may be indicated
appears to have fewer axonal connections with the rest of the brain (600). This disorder is now thought to include those previously
(594). This lack of connections may be the reason why PMG cortex called megalencephaly-polymicrogyria-polydactyly-hydrocephalus
does not function normally and why seizures emanate from the para- syndrome and macrocephaly capillary malformation syndromes
microgyral zone instead of the dysplastic cortex itself. (597–599).

FIG. 5-89. Parasagittal parietooccipital polymicrogyria. Parasagittal (A) and axial (B) T1-weighted images show polymi-
crogyria (black arrows) coursing through the parasagittal parietal white matter.

Barkovich_Chap05.indd 439 5/6/2011 8:59:43 PM


440 Pediatric Neuroimaging

FIG. 5-90. Megalencephaly with polymicrogyria and hydrocephalus in a 4-year-old with delayed development and
seizures. Sagittal T1-weighted image (A) shows enlarged third ventricle and pointed, herniating cerebellar tonsils. Axial
T2-weighted image (B) shows large lateral ventricles and perisylvian polymicrogyria (white arrows).

Megalencephaly, mega corpus callosum, and polymicrogyria macrocephaly and hemiparesis, ultimately developing epilepsy (>80%)
patients have large heads at birth and exhibit rapid head growth such and a mild to moderate developmental delay, depending upon the size
that they continue to increase relative to normals. In addition, they lack and location of the cleft within the brain (610,611,614). Patients with
motor and speech development and have a distinctive facies with frontal bilateral clefts tend to be severely retarded with early onset of epilepsy,
bossing, a low nasal bridge, and large eyes (601). MRI shows bilateral, severe motor anomalies, and, frequently, blindness (610–613). The
symmetrical megalencephaly with a thick corpus callosum, increased blindness probably results from optic nerve hypoplasia, which is seen
volume of white matter, wide sylvian fissures, and diffuse PMG. in up to one third of patients with schizencephaly (487,615–617). The
Proton MR spectroscopy seems to be normal in regions of PMG optic nerve hypoplasia, in conjunction with a high incidence of absence
(310). of the septum pellucidum, results in the classification of many affected
patients in the category of septooptic dysplasia (later section of this
Schizencephaly Schizencephaly, sometimes called agenetic por- chapter) (616). Some authors have noted that epilepsy is less common
encephaly, is the term used to describe gray-matter lined clefts that in bilateral than in unilateral schizencephaly (618). They speculate that
extend through the entire cerebral mantle, from the ependymal lining the bilateral clefts may impair spread of the epileptic discharges. How-
of the lateral ventricles to the pial covering of the cortex (602,603). ever, most reports suggest that seizures begin earlier and have worse
It is classified as a malformation due to abnormal cortical organiza- outcome when clefts are bilateral (610,613).
tion (7) because it is known to be associated with vascular disrup- For prognostic purposes, patients with schizencephaly are divided
tions and because an overwhelming number of cases seem to be into those with unilateral (∼60%) and bilateral clefts (∼40%). The
associated with PMG. Schizencephaly is a rare malformation, with clefts are further divided into those with fused lips (15%–20%) and
a population prevalence of approximately 1.5 per 100,000 popula- those with separated (open) lips; those with separated lips are divided
tion (604). It is associated with young parental age (604). One third into small clefts and large clefts (610,611,614,619). In the clefts with
of affected patients have a non-CNS abnormality, more than half of fused lips, the walls appose one another directly, obliterating the CSF
which may be classified as secondary to vascular disruption, includ- space within the cleft at that point (Fig. 5-91). When the lips are sepa-
ing gastroschisis, bowel atresias, and amniotic band syndrome. Some rated, CSF fills the cleft all the way from the lateral ventricle to the sub-
reported cases were associated with prenatal infection, while others arachnoid spaces surrounding the hemispheres (Figs. 5-92 and 5-93)
had chromosomal aneuploidy and still others were familial, suggest- (602,603,611,617).
ing genetic causes, but no genetic locus was identified (605–608). On pathologic exam, the gray matter lining schizencephalic clefts
Although past reports suggested that the malformation may be asso- is dysmorphic and does not exhibit normal cortical lamination. The
ciated with mutations of the EMX2 homeobox gene, this association clefts can be unilateral or bilateral and are most commonly located
was not verified and more recent studies have questioned it (609); no near the precentral and postcentral gyri (4); in fact, the locations in
genetic basis for familial schizencephaly has been found at the time which they occur are nearly identical to those in which PMG occurs
this chapter was written. Taken together, these observations suggest (572,611).
that schizencephaly has heterogeneous causes, many of which include Imaging studies of schizencephaly show a full thickness CSF-filled
vascular disruption (604). cleft through the affected hemisphere; gray matter, typically charac-
Patients with schizencephaly typically present with seizures, hemi- terized by a bumpy outer surface and an irregular gray matter-white
paresis, or variable developmental delay. The severity of the patients’ matter junction (Fig. 5-92B), lines the cleft (487,488,611,613,617).
symptoms is related to the amount of involved brain (610–613). Those The appearance of schizencephaly on fetal MRI is identical to that on
patients with a single cleft with fused lips generally have epilepsy and, postnatal MRI (Fig. 5-92A). Closed lip schizencephalies may look like
perhaps, a mild hemiparesis but are otherwise developmentally normal. transmantle heterotopia if the walls of the cleft are fused through the
Patients with unilateral clefts with separated lips usually present with entire cortical mantle; indeed, transmantle heterotopia (Fig. 5-78) may

Barkovich_Chap05.indd 440 5/6/2011 8:59:44 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 441

be an extreme closed lip schizencephaly. Although any portion of the


brain can be involved, the frontal horns and the anterior aspects of
the temporal horns are relatively spared (619). When schizencephaly
is bilateral (40%–50%), clefts are nearly (but not exactly) symmetrical
in about 80% (Fig. 5-92), but the size may vary (Fig. 5-93); their loca-
tion is frontal or parietal in approximately 60% to 65% of cases, and
temporal or parietal in approximately 25% to 30%. The clefts are open
bilaterally in approximately 60%, closed bilaterally in approximately
20%, and closed on one side and open on the other in approximately
20% (610,611,619). When schizencephaly is unilateral (50%–60%), the
lips are open in about 65% and closed in about 35%; the location is
mid to posterior frontal or parietal in approximately 80%, temporal
in approximately 10%, and occipital in approximately 10% (610,611).
The gyral pattern of the cortex adjacent to the cleft is usually abnormal,
demonstrating sulci that radiate into the cleft. The gray matter lining
the cleft may extend into the ventricle and resemble PVNH (Fig. 5-92).
If the cleft has narrow open lips or closed lips, a dimple is usually seen
in the wall of the lateral ventricle where the cleft communicates (Fig.
FIG. 5-91. Closed-lip schizencephaly. Coronal T1-weighted image shows 5-91). The dimple is often a helpful sign of continuity of the cleft with
irregular gray matter extending from the cortex to the ventricular surface. the ventricle when the lips of the cleft are fused and helps to differenti-
The lips of the cleft come in contact (white arrows) in only part of the ate schizencephaly from transmantle heterotopia or deep infoldings of
cleft. PMG. Of note, clefts with fused lips might be missed if the imaging plane

FIG. 5-92. Bilateral open-lip schizencephaly with small


clefts. A. Coronal image of 22-week fetus shows bilateral
gray matter-lined clefts (arrows) and absence of the sep-
tum pellucidum. Note the CSF is continuous from the
subarachnoid space to the ventricle. B. Axial T2-weighted
postnatal image shows that the gray matter lining the
clefts is thick and irregular, suggesting polymicrogyria;
these can resemble periventricular nodular heteroto-
pia when they extend into the ventricle (black arrow-
head). The clefts (black arrows) are completely open.
Note expanded calvarium (e) overlying the large cleft.
C. Parasagittal T1-weighted image shows the superior-
to-inferior extend of the cleft (white arrows).

Barkovich_Chap05.indd 441 5/6/2011 8:59:45 PM


442 Pediatric Neuroimaging

The calvarium is often expanded over the opening of an open lip


schizencephaly (Figs. 5-92 and 5-94). The expansion may result from
longstanding CSF pulsations that emanate from the lateral ventricles
and propagate through the cleft. Another possible cause of calvarial
expansion relates to a thin membrane (the “roofing membrane”) that
overlies the cleft at the cerebral surface and separates the ventricular
CSF within the cleft from that in the overlying subarachnoid space
(619); this membrane may expand beyond the cortex, cause localized
pressure on the inner table of the calvarium and slowly expand the
calvarium. When plagiocephaly is severe, the insertion of a ventricu-
loperitoneal shunt may help to dampen the pulsations and partially
reverse the cranial asymmetry.
The appearance of schizencephaly on fetal MR is identical to that
on postnatal MR (Fig. 5-92A). Keep in mind that visualization of the
cerebrum in three planes is especially important when imaging the
small fetal brain.

Focal Cortical Dysplasia Without Dysmorphic Neurons (Minor


Malformations of Cortical Development, Focal Cortical Dyspla-
sia Types I, III) Focal cortical dysplasia without dysmorphic neurons
FIG. 5-93. Coronal T2-weighted image shows bilateral schizencephaly has been divided into three histological groups: mMCD, FCD Type
with large open clefts (white arrows). The black arrowhead points to a ven- I, and FCD Type III (340,344,621). These groups have similar clini-
triculostomy catheter from a shunt. cal and imaging characteristics and can be considered together. Their
causes are unknown, but it is likely that many result from acquired
is parallel to the plane of the cleft. For that reason, imaging should always brain injuries (121), as Type I and mMCD are associated with a sig-
be performed in at least two planes (three planes is best) in patients nificant incidence of prenatal and perinatal adverse events (suggesting
who present with seizures or developmental delay. Dysmorphic cere- that they may be caused by such events [280]), whereas Type III are
bral cortex, most likely PMG, may be present in a “mirror-image” associated with hippocampal sclerosis (FCD Type IIIa), epileptogenic
location in the hemisphere contralateral to a unilateral schizenceph- tumors (DNET and ganglioglioma, FCD Type IIIb), vascular malfor-
aly, particularly when the schizencephaly is large and open-lipped mations (FCD Type IIIc), or scarring from injury due to trauma, por-
(Fig. 5-94) (572,611); thus, the contralateral hemisphere should always encephaly, or encephalitis (FCD Type IIId) (for more information, see
be scrutinized. The septum pellucidum is absent in approximately 70% [121,280,315,344,621] and references therein). These disorders tend
of patients with schizencephaly and is almost always absent when the to present at older ages than FCD Type II (discussed in the section
clefts are bilateral and frontal; when unilateral, the septum is more “Malformations Secondary to Abnormal Stem Cell Proliferation or
commonly absent when the cleft is open-lipped (Fig. 5-94) rather than Apoptosis” of this chapter), are more often found in adults, have lower
closed-lipped (620). seizure frequencies, and are more commonly found in the temporal
lobe (315).
Type I FCDs can be considered a focal structural abnormality of
the cerebral cortex and, usually, the underlying white matter. Abnor-
mal neurons are not seen, although large pyramidal neurons may
be aberrantly positioned in upper cortical layers; neither balloon
cells nor dysplastic neurons are identified and abnormal cells do not
extend medially to the ventricular surface (338,340,344,353,622,623).
The structural abnormality may be predominantly radially oriented
(FCD Type Ia), horizontally oriented (FCD Type Ib), or both (FCD
Type Ic) (344). Patients with these types of FCD almost always
present with partial seizure disorders and normal neurologic exams
(315).
The MR appearance of Type I FCD is variable, suggesting that
this is a heterogeneous disorder. At least 50% of affected patients have
a normal initial MRI. When the MRI is abnormal, the most common
appearance is that of focal blurring of the cortical-white matter junc-
tion on T1- and T2-weighted images with FLAIR hyperintensity (Fig.
5-95) (280,340). Subcortical white matter may be isointense to cortex,
sometimes because the affected cortex is abnormally hypointense,
but other times (particularly in temporal lobe epilepsy) because the
white matter is abnormally hyperintense on FLAIR and T2-weighted
sequences. Some degree of volume loss (gray matter or white mat-
FIG. 5-94. Axial T2-weighted image shows a large open lip right hemi- ter atrophy) is found in about half of affected patients (Fig. 5-96)
sphere schizencephaly (S) with expansion (black arrowheads) of the overly- (280,315), possibly reflecting the previously mentioned history of
ing calvarium. Note an infolding of polymicrogyria (white arrows) in the late prenatal or perinatal adverse events in some patients (FCD Type
contralateral hemisphere. IIId). As a result, the cortex may appear thin (Figs. 5-96 and 5-97);

Barkovich_Chap05.indd 442 5/6/2011 8:59:46 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 443

FIG. 5-95. Type I focal cortical dysplasia. Coronal T1- (A)


and T2- (B) weighted images show a focal area of blur-
ring of the cortical-white matter junction (white arrows).
Coronal FLAIR image shows hyperintensity (white arrows)
at the cortical-white matter junction.

indeed, thick cortex is unusual. The abnormality is typically subtle Type IIId) or vascular malformations (FCD Type IIIc) (344). Imaging
and the MR scan may be initially interpreted as normal (280,315,624). of FCD Type III may be seen as cortical thinning (FCD Type IIId, Fig.
Careful evaluation with thin sections and optimal gray matter-white 5-96) or as T2/FLAIR hyperintensity of the white matter adjacent to
matter contrast may be needed to show the focal cortical thinning or the hippocampal sclerosis (FCD Type IIIa, Fig. 5-98).
blurring of the cortical-white matter junction (Figs. 5-96 and 5-97) As mentioned in the opening paragraph of this section “Malforma-
(354,625,626). FLAIR images with narrow windows may also be useful tions of Cortical Development,” a number of papers have indicated that
to detect cortical or subcortical hyperintensity; if the brain is incom- functional imaging studies such as magnetic source imaging (MSI, which
pletely myelinated, however, the abnormality is nearly impossible to is MEG coregistered to MRI), PET scanning, or SPECT scanning may be
separate from immature white matter. Moreover, if dyslamination is useful in detecting small areas of FCD (284–289,293,295,304,313,624);
the only abnormality (this is technically an mMCD rather than FCD these studies are particularly useful for FCD Type I where the initial
Type I [342]), even high-resolution MRI may be completely normal MRI is often read as normal (315). As noted earlier, PET and SPECT are
(624). The relative signal intensity of dysplastic cortex and underly- nonspecific and correlative MRI is always needed. Our approach is to
ing white matter may change with age; therefore, serial MR studies use these other techniques in those patients who have some localization
may be necessary to find the focus in young infants (627). to their seizure disorder or their EEG, but a normal initial MR scan. If
As mentioned above, FCD is often associated with other condi- the PET study shows an area of hypometabolism or MSI shows local-
tions; these are now called FCD Type III (344). The best described of ized epileptic spikes, a high-resolution volumetric MR scan (see Chap-
these conditions are hippocampal sclerosis (FCD Type IIIa) (628,629), ter 1 for technique) is performed to generate images in multiple planes
neuronal-glial tumors (FCD Type IIIb, with dysembryoplastic neu- and is coregistered with the functional data for better anatomic local-
roepithelial tumors [DNETs] [630,631] and gangliogliomas [632], see ization. The cortex in that area is then closely scrutinized on the MRI
Chapter 7), and glial scars (ischemic [633] or traumatic [634], FCD for any abnormality of the cortical surface or gray-white junction.

Barkovich_Chap05.indd 443 5/6/2011 8:59:47 PM


444 Pediatric Neuroimaging

FIG. 5-96. Focal atrophy in Type I focal cortical dysplasia. Axial


T1-weighted image (A) shows thinning of the medial frontal cortex
(black arrows) and focal volume loss (note enlarged sulci). Coro-
nal T2-weighted image (B) better shows the volume loss and con-
firms cortical thinning (white arrows). Axial 18F-FDG PET image
(C) shows focally reduced uptake (white arrow) in the anterome-
dial frontal lobe and more generalized anterior frontal decreased
uptake (white arrowheads).

FIG. 5-97. Type I FCD can be extremely small and subtle, requiring thin section imaging in multiple planes. Axial T1 (A) and FLAIR (B) images
show focal blurring of the cortex-white matter junction at the fundus of the right superior frontal sulcus (arrows). Note that the cortex in the affected
area appears thin, with intensity similar to gray matter on the coronal T2-weighted image (C). This finding was missed on four previous MRI scans.

Barkovich_Chap05.indd 444 5/6/2011 8:59:48 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 445

FIG. 5-98. FCD Type III is defined as focal cortical dysplasia adjacent to other epileptogenic pathology such as
hippocampal sclerosis, tumors, or vascular malformations. These coronal FLAIR images show abnormal hyper-
intensity of temporal white matter (white arrows in A and B) adjacent to a small, sclerotic hippocampus (white
arrowhead in B).

ANOMALIES OF VENTRAL frequently seen in the more severe forms (638–641). The embryogenesis
PROSENCEPHALON DEVELOPMENT of holoprosencephaly has not been clearly elucidated. Mutations of
at least ten different genetic loci have been implicated in the develop-
Holoprosencephaly
ment of familial holoprosencephaly: HPE1 on chromosome 21q22.3
The holoprosencephalies are a group of disorders that are characterized (642); HPE2 (SIX3) on 2p21 (643); HPE3 (SHH) on 7q36 (644); HPE4
by a failure of differentiation and midline cleavage of the prosencepha- (TGIF) on 18p (636); and HPE5 (ZIC2) on 13q32 (645), HPE6 at 2q37
lon. In the normal embryo, a single prosencephalic vesicle is established (646), HPE7 (PTCH1) at 9q22.3 (647), HPE8 at 14q13 (648), HPE9
at the rostral end of the neural tube between days 22 and 24 of gestation (GLI12) at 2q14 (649). All of these genes seem to be involved in dorsal
by closure of the anterior neuropore. Fate maps of the early prosen- or ventral induction of the prosencephalon. Additional chromosomal
cephalon show that regions that will become the future structures of regions (on chromosomes 1, 3, 5, 6, and 20) have been implicated, as
the prosencephalon (the deep gray nuclei, cerebral cortex, optic nerves, well (636), supporting the genetic heterogeneity of this disorder. Muta-
olfactory regions, etc.) are already established before the neural tube tions are found far less commonly in sporadic cases of HPE (636,650).
closes. Those structures that will normally form the most rostral medial Familial cases of HPE are very heterogeneous; individuals carrying the
portions of the brain fail to form in holoprosencephaly (635). As a same mutation may range from very severe to clinically normal due to
result, the portions of the brain that are normally located more laterally the influence of environmental or teratogenic factors such as alcohol,
lie close to or within the midline and may remain unseparated across diabetes, cholesterol, retinoic acid, or modifier genes (651).
the rostral midline of the brain. In these locations (most commonly Mutations of human sonic hedgehog gene (SHH) at the HPE3 locus
the anterior prosencephalon and the third ventricle), the interhemi- (652), which cause an autosomal dominant form of holoprosencephaly
spheric fissure fails to form and, as a result, the cerebrum fails to divide (653), have been studied most extensively (636,654). SHH is a protein
into distinct cerebral hemispheres; in addition, the medial aspects of that has a role in maintenance of the notochord; as the notochord is
the hemispheres have failed to form so the structures that are normally essential for the production of prechordal mesenchyme (the prechordal
positioned more laterally are in the midline, not separated from identi- plate) and induction of the ventral forebrain, this discovery supports
cal structures of the contralateral hemisphere. In mild cases, cells that the concept that defects of dorso-ventral induction can cause holo-
would normally be positioned slightly off of the midline are formed, prosencephaly. Also well-studied are mutations of dorsalizing factors
but lie closer to the midline than normal. In severe cases, all structures (chemicals that induce development of the dorsal midline, such as the
normally destined to be anywhere close to the rostral midline remain interhemispheric fissure and choroid plexuses) such as bone morpho-
unformed and only a small ball of rostral telencephalic tissue forms. In genic proteins (BMPs) and the ZIC2 gene (645,655–657). Of note, both
these very severe cases, the premaxillary segments of the face are not overexpression of ventralizing factors and underexpression of dorsalizing
induced to form, either, with the result being arrhinia, midline facial factors have been shown to impair dorsal induction (658). Indeed, vari-
clefts, and hypotelorism (with cyclopia in extreme cases). ous mutations of ZIC2 are associated with all types of HPE: disturbances
Holoprosencephaly is caused by both teratogens and genetic fac- of the BMP pathway and ZIC2 mutations that impair roof plate develop-
tors (636). Holoprosencephaly can be seen in congenital anomaly syn- ment cause the MIH variant of HPE (67) (a disturbance of dorsal induc-
dromes (Smith-Lemli-Opitz and Pallister-Hall syndromes are among tion), while classic HPE is caused by different mutations of ZIC2 that
the best known); up to 45% of affected patients have clear cytogenetic result in impaired formation of the prechordal plate (659) (a disturbance
abnormalities such as Patau syndrome (trisomy 13), Edwards syn- of ventral induction). Thus, normal brain development results from a
drome (trisomy 18, less common), and triploidy (637). Facial dys- balance of normal dorsalizing and ventralizing factors; HPE results from
morphism, particularly hypotelorism and midline facial clefts, are disturbances of this balance in the developing prosencephalon.

Barkovich_Chap05.indd 445 5/6/2011 8:59:50 PM


446 Pediatric Neuroimaging

Clinical presentation depends upon the severity of the malforma- severe neurologic disorders, manifesting seizures, abnormal neonatal
tion. Severe cases are investigated at birth due to facial dysmorphism, reflexes, and abnormalities of tone from the time of birth, in addition
macrocephaly (from hydrocephalus), microcephaly, or neonatal to severe midline facial deformities and hypotelorism, resulting from
seizures. Less severe cases may be studied because of epilepsy, move- absence or hypoplasia of the premaxillary segment of the face. In the
ment disorder, or developmental delay. extreme forms, the orbits and globes are fused, resulting in cyclopia
DeMyer has divided holoprosencephaly into three subcategories in (664,665).
which the most severe malformation affects the ventral prosencepha- Imaging studies are severely abnormal, as well. In affected patients,
lon: alobar, semilobar, and lobar (640,641). These categories are useful the ventral medial portions of the brain (the anterior, inferior parts of
for classifying holoprosencephalies of different severities, (although the frontal lobes, caudates, hypothalami, and the basal ganglia) and
the divisions among these categories are far from distinct [635]). face (nasal bones, medial aspects of maxilla, ethmoids, and vomer)
MIH, also called syntelencephaly, is a variant in which the dorsal pros- never form. As a result, the hypothalami and basal ganglia are fused and
encephalon is more severely affected (66,660). The reader should be reduced in volume and the third ventricle is absent (Fig. 5-99) (635). No
aware, however, that the holoprosencephalies represent a continuum interhemispheric fissure, falx cerebri, or corpus callosum can be identi-
of forebrain malformations, that no clear distinction among the dif- fied. Most commonly, the cerebrum is composed of a pancake-like mass
ferent categories exists, and that other classification systems have been of tissue in the rostral-most portion of the calvarium. No sylvian fis-
suggested (66,616,635,638,640,641,660). Indeed, the authors have seen sures can be identified in most cases and, when present, these fissures
a lack of cerebral hemispheric separation at nearly every location in the lie in the most anterior portion of the brain close to the midline (666).
telencephalon, as well as some that result in lack of separation of the A crescent shaped holoventricle (i.e., no septum pellucidum or third
cerebellar hemispheres (rhombencephalosynapsis, section “Arrhinia/ ventricle) is continuous with a large dorsal cyst, which usually occupies
Arrhinencephaly” of this chapter), as well. most of the volume of the calvarium (Fig. 5-99). Alobar HPE is easily
recognized on fetal MRI or sonography by the absence of the septum
Classic Holoprosencephaly pellucidum and absent interhemispheric fissure (Fig. 5-99A and B). The
In classic holoprosencephaly, the most anterior, ventral aspects of the vascular supply to the cerebrum consists of multiple small vessels aris-
prosencephalon (anterior frontal lobe, hypothalamus, caudate heads, ing directly from the internal carotid and basilar arteries. In less severe
third ventricle) are most severely affected while the more posterior, cases, discrete anterior and middle cerebral arteries may be identified.
dorsal aspects (parietal and occipital lobes) are least affected. The anterior cerebral arteries in these cases are nearly always azygous,
with a single arterial trunk supplying branches to the midline sections
Alobar Holoprosencephaly of the brain.
Although alobar holoprosencephaly is the most common form of hol-
oprosencephaly diagnosed by prenatal ultrasound (661,662), affected Semilobar Holoprosencephaly
patients are rarely imaged postnatally by CT or MRI. This rarity prob- In the semilobar form of holoprosencephaly, the brain is more devel-
ably stems from the fact that most affected infants are stillborn and oped than in the alobar form and the facial anomalies are mild or absent.
others have a very short life span (663). Alobar holoprosencephaly is Affected children may be referred for microcephaly, macrocephaly
the most severe form of holoprosencephaly. Most affected children have (usually associated with a dorsal cyst), or developmental delay. Motor

FIG. 5-99. Severe fetal alobar holoprosencephaly. Sagittal (A) and axial (B) fetal MRI at 22 weeks gestational age
show a small crescent of brain (white arrows) in the anterior calvarium, containing a large monoventricle (v) and
continuous with a large dorsal cyst (c).

Barkovich_Chap05.indd 446 5/6/2011 8:59:50 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 447

FIG. 5-99. (Continued) Sagittal (C), coronal (D), and axial (E) postnatal
images show similar findings, with the exception that some sulcation has
occurred in the cerebral cortex. Note the small, fused mass of deep gray
matter in the ventral midline of (D).

abnormalities typically consist of upper extremity choreoathetosis and gradient of development in which the separation of the hemispheres,
lower extremity spasticity (667,668). development of the falx cerebri, and development of the cerebral
Imaging studies reveal that the interhemispheric fissure and hemispheres progress from the occipital pole to the frontal pole of
falx cerebri are partially formed posteriorly; however, the anterior the cerebrum. The anterior extent of corpus callosum development
(frontal) regions of the brain remain continuous across the midline correlates with the anterior extent of interhemispheric fissure forma-
and underdeveloped (Figs. 5-100 to 5-102). Rudimentary temporal tion (65). Because the severity of classic holoprosencephaly is related
horn formation is seen, although the hippocampus is incompletely to how completely the ventral/anterior (hypothalamus and low fron-
formed (Fig. 5-100). The septum pellucidum is absent in all forms of tal) regions of the brain are developed, the anterior extent of callosal
holoprosencephaly. The frontal lobes are poorly developed and the formation, which reflects the anterior extent of cerebral develop-
sylvian fissures are positioned abnormally anteriorly in the cerebrum ment, can be used as an approximate marker of brain development
(Figs. 5-101 and 5-102) (666). The callosal splenium is present with- in holoprosencephalic patients. In other words, the further anterior
out a callosal body or genu in many patients with semilobar holo- the corpus forms, the better developed the brain. The deep cerebral
prosencephaly (Figs. 5-102) (57,63,638). Holoprosencephaly is the nuclei are typically partially separated, but the hypothalami, caudate
only brain anomaly described in which the posterior corpus callosum heads, and thalami remain partly unseparated, resulting in a small
forms in the absence of any anterior callosal formation. As the spec- third ventricle (Figs. 5-101 and 5-102). In the most severe cases, the
trum of holoprosencephaly is observed from the most poorly differ- central gray matter may be significantly reduced in volume and fused
entiated alobar brain to the most differentiated lobar brain, we see a into a mass (Fig. 5-100) (635).

Barkovich_Chap05.indd 447 5/6/2011 8:59:51 PM


448 Pediatric Neuroimaging

FIG. 5-100. Severe semilobar holoprosencephaly with thalamic fusion and dorsal cyst.
Midline sagittal transfontanelle sonogram (A) shows an abnormal gyral pattern, presence
of a callosal splenium (small white arrows), and a moderate-sized dorsal cyst (large white
arrows). Note that p denotes posterior and a denotes anterior. Coronal transfontanelle image
(B) through the anterior cerebrum shows a large gray matter mass (white arrows) consisting
of fused thalami and basal ganglia. No interhemispheric fissure or ventricle is present at this
level. Coronal transfontanelle image more posteriorly (C) shows the central gray matter mass
separating the lateral ventricles. A rudimentary interhemispheric fissure (open white arrows)
is present. Axial noncontrast CT image (D) shows the central gray matter mass separating
rudimentary temporal horns. No interhemispheric fissure is seen. CT image at a higher level
(E) shows the relationship of the dorsal cyst (white arrows) to the lateral ventricles. Axial
T1-weighted images (F and G) after shunting of the cyst shows the brain with the appearance
of a ball.

Barkovich_Chap05.indd 448 5/6/2011 8:59:52 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 449

FIG. 5-100. (Continued)

On fetal MRI, semilobar holoprosencephaly is identified better if horn formation is present, and the posterior half of the callosal body is
the fetus is more mature; 20 weeks is too early to see the absence of the formed (in addition to the splenium), holoprosencephaly can be classified
anterior portions of the corpus callosum and the lack of separation as lobar (Fig. 5-104).
of the anterior-ventral cerebral hemispheres, but these features and As the abnormalities in lobar holoprosencephaly are rather subtle
the small frontal lobes may be detected by 23 weeks (Fig. 5-102A–C). and are confined to the ventral/anterior portions of the cerebrum, this
Absence of the septum pellucidum may not be identified unless the diagnosis is difficult on fetal MRI unless thin (3 mm), high quality
ventricles are enlarged. coronal images are obtained through the anterior portions of the cere-
Dorsal cysts are sometimes seen in semilobar holoprosencephaly, brum to identify the abnormal configuration of the frontal horns of
almost always when thalamic fusion is present (Figs. 5-100 and 5-101) the lateral ventricles, absence of the septum pellucidum and the lack of
(669). In all likelihood, a combination of thalamic fusion and aqueduc- separation of the ventral portions of the frontal lobes and hypothala-
tal dysgenesis prevents egress of CSF into the fourth ventricle in these mus (Fig. 5-103).
patients; the dorsal cyst thus represents a dilated pineal or suprapineal
recess of the fourth ventricle. After shunting of the cyst, the brain takes Syntelencephaly (Middle Interhemispheric Variant)
on the appearance of a ball (Fig. 5-100) (638–641,670). Myelination is
In some patients, the interhemispheric fissure is formed in the anterior
typically delayed (671).
frontal and in the occipital lobes, but the hemispheres are fused in the
posterior frontal and parietal regions (66). This condition is called MIH
Lobar Holoprosencephaly variant of HPE or syntelencephaly. This malformation also seems to
Patients with lobar holoprosencephaly have less severe clinical mani- result from a defect in dorsal-ventral induction of the telencephalon, but
festations than do patients with alobar or semilobar holoprosen- in MIH, the problem is reduced induction of the dorsal midline struc-
cephaly. Affected patients typically present with mild or moderate tures (67,660) (in contrast to classic HPE in which ventral induction is
developmental delay, hypothalamic-pituitary dysfunction, or visual reduced). Indeed, animal models suggest that failed induction of the roof
problems (667,668). Some patients are initially classified as having plate leads to MIH, possibly as a result of reduced ZIC2 function (ZIC2
“cerebral palsy.” The point at which a brain is identified as being lobar, is a zinc finger gene that is expressed in the dorsal and ventral midline
in contrast to semilobar, holoprosencephaly is poorly defined. The of the developing embryo) or focally increased BMP expression (BMP is
frontal lobes are more fully developed than in semilobar holoprosen- involved in dorso-ventral induction of the embryo) (67). Patients are less
cephaly; as a result, frontal horns of the lateral ventricles are present severely affected clinically than in semilobar or alobar holoprosenceph-
(although they may be dysmorphic) and the sylvian fissures may look aly, usually manifesting mild to moderate cognitive impairment, appen-
normal or nearly normal (666). The deep cerebral nuclei are nearly dicular spasticity, and mild visual impairment (66,672). Choreiform
completely formed (Fig. 5-103) (635). In these patients, the inter- movements and hypothalamic insufficiency, present in most patients
hemispheric fissure and falx cerebri extend into the frontal area of with classic holoprosencephaly, are conspicuously missing from patients
the brain, although the anterior falx may be hypoplastic. The septum with MIH (668). Facial features include normal interocular distance, or
pellucidum is absent, even in mild cases (Fig. 5-104). The hippocampi even hypertelorism, and a broad, flat nose rather than hypotelorism as
are normal or nearly normal and the temporal horns and third ven- in classic HPE (655). Most patients are microcephalic.
tricles are better defined than in the semilobar brains (638–641,670). Imaging in MIH shows normal appearing basal forebrain with
Thus, as a general rule, if the third ventricle is fully formed, some frontal normal basal ganglia and olfactory sulci, and normal appearing

Barkovich_Chap05.indd 449 5/6/2011 8:59:54 PM


450 Pediatric Neuroimaging

FIG. 5-101. Moderately severe semilobar holoprosencephaly.


Sagittal T1-weighted image (A) shows large dorsal cyst with cerebel-
lum inferiorly displaced and compressed. Axial T2-weighted images
(B and C) show that the sylvian fissures (small white arrows) are
abnormally anterior and medially rotated and the frontal lobes are
small. No interhemispheric fissure is present anteriorly. The basal
ganglia and thalami are small and nonseparated in the midline,
resulting in a small third ventricle. At a higher level (C), a crescentic
monventricle has no septum pellucidum.

anterior interhemispheric fissure. The anterior falx cerebri may be


Arrhinia/Arrhinencephaly
normal or slightly hypoplastic. In the posterior frontal or parietal
lobes however, the interhemispheric fissure and falx become dysplas- Absence of the olfactory lobe of the brain, a condition known as arrhi-
tic and then disappear as the two hemispheres become continuous nencephaly, may be isolated or may be associated with anomalies of
across the midline (Fig. 5-105). Sometimes sulci (typically extensions the base of the brain and midline structures of the face (absence of the
of the sylvian fissures) are continuous or nearly continuous across nose itself is known as arrhinia) (4). The olfactory sulci and olfactory
the midline, as well (Fig. 5-105D). We have seen one case of MIH bulbs are invariably absent or dysplastic in these cases. In the fetus,
associated with an occipital cephalocele. Fetal MRI of MIH shows olfactory sulci begin to develop at about the 16th gestational week.
absence of the septum pellucidum, absence of the body of the corpus The sulci are just lateral and parallel to the interhemispheric fissure.
callosum, and lack of separation of the cerebral hemispheres in the They remain shallow until 28 to 30 weeks, when they begin to deepen
posterior frontal and anterior parietal lobes (Fig. 5-106) (673). This from posterior to anterior. They can currently be identified by fetal
is one of the few conditions in which the callosal genu and splenium MRI by about the 30th gestational week (Fig. 5-107A and B) (674). The
are formed in the absence of the callosal body (see section “Anomalies olfactory bulbs lie just above the cribriform plates, where they receive
of the Cerebral Commissures” on Callosal Anomalies earlier in this olfactory nerves from the nasal cavities. They lead into the thinner
chapter and Fig. 5-105A). olfactory tracts, which conduct the axons of the secondary olfactory

Barkovich_Chap05.indd 450 5/6/2011 8:59:54 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 451

FIG. 5-102. Relatively mild semilobar holoprosencephaly. A. Midline


sagittal image from fetal MRI shows the presence of a posterior inter-
hemispheric fissure and posterior corpus callosum (white arrows). No
CSF is seen in the anterior midline of the cerebrum. B and C. Axial (B)
and coronal (C) images from fetal MRI show only a very rudimentary
interhemispheric fissure (black arrows) in the anterior cerebrum. Syl-
vian fissures (white arrowheads in B) are rotated anteriorly. No discrete
basal ganglia are seen. D. Midline sagittal T1-weighted postnatal image
from a different patient shows small frontal lobes and the presence of
only a posterior corpus callosum (black arrows). E. Axial T2-weighted
image shows larger, better separated basal ganglia with a tiny remnant
of anterior third ventricle (white arrowhead). Sylvian fissures (white
arrows) are abnormally anterior and medially rotated.

Barkovich_Chap05.indd 451 5/6/2011 8:59:55 PM


452 Pediatric Neuroimaging

FIG. 5-103. Prenatal and postnatal MR images of a patient with severe


lobar holoprosencephaly. A. Coronal image of a 30-week fetus immediately
anterior the frontal horns of the lateral ventricles shows a rudimentary
interhemispheric fissure (black arrows, note also the absence of an anterior
falx cerebri) dorsally and lack of separation of the cerebral hemispheres
ventrally. B. Axial image shows dysmorphic frontal horns (arrows) and
shallow anterior interhemispheric fissure. C. Postnatal axial T2-weighted
image shows the dysmorphic frontal horns (white arrows), shallow anterior
interhemispheric fissure, and azygous anterior cerebral artery (black arrow).
Note the caudate heads (c) touching in the midline, a finding more common
in semilobar HPE. This illustrates the lack of clear differentiation among the
subtypes of holoprosencephaly.

FIG. 5-104. Mild lobar holoprosencephaly. Midline sagittal T1-weighted image (A) shows the anterior corpus callosum terminating at the superior genu
(black arrow), with absence of the inferior genu and rostrum. Axial T1-weighted image (B) shows continuity of the caudates across the midline (white
arrow). Note also mild hypotelorism. Coronal T1-weighted image (C) shows very narrow frontal horns of the lateral ventricle without septum pellucidum
and lack of separation of the walls of the hypothalamus (white arrows).

Barkovich_Chap05.indd 452 5/6/2011 8:59:56 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 453

FIG. 5-105. Middle interhemispheric variant of holoprosencephaly. Sagittal T1-weighted image (A) shows the presence of a
callosal splenium (white arrows) and genu (white arrowhead) without intervening callosal body. Coronal T2-weighted image
(B) shows continuity of the posterior frontal lobes across the midline without intervening interhemispheric fissure. Note char-
acteristic gray matter (white arrow) on the surface of the narrow, nonseparated lateral ventricles. Axial T2-weighted images
(C and D) show an anterior and posterior interhemispheric fissure with absent interhemispheric fissure in the posterior frontal
lobes. Sylvian fissures, which appear nearly normal in (C), continue upward to meet over the cerebral hemispheres (white
arrows in D).

neurons in the lateral, intermediate, and medial olfactory striae to their retarded growth and mental development, genital anomalies, and
specific terminations within the olfactory cortex. Although the olfac- ear malformation) in an autopsy series (677) and small olfactory
tory bulbs can be first identified by histology at Carnegie stage 18 (44th bulbs with shallow olfactory sulci were seen in all ten patients in
day of development), they cannot be identified by fetal MRI until after a series studied by MRI (678). Ocular anomalies (hypertelorism,
30 weeks because of their small size and the current limits of technol- microphthalmia, and iris colobomas) (679) and other developmental
ogy (674). They are best identified postnatally by thin section (3 mm anomalies of the nose and paranasal sinuses (680) may be seen as
or less) coronal T2-weighted images (Fig. 5-107C), where they are seen well. Arrhinencephaly is most likely the result of either a severe focal
to undergo a characteristic maturation pattern (675). injury in utero or mutation of a gene involved in early development
Arrhinencephaly is typically associated with multiple congeni- of the rostroventral prosencephalon. When multiple anomalies are
tal anomalies, with cleft lip, cleft palate, hypoplasia/absence of the present, the condition is usually diagnosed in early infancy. In the
nose and nasal cavity, and holoprosencephaly being the most com- rare cases when the disorder of the olfactory apparatus is isolated,
mon (676). Olfactory agenesis has been reported in 23% of patients the anomaly may remain cryptic until the child’s absence of olfactory
with CHARGE association (coloboma, heart disease, choanal atresia, sensation is discovered.

Barkovich_Chap05.indd 453 5/6/2011 8:59:58 PM


454 Pediatric Neuroimaging

FIG. 5-106. Middle interhemispheric variant of


holoprosencephaly, fetal MRI. Inferior axial image
(A) shows absence of the septum pellucidum and
presence of anterior and posterior interhemispheric
fissures. More superior image (B) shows continuity
of the hemispheres across the midline (white arrows).
Coronal image (C) confirms continuity of the hemi-
spheres across the posterior frontal midline.

It is important to specifically look at the olfactory structures and


Septooptic Dysplasia
the midline structures of the brain in any patient with abnormalities
of olfaction or midline facial anomalies, particularly if the CHARGE Septooptic dysplasia, as described by de Morsier in 1956, consisted
association is suspected, as this finding strongly supports the diagno- of two major findings: hypoplasia of the optic nerves and hypoplasia
sis. Indeed, neuroimaging studies are usually performed in patients or absence of the septum pellucidum (685). It is currently considered
with facial anomalies primarily to assess associated anomalies of the to be a heterogeneous disorder loosely defined by any combination
brain; therefore, MRI is the study of choice. On CT, arrhinia shows of optic nerve hypoplasia, pituitary gland hypoplasia, and midline
variably severe absence of the nose and nasal cavity, with a high abnormalities of the ventral or rostral cerebrum (686). In all prob-
arched hard palate and absence of the nasal bones (Fig. 5-108) (681). ability, the “syndrome” of septooptic dysplasia is the end result of
In milder cases, congenital nasal pyriform aperture stenosis or choa- several different genetic abnormalities (primarily abnormal expres-
nal atresia may be present, just as they may be present in holoprosen- sion of genes in the ventral and rostral prosencephalon) and in utero
cephaly (682,683). MRI typically shows arrhinencephaly as absence of injuries (616,687–690). One family with septooptic dysplasia and
the olfactory sulci, olfactory bulbs, and olfactory tracts (Fig. 5-108A) several individuals with a sporadic form of the syndrome have been
(684). Anomalies of the hypothalamic-pituitary axis (HPA), corpus shown to have mutations in a gene called HESX1, which is produced
callosum, and ventral forebrain are the most common associated find- by tissue adjacent to the developing prosencephalon and is located at
ings. When anosmia or hyposmia is associated with hypogonadotropic chromosome 3p21.1–3p21.2 (686,691,692). This location suggests that
hypogonadism, the diagnosis of Kallmann syndrome should be con- proper induction of the prosencephalic midline depends upon HESX1
sidered (see section “Anomalies of the Hypothalamic-Pituitary Axis” expression, and supports the concept of an overlap of these patients
later in this chapter). with those manifesting holoprosencephaly. HESX1 is also produced

Barkovich_Chap05.indd 454 5/6/2011 8:59:59 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 455

FIG. 5-107. Normal olfactory apparatus on MRI.


Axial (A) and coronal (B) scans of 30-week fetus show
the olfactory sulci (white arrows) between the gyrus
rectus and medial olfactory gyrus. Because they form
from posterior to anterior, they are seen earliest just
anterior to the suprasellar cistern (A). Olfactory bulbs
are not seen in the fetus. On postnatal scans (C), both
the olfactory bulbs (white arrows just above the cribri-
form plate) and olfactory sulci (white arrowheads) are
easily seen on coronal T2-weighted images.

(at a later stage of development) by Rathke pouch primordium, a fact conjunction with neuroimaging. When hypoplasia of the optic discs
that possibly explains the observed hypopituitarism of many affected is seen in association with partial or complete absence of the septum
patients (693). More recently, duplications of SOX3 and mutations of pellucidum, the diagnosis is established.
SOX2 and SOX3, transcription factors involved in pituitary develop- Imaging studies show hypoplasia or complete absence of the
ment, have been implicated in the etiology of SOD (694). septum pellucidum, resulting in box-like frontal horns (Fig. 5-109)
Approximately, two thirds of patients with SOD have hypo- (616,639,700,701). Patients with complete septal absence have a higher
thalamic-pituitary dysfunction (695–699); indeed, some authors incidence of hypothalamic-pituitary dysfunction and worse neurologi-
(particularly endocrinologists) consider it an essential part of the syn- cal and developmental outcome than those with partial septal absence
drome. The clinical presentation is variable, as expected from a het- (702). The absence of the septum can often be identified on the sagittal
erogeneous disorder. Visual symptoms may include nystagmus and image by the low position of the fornices (Fig. 5-109A). High qual-
diminished visual acuity; however, patients can have normal vision ity, thin section coronal MR images allow confirmation of the absent
(696). Occasionally, hypotelorism is present. When hypothalamic- septum and facilitate the diagnosis of hypoplasia of the optic nerves
pituitary dysfunction is present, it is usually manifested as growth in severe cases (Fig. 5-109C). However, determining mild hypoplasia
retardation secondary to deficient secretion of growth hormone of the optic nerves, chiasm and tracts by neuroimaging is extremely
and thyroid stimulating hormone (695,698,699). The diagnosis of difficult; in fact, optic nerve hypoplasia is recognized on neuroimag-
septooptic dysplasia is made by ophthalmological examination in ing studies in only about 50% of affected patients (616). Severe optic

Barkovich_Chap05.indd 455 5/6/2011 8:59:59 PM


456 Pediatric Neuroimaging

FIG. 5-108. Arrhinia with arrhinen-


cephaly. Coronal T1-weighted image
(A) shows absence of the olfactory
sulcus and bulbs. Axial CT image
(B) shows that the nasal cavity is
absent. Oblique view of 3D reforma-
tion (C) shows absence of the nasal
bones. (This case courtesy Clark Car-
roll, MD.)

nerve hypoplasia is best identified on sagittal and coronal, rather than normal septum and HPA on MRI had normal endocrine function,
axial, MR images; bulbous dilatation of the anterior recess of the third 22% of those with abnormal septum and normal HPA had abnormal
ventricle and a large suprasellar cistern may be associated. Of note, endocrine function, 35% of those with normal septum and abnormal
patients with unilateral optic nerve hypoplasia uncommonly have HPA had abnormal endocrine function, and 56% of those with abnor-
brain anomalies other than minor pituitary pathology (Fig. 5-109) mal septum and HPA had abnormal endocrine function. Therefore,
(702). Brodsky et al. (687) showed that patients with neuroradiologic MR evaluation of the septum pellucidum and HPA may be useful to
findings limited to isolated optic nerve hypoplasia or optic nerve hyp- predict the likely spectrum of endocrinopathy (703).
oplasia in conjunction with partial or complete absence of the sep- Several distinct MR subgroups have been identified in patients
tum seem to have a good developmental prognosis, whereas those with septooptic dysplasia (616,687). One group shows a high incidence
with hemispheric anomalies or posterior pituitary ectopia have more of malformations of cortical development (especially schizencephaly,
guarded prognoses. Another study showed that 49% of patients with PMG, and gray matter heterotopia [616,687,702]) and partial absence
optic nerve hypoplasia have an abnormal septum pellucidum, while of the septum pellucidum (Fig. 5-110); these patients are often found
64% have a hypothalamic-pituitary abnormality (703). Patients with to have incomplete rotation of the hippocampi, as well (702). The

Barkovich_Chap05.indd 456 5/6/2011 9:00:00 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 457

FIG. 5-109. Septooptic dysplasia with ectopic posterior pituitary


lobe. Sagittal T1-weighted image (A) shows the hyperintense pos-
terior pituitary lobe (white arrow) at the median eminence of
the hypothalamus instead of in the sella turcica. The fornices
(white arrowheads) are too low, suggesting absence of the septum
pellucidum. Coronal T1-weighted image (B) confirms absence of
the septum and the presence of an ectopic posterior pituitary (white
arrow). More anterior coronal STIR image (C) shows a hypoplastic
right optic nerve (white arrow).

finding of hippocampal anomalies is strongly associated with callosal neurologic deficits are present in the majority of patients with appar-
anomalies and neurologic/developmental difficulties (702). Small optic ently isolated absence of the septum (705). Because isolated septal
nerves in this group may be the result of transsynaptic degeneration of agenesis is rare, the brain should be carefully scrutinized for associated
the optic nerves secondary to prenatal destruction/hypogenesis of the anomalies when the septum is absent. Abnormalities associated with
optic radiations. A second group has findings of complete absence of septal absence include holoprosencephaly, callosal agenesis, septooptic
the septum pellucidum and hypoplasia of the cerebral white matter, dysplasia, schizencephaly, bilateral PMG, malformations secondary to
often resulting in ventriculomegaly, but normal cerebral cortex. Some abnormal pial basement membrane formation such as WWS and MEB,
patients in this second subset have hypoplasia of the anterior falx cere- chronic severe hydrocephalus, hypothalamic-pituitary anomalies, and
bri or the genu of the corpus callosum (Fig. 5-111), compatible with Chiari II malformations (260,575,701,705).
impaired induction of the rostral end of the neural tube, in addition to
the ventral region where the hypothalamus is generated. Some patients
Anomalies of the Hypothalamic-Pituitary Axis
(perhaps a third subset) with septooptic dysplasia have posterior pitu-
itary ectopia (Fig. 5-109) (687,704) and a fourth subset likely includes Abnormalities of the pituitary gland are often visible by MRI. The pitu-
those in the family described by Dattani et al. (686,691), who have itary gland may be too small or it may be enlarged by congenital cysts
hypogenesis of the corpus callosum. (cysts are discussed in Chapter 7); the posterior pituitary gland (some-
times referred to as the pituitary “bright spot” for its hyperintense signal
on T1-weighted MR images) may be absent or ectopic (in the pituitary
Isolated Absence of the Septum Pellucidum
stalk or in the hypothalamus proper at the median eminence); or, very
Isolated absence of the septum pellucidum is a relatively rare struc- rarely, the pituitary gland and stalk may be duplicated or even tripli-
tural anomaly that may have no neurologic manifestations. However, cated. Abnormalities of the hypothalamus are more difficult to detect

Barkovich_Chap05.indd 457 5/6/2011 9:00:02 PM


458 Pediatric Neuroimaging

FIG. 5-110. Septooptic dysplasia with schizencephaly. Coronal SPGR T1-weighted images show hypopla-
sia of the left intracranial optic nerve (black arrow in A) and, more posteriorly, bilateral schizencephalies
(white arrows in B).

by imaging, as it is difficult to differentiate the hypothalamus from ridge (706). Whatever the origin, Rathke pouch contacts and comes to
surrounding structures, even with very thin section coronal images, and lie anterior to a downward extension of the embryonic hypothalamus
it is so small that subtle anomalies are difficult to appreciate. Nonethe- known as the neurohypophysis, or posterior pituitary lobe (Fig. 5-112).
less, developmental hypothalamic abnormalities, such as hamartomas The connection of Rathke pouch with the buccal cavity then undergoes
(discussed in Chapter 7) or lack of midline separation, may occasion- obliteration, although portions of it may persist as the craniopharyn-
ally be detected if the region is carefully scrutinized. A basic under- geal canal or as nests of ectopic pituitary tissue in the nasopharynx
standing of the embryology of the region facilitates an understanding or sphenoid sinus. After its connection to the buccal cavity is obliter-
of hypothalamic and pituitary anomalies. ated between 42 and 44 days of gestational age, Rathke pouch becomes
known as the adenohypophysis, or anterior pituitary lobe (707,708).
Embryology The molecular biology of this transition has been elucidated and is
Between 28 and 32 days of embryonic life, a shallow vesicle known nicely reviewed in several publications (709,710).
as Rathke pouch or Rathke cleft appears on the roof of the foregut, Between 37 and 42 days gestational age, lateral lobes form in the
immediately rostral to the notochord. Traditionally, Rathke pouch has developing adenohypophysis. These lobes, known as the tuberal pro-
been considered to arise from the buccal cavity; however, recent evi- cesses, will differentiate into the pars tuberalis. Between 42 and 44 days,
dence supports origin from neuroectoderm, along the anterior neural the tuberal processes surround the infundibulum and developing

FIG. 5-111. Septooptic dysplasia with dysgenesis of the corpus callosum. Sagittal T1-weighted image (A) shows lack or formation of the
callosal genu despite a normal appearing body and splenium. Coronal T1-weighted image (B) shows absence of the septum pellucidum and
hypoplasia of the left optic nerve (black arrow).

Barkovich_Chap05.indd 458 5/6/2011 9:00:03 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 459

FIG. 5-112. Formation of the pituitary gland. A. Between 28


and 32 days of embryonic life Rathke pouch (RP) appears on
the roof of the foregut, immediately rostral to the notochord.
Rathke pouch contacts, and comes to lie anterior to a downward
extension of the embryonic hypothalamus known as the neuro-
hypophysis, or posterior pituitary lobe (nh). B. The connection
of Rathke pouch with the buccal cavity undergoes obliteration
between 42 and 44 days gestational age, at which time Rathke
pouch becomes known as the adenohypophysis, or anterior
pituitary lobe (ah). C. Between 44 and 48 gestational days, the
midline anterior lobe cells that are in close approximation to
the posterior lobe differentiate into the primordium of the pars
intermedia (pi). All of the components of the mature hypotha-
lamic-pituitary axis are now in place and will slowly evolve into
the mature system.

neurohypophysis laterally and give some definition to the developing of the adrenal glands, thyroid, testes, ovaries, and penis. The sella is
hypophysis. Between 44 and 48 gestational days, the midline anterior- small and flat, and sometimes is covered by a layer of dura. Pituitary
lobe cells that are in close proximity to the posterior lobe differentiate hypoplasia (Fig. 5-113) is much more common than aplasia; more
into the primordium of the pars intermedia. All of the components of importantly, affected patients can survive with hormonal replacement
the mature HPA are now in place by day 49 and will slowly evolve into therapy (717,718). These patients may have short stature as a result of
the mature system (707,708). deficiency of growth hormone and, therefore, fall into the category of
Functional specialization of the pituitary gland involves the for- pituitary dwarfism (see next section). Mutations of a number of differ-
mation of the portal circulatory system and differentiation of hor- ent genes have been discovered to cause pituitary hypoplasia, with vari-
mone-secreting cells. It is postulated that this differentiation requires able types of inheritance and different specifics regarding the effects
directed contact of the gland with the hypothalamus (711). The genetic upon production of specific hormones and development of the ante-
basis of pituitary cell differentiation is being established and seems to rior and posterior lobes of the pituitary (713,719).
involve transient signaling gradients that induce nuclear genes to pro- Very rarely, the hypothalamus and pituitary gland may be dupli-
duce mediators that lead to cell specification (712). These mediators cated (Fig. 5-114), or even triplicated (720); other anomalies of the
include numerous transcription factors acting as repressors or activa- face and brain are nearly always present (721–723). The most common
tors, and associated coregulators that arise from the adenohypophysis of these are facial dysmorphism (ranging from hypertelorism to facial
and neighboring structures (709,710,713). Details of these factors are clefting), anomalies of the tongue, cleft palate, pharyngeal teratomas,
beyond the scope of this book. Suffice it to say that mutations of certain agenesis of the corpus callosum, anomalies of the circle of Willis (par-
genes that are important in pituitary development cause fairly specific ticularly partial fenestration of the basilar artery [724]), and anoma-
deficiencies in anterior and posterior pituitary development as well as lies of the olfactory bulbs and tracts (722,723). Delayed or precocious
development of other brain and visceral structures (713). puberty may be found in affected patients (725).
The development of the hypothalamus is not as well understood.
It is clear that hypothalamic cells are generated in the floor plate of Pituitary Dwarfism
the developing neural tube very early in the region of Henson node, Pituitary dwarfism is composed of a heterogeneous group of disorders,
then move rostrally under the influence of the prechordal plate and its all caused by growth hormone deficiency and characterized by short
secreted proteins such as SHH (714). Therefore, disorders that result stature, slow growth, defective dentition, and delayed skeletal matura-
from abnormal induction of the ventral midline forebrain, such as hol- tion. Males are affected two to three times as commonly as females.
oprosencephaly, nearly always result in abnormalities of the hypothal- The hormonal deficiency may be limited to growth hormone, or may
amus and abnormal hypothalamic function (715). In addition, some involve multiple adenohypophyseal and neurohypophyseal hormones.
hypothalamic cells migrate to the hypothalamus from other regions of This diagnosis is suspected if a patient is exceptionally short for age
the developing embryo (716). Therefore disorders of cell migration can according to standard growth charts, is growing poorly for age without
also lead to hypothalamic dysfunction. From an imaging perspective, evidence of any other disorder, or has short stature and any signs or
the important point is to evaluate the hypothalamus, in addition to the symptoms of CNS abnormality (726,727). A number of familial and
pituitary gland, with thin section, T1- and T2-weighted images in all genetic causes of isolated growth hormone deficiency and combined
patients with hypothalamic-pituitary dysfunction. pituitary hormone deficiencies (CPHD) have been described (713,728),
but these are rarely found in clinical practice (713,729).
Pituitary Absence, Hypoplasia, and Duplication Imaging studies show a range of features, including a small sella
Absence or hypoplasia of the pituitary gland is a rare anomaly that turcica and anterior pituitary lobe (Fig. 5-113), hypoplasia or absence of
involves absence or hypoplasia of both the anterior and posterior lobes the pituitary stalk, and presence of the high signal intensity neurohypo-
and, in many cases, the pituitary stalk (717); this disorder is nearly always physis (posterior lobe) in the infundibulum or at the median eminence
fatal at birth. Commonly associated anomalies include hypoplasia of the hypothalamus (“ectopic bright spot”) (Fig. 5-115) (730–733).

Barkovich_Chap05.indd 459 5/6/2011 9:00:04 PM


460 Pediatric Neuroimaging

FIG. 5-113. Severe anterior pituitary hypoplasia. Sagittal (A) and coronal (B) T1-weighted images show a tiny anterior pituitary
lobe (white arrow), which is smaller than the hyperintense posterior pituitary.

FIG. 5-114. Pituitary duplication. Sagittal T1-weighted image (A) shows an abnor-
mally thick floor (open white arrows) of the third ventricle. Coronal T1-weighted
images show a thick bar of gray matter at the floor of the third ventricle with two pitu-
itary stalks (small white arrows in B) exiting from it and two separate pituitary glands
(black arrows in C).

Barkovich_Chap05.indd 460 5/6/2011 9:00:05 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 461

FIG. 5-115. Anterior pituitary hypoplasia with ectopic posterior pituitary lobe. Sagittal and axial T1-weighted images show
a small infundibulum (not visible), small anterior pituitary lobe, and ectopic posterior pituitary lobe at the median eminence
of the hypothalamus (white arrow). Such patients commonly have other anomalies such as absent corpus callosum, and large
anterior commissure (white arrowhead).

Only about 40% of patients with pituitary dwarfism will have the full located within the median eminence of the hypothalamus) is pres-
spectrum of hypothalamic-pituitary imaging features; those patients ent, patients have a higher incidence of CPHD (49%), breech delivery
with the full spectrum are likely to have multiple hormone deficien- (32%) and associated brain and skull base anomalies (12%) (727,735),
cies (CPHD) and lower levels of growth hormone (727,731,732). Post including callosal anomalies (Fig. 5-115), optic nerve hypoplasia, and
contrast thin section MRI will not visualize the pituitary stalk in such PVNH (529,713,719).
patients and the ectopic posterior pituitary gland will be located in the Although most patients with CPHD present in the neonatal period
median eminence (Fig. 5-115) (733). Conversely, MR scans of patients and have a very small pituitary gland, patients with homozygous or
with isolated growth hormone deficiency are more likely to have a nor- compound heterozygous mutations of the PROP1 gene present with
mal sized adenohypophysis and a visible infundibulum than those with profound growth retardation in childhood (736). PROP1 is a transcrip-
multiple hormonal deficiencies (733,734); the ectopic posterior pitu- tion factor whose expression leads to normal development of pituitary
itary gland will typically be found along the pituitary stalk (Fig. 5-116). gonadotropes, somatotropes, lactotropes, and thyrotropes. Inactivating
This less severe anomaly is sometimes called “partial” pituitary ectopia mutations lead to deficiencies in luteinizing and follicle-stimulating
(733). When complete posterior pituitary ectopia (posterior pituitary hormones, growth hormone, prolactin, and thyroid stimulating

FIG. 5-116. “Partial” posterior pituitary ectopia. Postcontrast sagittal (A) and noncontrast coronal (B) T1-weighted
images show a large posterior pituitary gland (white arrows) that is located along the intact pituitary stalk. The anterior
pituitary gland is normal in size for a young child. No other anomalies were identified.

Barkovich_Chap05.indd 461 5/6/2011 9:00:07 PM


462 Pediatric Neuroimaging

hormone. Mutations of PROP1 seem to account for about 40% of treatments can restore reproductive function and patients can lead
cases of CPHD (737–739). In contrast to most types of CPHD, which relatively normal lifestyles (752). MRI can be important in establish-
are characterized by a small gland and small sella turcica, imaging of ing the diagnosis (684), as it allows assessment of the formation of the
CPHD patients with PROP1 mutations is characterized by enlargement olfactory sulcus and bulbs, which are hypoplastic or absent in Kall-
of the pituitary gland, with intrinsic T1 hyperintensity and marked T2 mann syndrome. Although axial images can suggest the absence of the
hypointensity of the anterior pituitary lobe (740,741). olfactory sulcus (684,753), coronal scans, particularly thin section T2
RARE (FSE/TSE) images, will show the anatomy more definitively (Fig.
Kallmann Syndrome (Hypogonadotropic 5-117). These observations can be made on fetal MRI, as well, if high
Hypogonadism) quality coronal images are obtained. If the hypothalamic hypofunc-
Kallmann syndrome (OMIM 308700) (742) is a genetically heteroge- tion is severe, sagittal images may show a small pituitary gland (Fig.
neous disorder of neuronal migration in which olfactory cells and cells 5-117A). One should also look for associated cleft palate, dental agen-
that normally express luteinizing hormone releasing hormone fail to esis, or renal anomalies (728).
migrate from the olfactory placode into the forebrain. Normally, both
of these cell types originate in the olfactory placode (in the nasal fossa) Hypothalamic Dysgenesis
and migrate superiorly and anteriorly along olfactory axons toward the As stated in the introduction of this section, hypothalamic anomalies
developing cerebral hemispheres (716). The migration is facilitated by are difficult to detect by MRI because the hypothalamus is so small and
the recognition of specific neural-cell adhesion molecules on the sur- is difficult to differentiate from surrounding structures. When patients
face of cells along the pathway of migration (743,744). In X-linked Kall- have abnormalities of hypothalamic function (usually manifested as
mann syndrome (OMIM 147950), the KAL1 gene (at Xp22.3), which diabetes insipidus) or abnormal levels of the hormones produced in
encodes a small extracellular matrix protein that presumably acts as a the hypothalamus (usually releasing factors for pituitary hormones),
permissive substrate for outgrowth of olfactory axons (50), is deleted; we typically look for either atrophy (presumably secondary to prior
as a result, the cells generated in the olfactory placode remain in the injury) of the hypothalamus (typically manifested as asymmetric
nasal cavity (745). In autosomal dominant Kallmann syndrome, the enlargement of the anterior third ventricle), tumor of the suprasellar/
cause is a loss of function mutation of the gene for fibroblast growth third ventricle region (see Chapter 7), or lack of normal development
factor receptor 1 (FGFR1 [also called KAL2], located at chromosome of the hypothalamic/suprasellar structures. In terms of malformations,
8p11.2-p11.1 [746]). FGFR1 mutations appear to cause about 10% of we particularly scrutinize the optic chiasm to see that it is normally
Kallmann syndrome cases by impairing formation of gonadotropin formed and separated from the hypothalamus, and the walls of the
releasing hormone (747). (Of interest, gain of function mutations of anterior third ventricle to make sure they are completely separated
FGFR1 cause craniosynostosis; see later section of this chapter.) Other (Fig. 5-118). The neurohypophysis should be evaluated as well to
genes causing Kallmann syndrome include PROKR2 at chromosome ensure that its migration to the sella turcica has not been arrested at
20p13 (KAL3, OMIM 244200 [748]), PROK2 at chromosome 3p21.1 the median eminence or in the pituitary stalk (719).
(KAL4, OMIM 610628 [748]), CHD7 at chromosome 8q12.1 (KAL5,
OMIM 612370 [749]), and FGF8 at chromosome 10q24 (KAL6, OMIM Anomalies of the Eyes
612702 [750]).
Patients with Kallmann syndrome typically present with anosmia Embryology of the Eye and Orbit
or hyposmia and, in older children, with hypogonadism. Occasionally, At about the 22nd postconceptional day, the lateral walls of the dien-
renal anomalies, cleft palate, and dental agenesis may be associated cephalic portion of the (still open) neural tube take the shape of two
(751). Establishing the proper diagnosis is important, as hormonal broad and shallow concavities; these are the incipient optic vesicles

FIG. 5-117. Kallmann Syndrome. Sagittal T1-weighted image shows


pituitary hypoplasia (white arrowheads) and small corpus callosum.
Coronal T1-weighted (B) and T2-weighted (C) images show absence of
the normal olfactory sulci separating the gyri recti from the medial orbital
gyri. Coronal T2-weighted image (D) shows normal olfactory sulci (white
arrowheads) and bulbs (arrows) for comparison.

Barkovich_Chap05.indd 462 5/6/2011 9:00:08 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 463

FIG. 5-117. (Continued)

FIG. 5-118. Hypothalamic anomaly in a child with hypotha-


lamic dysfunction. Sagittal T1-weighted image (A) shows hypo-
genesis of the corpus callosum (white arrows) without a visible
third ventricle, anterior commissure, or posterior commissure.
The pituitary gland looks normal. A small region of ectopic pos-
terior pituitary gland (white arrowhead) is visible at the median
eminence. Coronal T1-weighted image (B) shows a thick, asym-
metric (to the right side) optic chiasm (white arrowheads).
Coronal T2-weighted image (C) shows lack or separation of the
hypothalami resulting in continuity of the hypothalamic gray
matter across the midline (black arrows) and obliteration of the
anterior third ventricle.

Barkovich_Chap05.indd 463 5/6/2011 9:00:09 PM


464 Pediatric Neuroimaging

FIG. 5-119. Development of the eye. A. During the early fourth fetal week, the optic vesicle (OV) evagi-
nates from the diencephalic forebrain wall (FW) toward the lens placode (LP), which is a focal thickening
of ectoderm. As the ectoderm thickens, the optic vesicle becomes indented. B. Later in the fourth week, the
optic vesicle becomes cup-shaped (OC) under the continued influence of the lens placode. At the same
time, a second invagination occurs, forming a groove (the optic fetal fissure, OFF) running from the devel-
oping cup posteriorly to the forebrain wall. C. By the end of the fourth week, the optic cup invaginates such
that the sector that was closest to the ectoderm, the incipient retina (neuroretinal layer, NRL), has doubled
back against the incipient pigment epithelium (pigment layer, PL). These are still separated by the optic
ventricle (V), which is in communication with the third ventricle through the optic stalk (OS). The lens
vesicle (LV) has almost completely budded off of the ectoderm at this time.

(Fig. 5-119). Under the influence of the optic vesicle, the overlying the iris. A mass of cells that bud off from the iris will differentiate into
ectoderm begins to thicken to form the lens plate. The lens plate also the pupilloconstrictor and pupillodilator muscles (25,754–756). The
influences the development of the optic vesicle, resulting in thickening of genetics of ocular development are complex but are slowly being elu-
the underlying neural tissue that is destined to become the neural retinal cidated; currently, however, the mechanisms by which mutations cause
layer. Late in the fourth gestational week, the optic vesicle invaginates ocular anomalies are unclear (757–759).
to form the optic cup and the lens plate extends into the hollow of the
cup. At the same time, a second invagination occurs to form a groove Ocular Malformations
running lengthwise along the ventral aspect of the optic cup; this is As the genetic basis of ocular malformations remains unclear at this
the optic fetal fissure, also called the choroidal fissure (Fig. 5-119). time, these malformations are most easily classified by differentiating
Mesenchyme will migrate through this fissure to form the primary vit- them into anophthalmia, microphthalmic malformations, and mac-
reous and the hyaloid artery. The optic cup invaginates deeply such rophthalmic malformations. Ocular anomalies associated with specific
that the sector of its wall that was closest to the surface ectoderm at syndromes, such as Trisomy 13, septooptic dysplasia, WWS, MEB, and
the vesicle stage (that portion destined to become the neural retinal the phakomatoses, are discussed in the sections and chapters on those
layer) becomes doubled back against the sector that will become the specific disorders.
pigment epithelium (Fig. 5-119C). The pigment epithelium will ulti-
mately differentiate into rods, cones, and ganglion cells. A cup-shaped, Anophthalmia
CSF-containing slit develops between the incipient neural retinal and The term anophthalmia refers to congenital absence of one or both
pigment epithelium layers (Fig. 5-119C), remaining continuous with eyes. Some authors contend that true primary anophthalmia is incom-
the third ventricle. During the fifth and early sixth gestational week, patible with life and that the condition seen in live newborns is best
the apposed margins of the fetal optic fissure fuse so that the site at termed extreme microphthalmia or clinical anophthalmia (760,761).
which the initial retinal axons had passed from the vitreal surface into We will use the term for any condition where no ocular globe or optic
the fetal fissure becomes enclosed within the retina as the optic nerve nerve is identified. It can be divided into two major forms. Primary
head. The axon bundles from the developing neural retinal layer run anophthalmia, in which the ocular primordia never form, is very rare
within the thinner, more posterior aspect of the fetal fissure known and is probably the result of a mutation of genes that are important in
as the optic stalk; these will develop into the optic nerve. In the sev- the development of the optic vesicle. The gene most strongly linked
enth gestational week, the margin of the optic cup grows forward and to anophthalmia is SOX2 at chromosome 3q26.3-q27, mutations of
encloses the anterior portion of the lens to form the ciliary body. Later, which have been shown to account for 10% to 20% of severe bilateral
the optic cup extends even farther forward to form the epithelium of anophthalmia/microphthalmia (762); affected infants commonly have

Barkovich_Chap05.indd 464 5/6/2011 9:00:10 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 465

but also mutations of SOX2, PAX6, CHD7, PAX2, SIX3, HESX1, SHH,
SIX6, RAX, and BCOR [757–759,765]), and inborn errors of metabo-
lism (most commonly Lowe syndrome, see Chapter 3). In children,
secondary microphthalmia is most commonly the result of retinopa-
thy of prematurity (ROP), ocular infection, or prenatal toxin exposure
(760,764,765); the imaging findings in secondary microphthalmia are
nonspecific, consisting of small ocular globe with small optic nerve.
Those conditions with specific imaging findings will be discussed in
some detail.

Colobomas Coloboma is the name give to a fissure or discontinuity


in any ocular structure; the most typical form is that resulting from
failure of the choroidal fissure to close properly (760); mutations of
many genes have been implicated in their formation, the two best char-
acterized being PAX6 at 11p13 (766) and GDF6 at 8q21.2-q22.1 (767).
Ocular colobomas are fairly common, often bilateral, and are usually
mild (the most common form is located in the inferior temporal aspect
of the iris and is cryptic to imaging); as a result, imaging studies of
most patients with colobomas are normal (768,769). They are asso-
ciated with many brain malformation syndromes, including Joubert
FIG. 5-120. Anophthalmia. Axial T1-weighted image shows neither optic syndrome (JS), Aicardi syndrome, and WWS (769). Two variants of
nerves nor globes. Extraocular muscles are identified. colobomas have rather typical imaging findings. The morning glory
syndrome (named for the characteristic ophthalmoscopic appearance)
is a coloboma that affects the optic nerve at the optic disc; the syn-
associated esophageal atresia, vertebral segmentation anomalies, facial drome is commonly associated with brain anomalies such as callosal
dysmorphisms, and anomalies of male genitalia (759,761). Mutations agenesis and basilar encephaloceles (770). Imaging studies have a char-
of OTX2 on chromosome 14q22 and RAX on chromosome 18q21.3 acteristic appearance in which a section of retina at the site of the optic
have also been associated with primary anophthalmia (757,759). With nerve head is displaced posteriorly (Fig. 5-121). The displaced por-
either cause, the brain (and in particular the “visual cortex”) appears tion may be rectangular or cone-shaped. Contiguous retrobulbar cysts
normal (763). Secondary anophthalmia is more common and is gen- may be present (Fig. 5-121B) (770). Coloboma with cyst (also called
erally related to infection (such as rubella), trauma, vascular events, microphthalmia with cyst) is a malformation caused by extensive pro-
toxic/metabolic events (hypervitaminosis or hypovitaminosis A), or liferation of the embryonic retina with consequent separation of the
exposure to toxins occurring during the early part of the fourth ges- inner and outer retinal layers (760,771). The walls of these cysts may
tational week (764). In true anophthalmia (primary or secondary), fuse with the wall of the globe to create a continuity of the cyst with
imaging shows that neither the optic nerves nor the globes are present the vitreous cavity (Fig. 5-121B); large cysts, therefore, may be associ-
(Fig. 5-120). Extraocular muscles, the lachrymal apparatus, and blood ated with extrusion of vitreous humor into the cyst and consequent
vessels may be identified. diminished size and displacement of the globe (Fig. 5-122). Indeed,
large cysts may be larger than the globe itself (Figs. 5-121B and 5-122)
Microphthalmic Malformations and may cause proptosis (Fig. 5-122). The globe can be differentiated
Microphthalmia refers to a condition in which the eye is abnormally from the cyst by the presence of a lens. Imaging studies will often dem-
small. It can be unilateral or bilateral and may result from a number of onstrate continuity of the globe and the retrobulbar cyst, confirming
different processes. Primary microphthalmia is a finding in many syn- the diagnosis and aiding differentiation from other conditions (768).
dromes (765); it can be the result of primary ocular hypoplasia, ocular
coloboma, PHPV, congenital infections (most commonly rubella, see Persistent Hyperplastic Primary Vitreous PHPV is a malformation
Chapter 11), chromosomal disorders (most commonly trisomy 13, that is caused by a failure of regression of the fibrovascular tissue of the

FIG. 5-121. Colobomas. Axial T2-weighted image (A) shows a small left coloboma (white arrows) with the posterior midline wall of the globe
being posteriorly displaced. Axial T1-weighted image (B) shows a large right retrobulbar cyst (large black arrows), extending posteriorly from a
small colobomatous globe (small black arrows). The contralateral (left) globe has been previously treated for retinal dysplasia.

Barkovich_Chap05.indd 465 5/6/2011 9:00:11 PM


466 Pediatric Neuroimaging

the eye. In either case, patients typically present at birth or during the
first months after birth with microphthalmia, leukocoria (white pupil),
and congenital cataract (764,772). Indeed, PHPV is the second most
common cause of leukocoria (after retinoblastoma, see Chapter 7),
accounting for 20% to 25% of cases (773). Both autosomal domi-
nant (OMIM 611308) and recessive (OMIM 611311) forms have been
described. Most cases are unilateral, although bilateral PHPV is much
more common when associated with Norrie disease, WWS, and other
neurologic and systemic abnormalities (774).
Pathologically, patients with anterior PHPV have a shallow ante-
rior chamber, enlarged vessels in the iris, and a retrolental fibrovascular
membrane. Cataracts and intralenticular hemorrhage usually develop,
and the latter may lead to glaucoma. When posterior PHPV is present,
a fibrovascular stalk is seen traversing the globe from the optic nerve to
the posterior lens along a remnant of the hyaloid canal (Cloquet canal).
This is associated with elongation of the ciliary processes, dysplasia of
the optic disc, an indistinct macula and, sometimes, retinal folds. The
stalk may exert traction on the retina, resulting in retinal detachment
and subretinal hemorrhage. Malformations of the optic nerve and
retina are common (774,775).
FIG. 5-122. Coloboma with cyst (microphthalmia with cyst). The large The diagnosis can be made with any imaging modality. Ocular
cyst (large white arrows) is in continuity with the vitreous cavity (small ultrasound shows an echogenic mass of variable size posterior to the
white arrows), which can be identified by the presence of the lens (white lens; a hyperechoic band extends from the posterior surface of the
arrowhead). The large size of the cyst has resulted in proptosis. mass to the posterior pole of the globe. Doppler imaging will shows
flow within this band, representing persistence of the hyaloid artery
in Cloquet canal. Retinal detachment may be seen as a curvilinear,
primary vitreous and of the hyaloid artery, which delivers blood to the echogenic structure within the anechoic vitreous (776). CT of PHPV
primary vitreous and the developing lens (760,772). The embryonic shows microphthalmia and portions or all of a vertical septum (the
intraocular vascular system is composed of two components: an ante- hyaloid artery running through Cloquet canal) extending from the
rior system near the iris and a posterior retrolental system within the head of the optic nerve to the site of the primary vitreous (immedi-
vitreous. Persistence of these vessels often results in vascular prolifera- ately behind the lens) (Fig. 5-123) (772,777). A generalized increase in
tion, which can cause PHPV in the anterior or posterior segments of attenuation of the vitreous is sometimes present. It is also important

FIG. 5-123. Persistent hyperplastic primary vitreous. Axial contrast-en-


hanced CT (A), contrast-enhanced, fat-suppressed T1-weighted MRI (B),
and axial fat-suppressed T2-weighted MRI (C) show a small left globe with
abnormal vitreal signal, an abnormally shaped lens, and abnormal struc-
ture located behind the lens (the persistent primary vitreous, black arrows)
and a tract (black arrowheads) extending from the lens to the optic nerve
head (seen best on MRI).

Barkovich_Chap05.indd 466 5/6/2011 9:00:12 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 467

to confirm that no calcium is present within the globe; this confirms


that the leukocoria is not the result of a retinoblastoma (see Chapter
7). MRI better shows a variably enhancing mass located behind the
lens, Cloquet canal, the abnormal lens, elongated ciliary processes, and
associated retinal detachment with subretinal hemorrhage (Figs. 5-123
and 5-49) (776–778). Moreover, MRI does not result in exposure of
the developing eyes to ionizing radiation. At UCSF, MRI is typically
performed if ocular ultrasound strongly suggests either retinoblastoma
or PHPV; CT is still used for equivocal cases.

Retinopathy of Prematurity ROP is mentioned here primarily


because it is still occasionally seen in pediatric practices, and it should
not be mistaken for other disorders. ROP is caused by the vasocon-
strictive effect of high blood levels of oxygen (fortunately, the use of
antenatal steroids has markedly reduced the incidence of lung disease
associated with prematurity). The vasoconstriction results in chronic
retinal ischemia, which, in turn, causes neovascularization. The neo-
vascularization and its subsequent regression cause subretinal exuda-
tion, hemorrhage, and scarring; both the scarring and the subretinal
exudate may result in chronic retinal detachment and, eventually, FIG. 5-124. Congenital glaucoma (buphthalmos). Axial CT image
microphthalmia. shows enlargement of the right globe in a patient with neurofibromatosis.
Imaging studies show changes that are nearly always bilateral but Enlargement is greater from front to back because much of the enlarge-
often asymmetrical. Microphthalmia is always bilateral and, therefore, ment is in the anterior chamber and the orbital walls restrict medial-lateral
may be difficult to appreciate; clinical history of prematurity should enlargement.
always be sought when bilateral chronic retinal detachment is present.
Retinal detachment is more common in the temporal portions of the bilateral in up to 80% of affected patients (764). Imaging studies show
globe. If retinal detachment is acute, subretinal fluid will be of high enlargement of the entire globe. Often the affected globe will appear
attenuation on CT due to the presence of acute hemorrhage. Calci- more elongated from front to back because (a) much of the enlarge-
fication is rare and is seen only in long standing disease (779). MRI ment is in the anterior chamber and (b) the medial and lateral orbital
will show high signal intensity on T1-weighted images and low signal walls restrict medial-lateral enlargement (Fig. 5-124).
intensity on T2- and T2*-weighted images. More chronic detachment
will show low attenuation subretinal fluid on CT, intermediate to low Other Congenital Ocular Anomalies
signal intensity on T1-weighted MR images and variable intensity on Coats disease is a congenital, nonhereditary ocular anomaly caused by
T2 and gradient-echo images depending upon the amount of deoxyhe- a vascular disorder of the retina in which lipoproteinaceous exudate
moglobin remaining (see [780] and Chapter 4). Scarring in the poste- accumulates in the subretinal space, leading to retinal detachment. In
rior vitreous may be difficult to separate from retinoblastoma by MRI. addition, subretinal and posterior vitreous hemorrhages result from
Ocular ultrasound is the study of choice to differentiate the noncalci- rupture of small aneurysms within the retinal vascular malformation.
fied scar of ROP from the calcified retinoblastoma (calcification causes Males are most commonly affected (70%–80%), typically in the middle
acoustic shadowing). Most important is a history of premature birth years of the first decade of life, but the disorder may be detected at
with associated lung disease. If no history is available, look for bilat- nearly any age. The malformation is almost always unilateral (776,786).
erality of the disease, microphthalmia, and the frequently associated Affected children typically present with diminished vision in the
finding of brain injury secondary to prematurity (see Chapter 4); if affected eye and are found on examination to have leukocoria; there-
these findings are present, the abnormality will nearly always be ROP. fore, differentiation from retinoblastoma (see Chapter 7) is essential.
Another important disease to exclude is infection by Toxocara canii
Macrophthalmia (also known as Toxocara endophthalmitis or larval granulomatosis,
Macrophthalmia refers to a condition in which the eye is abnormally and also discussed in Chapter 7). Ultrasound shows Coats disease as a
large. It is less common than microphthalmia and is usually the result of hyperechoic mass in the posterior vitreous without posterior acoustic
congenital glaucoma (also called buphthalmos) secondary to increased shadowing (776). Hemorrhage is common in the vitreous and subreti-
resistance to resorption of aqueous humor (781). The normal sagittal nal space. The CT features of Coats disease (Fig. 5-125A) are nonspe-
diameter of the neonatal globe is 16 to 17 mm, increasing to 22.5 to 23 cific, showing a normal sized (sometimes slightly enlarged) eye with
mm by age 3 years and then to 24 mm by age 13 years; it remains stable retinal detachment, filling of the subretinal space with slightly dense
in size thereafter (782). In congenital glaucoma, the flow of aqueous proteinaceous material, and variable obliteration of the vitreous space
humor from the anterior chamber is obstructed; as a result the anterior (777,786). Of importance, calcification is absent in the vast majority of
chamber is enlarged from its normal depth of 3 mm (783). Congeni- cases of Coats disease (but present in most cases of retinoblastoma) and
tal glaucoma may be an isolated finding (autosomal recessive primary the abnormal tissue is always intraocular (in contrast to retinoblastoma
congenital glaucoma, OMIM 231300, is caused by mutation of CYP1B1 where extraocular extension may occur). MRI may be useful in that
at chromosome 2p22-p21 [784]), or it may be seen in association with the proteinaceous exudate in Coats disease is of uniform signal inten-
systemic disorders such as Marfan syndrome and Smith-Lemli-Opitz sity, intermediate between brain tissue and vitreous, on T2-weighted
syndrome, some of the phakomatoses (particularly neurofibromato- images and does not enhance on postcontrast images (Fig. 5-125B–D)
sis Type 1 and Sturge-Weber syndrome, see Chapter 6), some meta- (777), whereas the tumor portion of an eye with retinoblastoma will be
bolic disorders (particularly Lowe syndrome and homocystinuria, see of low intensity on T2-weighted images and enhances uniformly other
Chapter 3), and congenital infections (rubella, syphilis) (782,785). It is than in areas of necrosis (see Chapter 7) (787).

Barkovich_Chap05.indd 467 5/6/2011 9:00:13 PM


468 Pediatric Neuroimaging

FIG. 5-125. Coats disease. A. Axial noncontrast CT image shows that the globes are of equal size. A hyperdense proteinaceous exu-
date (white arrowheads) lies in the back of the right globe. No calcification is seen, allowing differentiation from retinoblastoma. B.
Axial T2-weighted image shows that the exudate is homogeneous, other than some hypointensity at the interface with vitreous, and
of signal intensity intermediate between vitreous and brain tissue. C and D. Axial precontrast (C) and postcontrast (D) T1-weighted
images show the exudate to be nonenhancing, homogeneous, and of intensity similar to that of brain tissue.

other than the formation of the diencephalic-mesencephalic boundary


ANOMALIES OF MIDBRAIN-HINDBRAIN (DMB) and the midbrain-hindbrain boundary (MHB), are beyond the
DEVELOPMENT scope of this book. In murine and chick models, the DMB appears to
Overview of Midbrain and Hindbrain form as the result of interactions among the Pax6, Pax2, and En1/2
molecular markers, Pax6 endowing diencephalic fate by repressing
Development Pax2 and En1, while En1 represses Pax6 expression in the mesencepha-
Malformations of the midbrain and hindbrain (brainstem and cer- lon (789) (Fig. 5-126). Changes in the expression of these markers will
ebellum) are being found more frequently in children with mental shift the DMB caudally (more Pax6) or rostrally (more Pax2/En1). The
retardation and congenital neurologic dysfunction (3,9,10). As with all location of the MHB is determined by the expression of Otx2 in the
malformations, understanding of midbrain-hindbrain malformations caudal midbrain and Gbx2 in the rostral hindbrain; increases in the
is greatly aided by an understanding of development of that region. expression of Otx2 or decrease in Gbx2 shifts the MHB caudally, while
Therefore, a few paragraphs will be devoted to an overview of mid- decrease in Otx2 or increase in Gbx2 causes a rostral MHB shift (790).
hindbrain development. The interaction of Otx2 and Gbx2 also specifies the location of the isth-
mus organizer (IsO, Fig. 5-126), a critical structure that organizes gene
Patterning expression and directs specification of cell type (791,792) and the nor-
After the neural tube is formed, it develops a series of vesicles at its mal development of the cerebellum (793).
anterior (rostral) end: the prosencephalon (also called forebrain, which At the same times that AP patterning is taking place, an analogous
will divide into the dorsal telencephalon and ventral diencephalon), process is occurring along the dorsoventral (DV) axis. DV patterning
the mesencephalon (midbrain), and the rhombencephalon (hindbrain, depends on the relative amounts of dorsalizing factors (members of
which will divide into the rostral metencephalon and caudal myel- the BMP family, which are produced by non-neural ectoderm) and
encephalon). This differentiation along the anteroposterior (AP) axis ventralizing factors (members of the SHH family, which emanate from
(also called the rostral-caudal axis) is called patterning, a name given the notochord and floor plate) (714,792,794). Along the DV axis, the
to the early differentiation of the neural tube (788). The mechanisms mesencephalon is divided into the tegmentum (ventral region) and
that result in early AP patterning are currently poorly understood and, tectum (dorsal region), while the metencephalon is divided into the

Barkovich_Chap05.indd 468 5/6/2011 9:00:14 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 469

FIG. 5-126. Anteroposterior patterning and the Isthmus Organizer. Regionalization of the
brain starts with the formation of patterning centers that secrete signaling molecules such as
the FGFs. Fgf8 and Fgf17 are important signaling molecules at both the anterior forebrain and
the midbrain-hindbrain junction. In the forebrain, it helps to direct formation of the prefron-
tal cortex and other rostral structures by inducing cells to secrete the transcription factor Pax6.
At the midbrain-hindbrain junction, the patterning center known as the IsO is induced to
secrete Fgf8 and Fgf17 by the interaction of transcription factor Gbx2 from the rhombenceph-
alon and Otx2 from the caudal mesencephalon. The secretion of Fgf8 and Fgf17 then induces
further changes crucial to formation of the midbrain-hindbrain junction and the formation
of the cerebellum. The junction of the prosencephalon (pros) and mesencephalon is directed
by the interaction of Pax6 from the caudal prosencephalon (diencephalon) and En1/Pax2 from
the rostral mesencephalon. Changes in the concentration of Gbx2 or Otx2 alter the location of
the midbrain-hindbrain junction and can affect cerebellar formation, as the cerebellum forms
from the most rostral portion of the hindbrain. Similarly, alterations of Pax6 or En1/Pax2 will
alter the location of the diencephalic-mesencephalic junction.

pons (ventral region) and the cerebellum (dorsal region). The subtype nuclei migrate radially outward from the ventricular zone (Fig. 5-127)
of the neurons in these regions is specified by expression of local HOX (802). In contrast, the neurons of the granular layer of the cerebel-
genes (795), the influence of local organizers such as the IsO (Fig. 5-126 lar cortex and of the glutamatergic deep cerebellar nuclei originate
[796]), and on the presence of graded doses of signaling molecules, in the rostral rhombic lips and have more complex migration routes
such as SHH and BMP, from the floor plate and roof plate (792), all that are guided by adhesion molecules, neurotrophins, and repulsive
influenced by local organizers, especially the IsO (796–798). molecules that may be on the surface of cells or in the interstitium
(Fig. 5-127) (793,803–807). Deep cerebellar nuclear neurons in mice
Cell Proliferation and Migration migrate rostrally in a subpial stream, express sequentially changing
On a macroscopic scale, the pontine flexure appears at about the fifth transcription factors (Pax6, Tbr2, and Tbr1) as they migrate to a
gestational week, at which time the fourth ventricle has a thin roof nuclear transitory zone (NTZ). A subset of rhombic lip-derived cells
and laterally positioned dorsal (alar) plates (25,799). During the fifth also express reelin, a key regulator of Purkinje cell migrations. In later
gestational week, cellular proliferation within the alar plates in con- stages of development, the NTZ organizes into distinct deep cerebel-
junction with formation of the pontine flexure forms the rhombic lar nuclei (Fig. 5-127) (808). Other cells migrate tangentially from the
lips, situated at the lateral walls of the fourth ventricle. The neuroepi- rhombic lips, over the cerebellar surface to form a transient EGL (see
thelial zones, in the roof of the fourth ventricle and the rhombic lips, Fig. 5-127), which acts as a secondary germinal matrix (800,809,810).
are the locations of the germinal matrices where the cells of the cer- The EGL forms between the 10th and 11th postconceptional weeks
ebellum and many brainstem nuclei will form (Figs. 5-127 and 5-128) and persists until about 15 postnatal months. After migrating through
(800,801). Between 9 and 13 postconceptional weeks, the Purkinje cells the EGL, the granule cell neuroblasts migrate inwards between clus-
of the cerebellar cortex and GABAergic neurons of the deep cerebellar ters of Purkinje cells with the presumed aid of glial (Bergman) fibers,

FIG. 5-127. Embryology of the cerebellar cortex. The cells that form the
cerebellum are generated in germinal zones lining the fourth ventricle. The
cerebellar ventricular zone in the rostral dorsal rhombencephalon gener-
ates neurons that will become GABAergic Purkinje cells and interneurons.
Some neurons migrate dorsally into the developing cerebellar hemispheres,
while others migrate to a transient nuclear transitory zone (not shown)
before migrating to form the deep cerebellar nuclei. Lateral to the cerebellar
ventricular zones are the rostral rhombic lips, where glutamatergic neurons
(granule neurons, projection neurons of the deep cerebellar nuclei, unipolar
brush cells) are generated. Some of these migrate to the nuclear transitory
zone and the deep cerebellar nuclei, while others migrate dorsally and later-
ally to form a transitory EGL. The developing granule cells migrate through
the EGL and undergo many mitoses there before terminating their migration
and then coursing inward, through the cerebellar molecular and Purkinje cell
layers before finally terminating their migration in the internal granular layer
of the cerebellar cortex. Cells formed in the more caudal rhombic lips form
the brainstem nuclei that connect to the cerebellum (see text).

Barkovich_Chap05.indd 469 5/6/2011 9:00:15 PM


470 Pediatric Neuroimaging

FIG. 5-128. Brainstem embryology. Many of the neu-


rons of brainstem nuclei that connect to the cerebellum
are formed in the caudal rhombic lips (RL). These cells
initially migrate tangentially to the surface of the brain-
stem (1) until they receive chemical signals directing
them to either cross the midline (2) or turn and migrate
radially inward for a variable distance (3) before stop-
ping at the site where a nucleus will form. These neurons
then sprout axons that migrate through the milieu of the
developing brainstem (4) to form synapses with specific
targets.

to form the internal granular layer (Fig. 5-127) (25,811,812). Also nerve from rhombomeres 4–5 (819). Due to their compartmental
formed at this time are the stellate and basket cells of the cerebel- identity, the neuronal progenitors display programmed migratory
lar cortex (25,799,801,813,814). Several genes, the most important behaviors and send axons along defined trajectories to their peripheral
being PAX6, ZIC1, and MATH1, support granule cell viability and targets. While the position of the neural cell progenitors along the AP
migration, while the Bergman glia are preserved by the expression of axis determines the identity of the nucleus, its sensory or motor func-
PAX3 (815,816). Malexpression of these genes likely causes cerebellar tion is determined by its position along the DV axis. Graded expres-
malformations. sion of specific chemicals (SHH, Pax6, and Nkx.2.2) along the DV axis
The cerebellum contains five main classes of neurons, of which two appears to generate domains conducive to either motor (ventral) or
(Purkinje cells and inhibitory interneurons) produce GABA as a neu- sensory (dorsal) neuron development (819). Another chemical, the
rotransmitter (these are called GABAergic neurons) and three (deep cer- paired-like homeodomain protein Phox2b, is required for the forma-
ebellar nuclear projection neurons, granule neurons, and unipolar brush tion of all branchial and visceral, but not somatic, motor neurons in
cells) use glutamate as a neurotransmitter (glutamatergic neurons). the hindbrain (820).
Morphological and genetic approaches both suggest that all GABAer-
gic types are produced from the cerebellar ventricular zone (Fig. 5-127) Cell Connections (Neurite Formation, Axon Migration,
(808). Among glutamatergic types, deep cerebellar nuclear projection and Synapse Formation)
neurons come from the rhombic lip (808). Granule cells are produced Although multiple connections exist among cerebellar neurons, there
from the EGL, which is a rhombic lip derivative (809,817). The origins are only two major afferent projections to the cerebellar cortex. Mossy
of unipolar brush cells have been difficult to discern (818), but new data fibers, from the basis pontis and the spinal cord make contact and syn-
suggest that they are likewise produced from the rhombic lip (808). apse with granule cells, whereas climbing fibers from the contralateral
Together, these findings suggest that most or all glutamatergic neurons inferior olives make contact with Purkinje cell dendrites (801,814,821).
in the cerebellum originate from the rhombic lip (Fig. 5-127). Thus, cer- In addition, axons from locus coeruleus, raphe nuclei, and substantia
ebellar neurogenesis seems divided according to the type of neurotrans- nigra synapse with Purkinje cells. The only efferent fibers from the cer-
mitters that the cells primarily synthesize and secrete, much as cerebral ebellar cortex are from the Purkinje cells, which connect with many
neurogenesis is divided between the germinal zones in the more ventral brainstem nuclei, in addition to the cerebellar nuclei (822). The cere-
(subpallial) ganglionic eminences (which produce most, if not all, of bellar nuclei, in turn, connect with pontine and other brainstem nuclei
the GABAergic neurons) and the more dorsal (pallial) ventricular zones through the middle cerebellar peduncles. Most of these connections
(which produce most, if not all, of the glutamatergic neurons). develop while the cells are still migrating to their final locations.
While cells produced in the rostral rhombic lips form the cer-
ebellum, those produced in the caudal rhombic lips form brainstem Interactions of Brain and Meninges
nuclei that connect to the cerebellum: the precerebellar nuclei (inferior Two final important concepts concern the development of the cerebel-
olives, lateral reticular, and external cuneate nuclei). Thus, neurons of lum, particularly as it relates to posterior fossa cysts. The first concept
brainstem nuclei also migrate to their final location (Fig. 5-128). Their is that the development of the leptomeninges overlying the cerebel-
migration pathway consists of an initial tangential migration along the lum affect cerebellar growth; mutations in or damage to the posterior
periphery of the brainstem (some also migrate longitudinally along the fossa leptomeninges impair cerebellar development (823,824). The
AP axis of the brainstem), followed by radial migration inward. Dur- second concept concerns the complex development of the roof of the
ing or at the end of the radial migration, axons are extended to con- fourth ventricle. Bonnevie and Brodal (825) have shown that the roof
nect with the cerebellum or other CNS structures. With the exception of the fourth ventricle of the mouse is divided by a ridge of develop-
of the oculomotor (third nerve, deriving from the mesencephalon) ing choroid plexus into the anterior and posterior membranous areas
nuclei, cranial nerve nuclei are derived from rhombencephalic neu- on the 11th gestational day (Fig. 5-129). By the end of the 11th day,
ronal precursors: fourth nerve from rhombomere 1, fifth nerve from the anterior membranous area (above the choroid ridge) is incorpo-
rhombomeres 2–3, sixth nerve from rhombomeres 5–6, and seventh rated into the developing choroid plexus. The posterior membranous

Barkovich_Chap05.indd 470 5/6/2011 9:00:15 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 471

FIG. 5-129. Schematic showing normal and abnormal development of the fourth ventricle. Early in gestation,
the roof of the fourth ventricle is divided by a ridge of developing choroid plexus into anterior and posterior
membranous areas (A). Normally, the anterior membranous area is incorporated into the developing choroid
plexus (B). The posterior membranous area remains and eventually cavitates to form to the midline foramen of
Magendie (C). If the anterior membranous area is not incorporated into the developing choroid plexus (D) or if
there is delayed opening of the foramen of Magendie, the roof of the fourth ventricle can balloon posteriorly to
form a fourth ventricular-cisterna magna cyst (E), identical to what is seen in the Dandy-Walker malformation.

area (below the choroid ridge) remains; an area within it eventually medulla (resulting in a long pons and short medulla or, more com-
cavitates to form the midline foramen of Magendie. The lateral foram- monly, a short pons and long medulla) (9,10); affected patients typically
ina of Luschka open at some later, as yet unknown, time. Sometimes, have cranial neuropathies or long tract signs. Segmental shifts in the
the anterior membranous area balloons outward during development, brainstem are also seen in humans with Athabaskan brainstem dysgen-
forming a collection of ependymal lined CSF inferior to the vermis. esis syndrome (seen in Native American tribes) and Bosley-Salih-Alo-
This structure is known as Blake pouch. Persistence of Blake pouch, rainy syndrome (observed in Saudi and Turkish families), both caused
also called Blake pouch cyst, is one of the mimics of the Dandy-Walker by homozygosity for mutations of HOXA1 (832), resulting in horizon-
malformation (826–828). tal gaze abnormalities, hearing loss, facial weakness, hypoventilation,
mental retardation, and autism spectrum disorder (833). Anomalies of
Classification of Midbrain-Hindbrain the vascular system and the inner ear may be seen, as well (833).
Malformations
Disconnection Syndromes Brainstem disconnection syndromes
Several classification systems have been proposed for cerebellar malfor- (834–838) are disorders in which the superior portion of the brain-
mations (8,829,830). Most of these are morphological classifications. stem is connected to the inferior portion only by a thin cord of tis-
Although morphology is useful for imaging diagnosis, it does not help sue (Fig. 5-131). Affected infants are profoundly affected from birth,
in understanding the pathogenesis of disorders, nor the relationship of with mildly dysmorphic features (downward slanting eyes, low-set
disorders of the hindbrain to those supratentorial malformations that ears, mild micrognathia, and widely spaced nipples) (834). Neurologic
often accompany them. Moreover, classifications of cerebellar malfor- examination reveals weak suck, diminished corneal, gag, and oculo-
mations have not taken into account that the cerebellum is embryo- cephalic reflexes, pronounced flexor tone, and hyperactive deep tendon
logically a part of the dorsal pons and is, therefore, an extension of the reflexes with minimal ocular motility, and disturbed respirations that
brainstem. Recently, a classification of midbrain and hindbrain mal- require mechanical ventilation (837,838). In three reported patients,
formations has been proposed that is based on embryology and genet- the disconnection was in the lower midbrain/upper pons and in four
ics, as well as morphology (Table 5-6) (10). As this classification seems it was in the lower pons/upper medulla. Neuropathological analysis of
most useful for both understanding and diagnosis of the disorders, and two cases by Sarnat et al. (834) showed a thin midline cord passing
as it includes both midbrain and hindbrain, we will follow it in our from the upper segment to the lower segment with hypoplasia of the
discussion of malformations. cerebellar vermis and hemispheres and an anomalous basilar artery.
Histological investigation revealed a poorly organized mixture of neu-
Mid-Hindbrain Malformations Secondary to Early
rons in the tegmentum, but no evidence of any reactive astrogliosis to
Patterning Defects, or to Misspecification of Germinal
suggest hypoxia or ischemia; this was interpreted as providing evidence
Zones
in favor of a brainstem malformation, rather than a disruption. In con-
Antero-Posterior Patterning Defects trast, Barth et al. (838) found central cavitation that they interpreted a
These uncommon malformations result from abnormal segmentation syrinx and postulated a vascular cause.
of the midbrain and pons (resulting in long midbrain and short pons, Diagnosis is established by MRI, which shows a relatively nor-
(Fig. 5-130) or short midbrain and long pons [831]) or of the pons and mal upper brainstem that abruptly tapers into a thin band of tissue

Barkovich_Chap05.indd 471 5/6/2011 9:00:16 PM


472 Pediatric Neuroimaging

TABLE 5-6 Classification Scheme for Midbrain-Hindbrain Malformations


I. Malformations secondary to early antero-posterior and D. Malformation of neuronal migration that prominently affect
dorso-ventral patterning defects, or to misspecification of the brainstem and cerebellum
mid-hindbrain germinal zones 1. Lissencephaly with cerebellar hypoplasia
A. Antero-posterior patterning defects 2. Neuronal heterotopia with prominent brainstem and CBL
1. Gain, loss or transformation of the diencephalon and midbrain hypoplasia
a. Long or short midbrain 3. Polymicrogyria with cerebellar hypoplasia
2. Gain, loss or transformation of the midbrain and 4. Malformations with basement membrane and neuronal
rhombomere-1 migration deficits (cobblestone malformations)
a. Midbrain-pontine brainstem disconnection E. Diffuse molar tooth type dysplasias associated with defects in
b. Large midbrain with small pons ciliary proteins
c. Short midbrain with large pons and large anterior vermis 1. Syndromes affecting the brain with low frequency involve-
3. Gain, loss or transformation of lower hindbrain structures ment of the retina and kidney
a. Ponto-medullary brainstem disconnection 2. Syndromes affecting the brain, eyes, kidneys, liver and
b. Short pons with long medulla variable other systems
B. Dorso-ventral patterning defects III. Localized brain malformations that significantly affect the BS
1. Defects of alar and basal ventricular zones (VZs) germinal and CBL (pathogenesis partly or largely understood, includes
matrix local proliferation, cell specification, migration and axonal
2. Defects of alar VZ only guidance)
a. Cerebellar agenesis (with or without pancreatic agenesis) A. Multiple levels of mid-hindbrain
b. Profound cerebellar hypoplasia due to granule cell 1. Horizontal gaze palsy with progressive scoliosis
hypoplasia 2. Congenital fibrosis of extraocular muscles
c. Rhombencephalosynapsis 3. Duane radial ray syndrome
3. Defects of basal VZ only 4. Diffuse brainstem hypoplasia
a. Duane syndrome B. Midbrain malformations
b. Möbius syndrome 1. Dorsal midline cleft in Trisomy 14
II. Malformations associated with later generalized developmental C. Malformations of Rh1 including cerebellar malformations
disorders that significantly affect the brainstem and cerebellum 1. Cerebellar nodular heterotopia with overlying dysgenesis
(and have pathogenesis at least partly understood) 2. Cerebellar foliation anomalies
A. Developmental encephalopathies associated with mid- 3. Duplication of cerebellar hemisphere
hindbrain malformations D. Pons malformations
1. Cerebellar aplasia or hypoplasia 1. Pontine tegmental cap dysplasia
B. Mesenchymal-neuroepithelial signaling defects associated with 2. Pontine dysgenesis with dorsal or ventral clefts
mid-hindbrain malformations E. Medulla malformations
1. Dandy-Walker malformation 1. Medullary tegmental caps with callosal agenesis/hypogenesis
2. Mega cisterna magna IV. Combined hypoplasia and atrophy in putative prenatal onset
3. Retrocerebellar arachnoid cysts degenerative disorders
4. Cerebellar vermian hypoplasia A. Pontocerebellar hypoplasias
C. Malformations of neuronal and glial proliferation that promi- B. Mid-hindbrain malformations with CDG
nently affect the brainstem and cerebellum C. Other metabolic disorders with cerebellar or brainstem
1. Cerebellar tubers (tuberous sclerosis) hypoplasia or disruption
2. Lhermitte-Duclos disease/Cowden syndrome D. Cerebellar hemisphere hypoplasia (rare, more commonly
3. Microcephaly with disproportionate brainstem/cerebellar acquired than genetic, often associated with clefts or cortical
hypoplasia malformation)
Adapted from Barkovich AJ, Millen KJ, Dobyns WB. A developmental and genetic classification for midbrain-hindbrain malformations. Brain 2009;132:3199–3230,
with permission.

(sometimes not visible on imaging) that connects to the lower segment zones and structures derived from them, including abnormal forma-
of normal appearing brainstem. The cerebellum is typically small tion of brainstem nuclei, cranial nerves, or any cerebellar structures.
(Fig. 5-131). The vertebrobasilar vasculature is abnormal, with ves-
sels or the posterior circulation typically very narrow or even absent; Cerebellar Hypoplasia Cerebellar hypoplasia has many different
fetal posterior cerebral arteries are usually present. One reported case morphologic variations and many causes. For example, abnormal
showed absent cervical vertebral arteries with reconstitution via persis- development of the superior rhombic lip germinal zone may cause dif-
tent segmental arteries and PVNH (839). fuse granule cell hypoplasia (with consequent severe diffuse cerebellar
hypoplasia) while abnormal development of the cerebellar ventricu-
Dorso-Ventral Patterning Defects lar germinal zone due to mutation of the PTF1A gene causes cerebel-
Defects in dorso-ventral patterning are postulated to result in abnor- lar (and pancreatic) agenesis (840,841); surprisingly, some affected
mal development or function of specific mid-hindbrain ventricular patients may have relatively mild neurologic deficits (842). Indeed, poor

Barkovich_Chap05.indd 472 5/6/2011 9:00:17 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 473

(14,15,557,842,848–850). Some of these, such as the congenital


disorders of glycosylation (CDG) Type 1 and pontocerebellar hyp-
oplasia seem to be the result of cerebellar atrophy that begins before
birth; these are discussed in Chapter 3. Others are due to lack of for-
mation or lack of migration of the cells that compose the cerebellum
(discussed below in the section “Malformations of Neuronal Migration
that Prominently Affect the Brainstem and Cerebellum”). Unilateral
hemisphere hypoplasia might be due to early fetal injury or to genetic
mutations with somatic mosaicism (845).
Imaging of cerebellar hypoplasia shows a spectrum of findings,
depending upon the severity of the hypoplasia and the presence or
absence of associated anomalies. When hypoplasia is severe, the cer-
ebellum may appear essentially absent (Fig. 5-132A and B). When the
hemisphere is slightly larger, small fissures can be seen between the
small folia, allowing the differentiation of hypoplasia (normal fissures,
Fig. 5-132C) and dysgenesis. When less severe, the diagnosis of cerebel-
lar hypoplasia can be subtle. Localized vermian hypoplasia can best be
detected by acquiring high-resolution volumetric images, reformatting
an image in the midline sagittal plane, and looking for all vermian lob-
ules. When lobulation is normal, look for the rostrocaudal size of the
FIG. 5-130. Antero-posterior patterning defect. Sagittal T1-weighted vermis to run from the bottom of the inferior colliculus to the obex in
image shows an abnormally long midbrain (white arrows) and short pons, a neonate, and from the intercollicular sulcus (midway between the
suggesting a defect in AP patterning. superior colliculi and inferior colliculi of the quadrigeminal plate) to
the obex on the midline sagittal image; if smaller (Fig. 5-132D and E),
the vermis is hypoplastic. In the fetus, where vermian anatomy is more
correlation is found between the severity of the cerebellar deformity difficult to assess, it is easiest to measure the size of the vermis and
and the severity of the clinical syndrome; for example, some patients compare it to established standards (see Chapter 2 and published mea-
with isolated unilateral (843–845) and bilateral (842,845,846) cerebel- surements [851]); however, moderate and severe hypoplasia can be
lar hypoplasia have few or no cerebellar signs or symptoms and may detected on midline sagittal images (Fig. 5-132G). The cerebellar hemi-
be intellectually normal (847). Associated CNS anomalies (heterotopia, spheres are best assessed in the coronal plane, which best shows the
callosal anomalies, and PMG) are commonly associated, however; the horizontally oriented fissures (Fig. 5-132F and H). Note that the pons
presence of associated anomalies markedly increases the incidence of is often small in cerebellar hypoplasia (Fig. 5-132), probably because
neurodevelopmental disability (842). many axons from the cerebellum normally course through the ven-
Many cerebellar hypoplasias are the result of inborn errors of tral and central pons. When the cerebellum is small, the size of these
metabolism, congenital infections, or gross chromosomal anomalies axonal pathways is diminished. Thus, the presence of a small pons in
the setting of a small cerebellum should not automatically result in a
diagnosis of pontocerebellar hypoplasia. Pontocerebellar hypoplasia is
the name given to a distinct group of disorders, as described in the pre-
vious paragraph and, more completely, in Chapter 3. A small pons in
the setting of a small cerebellum only means that the small cerebellum
is a result of prenatal events.

Brain-stem Effects of Dorsal-Ventral Patterning Defects Defects


of the basal ventricular zone result in defects of specific cranial nerve
nuclei such as the abducens and facial nerves (852,853). Patients with
these latter disorders are sometimes described as having Möbius syn-
drome, a syndrome defined by congenital palsy of the sixth and sev-
enth cranial nerves (854); imaging findings vary (therefore, this is not
a radiologic diagnosis) from normal to pontine calcifications on CT to
pontine malformation to absence of the facial colliculus (855–858). It
is likely that Möbius syndrome is a heterogeneous disorder caused by
many different developmental and disruptive disorders involving the
pons; there is no characteristic radiologic appearance.

Rhombencephalosynapsis Rhombencephalosynapsis, originally


described by Obersteiner in 1914 (859), is an anomaly character-
ized by lack of separation of the cerebellar hemispheres, with absence
FIG. 5-131. Brainstem disconnection syndrome. Sagittal T1-weighted
or severe hypoplasia of the cerebellar vermis and midline continu-
image shows disconnection of the lower brainstem from the upper brain- ity (commonly called “fusion”) of the cerebellar hemispheres, deep
stem. The ventral pons (white arrowhead) and the cerebellum (c) are small. cerebellar nuclei, and superior cerebellar peduncles. All published
No neural tissue is seen in the location where the middle portion of the cases have been sporadic. Two patients with consanguineous parents
medulla would be expected (white arrows). have been reported (860,861). The cause is unknown, but one of the

Barkovich_Chap05.indd 473 5/6/2011 9:00:17 PM


474 Pediatric Neuroimaging

FIG. 5-132. Cerebellar hypoplasia, range of severity. Note the presence of pontine hypoplasia in all. A and
B. T1-weighted sagittal (A) and coronal (B) images show profound cerebellar hypoplasia, with only a tiny
amount of cerebellar tissue (white arrows). C. Sagittal T1-weighted image shows a very tiny vermis (white
arrow), but some normal vermian lobulation is visible. D. Sagittal T1-weighted image in a neonate shows mild
to moderate vermian hypoplasia. The normal neonatal vermis should extend from the inferior colliculi of the
midbrain tectum to the obex. E and F. Sagittal (E) and coronal (F) T1-weighted images show moderate verm-
ian hypoplasia. The cerebellar hemispheric hypoplasia is asymmetric, with the right hemisphere (white arrows
in F) smaller. Note cerebral periventricular nodular heterotopia (black arrows in F).

Barkovich_Chap05.indd 474 5/6/2011 9:00:17 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 475

FIG. 5-132. (Continued) G and H. Sagittal (G) and coronal (H) fetal MR images from different patients show moderate
vermian hypoplasia (G) and right cerebellar hemisphere hypoplasia (H).

authors has seen two cases associated with holoprosencephaly, sug- palsy with epilepsy and mental retardation to mild truncal ataxia
gesting a defect in dorsal-ventral patterning. Rhombencephalosyn- and normal cognition; most patients have some degree of cognitive
apsis is a part of the cerebello-trigeminal-dermal dysplasia syndrome dysfunction, often associated with attention deficit and hyperactivity
(OMIM 601853, also called Gomez-Lopez-Hernandez syndrome), in (869,870). Motor milestones are often delayed and physical exam will
which it is associated with trigeminal anesthesia and bilateral bands of typically show a cerebellar syndrome with dysmetria, dysdiadochoki-
temporal alopecia, as well as short stature, mental retardation, mid- nesis, and dysrhythmokinesis.
face hypoplasia, oxycephaly due to bilateral lambdoid suture synos- In addition to partial (20% of affected patients) or complete
tosis, low set ears, and fifth finger clinodactyly (862–864). It has also absence (80%) of the cerebellar vermis, autopsy studies show the most
been described in conjunction with the VACTERL-H association common brain anomalies to be midline continuity of the deep cerebel-
(vertebral anal, cardiac, tracheoesophageal, renal, and limb anomalies lar nuclei, superior cerebellar peduncles, and thalami; absence of the
with hydrocephalus) (865) and autosomal dominant polycystic kid- septum pellucidum; olivary hypoplasia; anomalies of the limbic system;
ney disease Type 1 (866). Affected fetuses may sometimes be identi- callosal agenesis or hypogenesis; and hydrocephalus (717,865). How-
fied by ventriculomegaly with absent septum pellucidum; high quality ever, many patients do not have all of these findings and a wide range
images may show midline continuity of the cerebellar hemispheres of other associated supratentorial, infratentorial, and non-neurologic
(Fig. 5-133) (865,867,868). Clinical presentation after birth is variable anomalies have been reported in association with the cerebellar defor-
and ranges from severe congenital hydrocephalus to severe cerebral mity (717,865,869,871).

FIG. 5-133. Prenatal and postnatal images of


rhombencephalosynapsis. A. Coronal fetal MRI
shows massive ventriculomegaly with absent
septum pellucidum. The cerebellum is dispro-
portionately small and appears continuous
across the midline. B. Axial fetal MRI confirms
continuity of cerebellar hemispheres across the
midline (white arrowheads) without intervening
vermis.

Barkovich_Chap05.indd 475 5/6/2011 9:00:19 PM


476 Pediatric Neuroimaging

FIG. 5-133. (Continued) C. Sagittal T1-weighted image 1 day after


birth shows massive hydrocephalus. D. Axial T2-weighted postnatal
image confirms absence of the septum pellucidum. E. Axial T2-weighted
image at the fourth ventricle confirms continuity of the cerebellar folia
and the deep cerebellar nuclei (white arrows) across the midline, con-
firming the diagnosis.

As affected patients may be initially detected due to a finding of fetal across the midline without intervening vermis. This feature is most
ventriculomegaly (Fig. 5-133), an effort should be made to identify a easily seen on coronal images through the posterior cerebellum, but
normal vermis in fetuses with severe ventriculomegaly and absent sep- it is also suggested by lack or the normal vermian folial pattern on the
tum pellucidum; if the midline sagittal image shows abnormal vermian midline sagittal image and the narrowing of the posterior aspect of the
foliation or coronal/axial images shows continuation of the cerebellar fourth ventricle on axial images (Fig. 5-134). If possible, DTI may be
hemispheres across the midline (Fig. 5-133), rhombencephalosynapsis helpful, as the color fractional anisotropy maps show axons running
should be the diagnosis. In affected neonates, imaging studies often superior-inferior in the midline instead of the normal left-right axon
show severe hydrocephalus with absent septum pellucidum and appar- orientation of the vermis (872).
ent aqueductal stenosis (Fig. 5-133C). The cerebellum should be scru- It is noteworthy that about 20% of affected patients may have a
tinized in such cases for evidence of a show a characteristic appearance small, dysplastic inferior vermis, referred to as incomplete rhomb-
of absence of the vermis with dorsal midline continuity of the cerebel- encephalosynapsis (865); in such patients, the diagnosis can still be
lar hemispheres, cerebellar nuclei (Fig. 5-133E), and superior cerebellar made by observation of continuous folia across the midline superiorly.
peduncles, in addition to the ventriculomegaly (Fig. 5-133D). The cer- Once the diagnosis is established, the cerebral cortex, ventricles, and
ebellar anomaly may be difficult to detect on CT, but may be suggested limbic system should be carefully evaluated. In addition to the previ-
if dorsal pointing of the fourth ventricle (Fig. 5-134D) is detected in ously discussed hydrocephalus and absent septum pellucidum, associ-
a patient with large lateral ventricles and absence of the septum pel- ated anomalies include cortical malformations and bilateral lambdoid
lucidum (present in >50%). The diagnosis is most easily established suture synostoses (869) (see section “Craniosynostosis Syndromes” of
on MRI by the observation that the cerebellar folia appear continuous this chapter).

Barkovich_Chap05.indd 476 5/6/2011 9:00:20 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 477

FIG. 5-134. Classic rhombencephalosynapsis. Sagittal T1-weighted image (A) shows abnormal foliation of the vermis.
Coronal T1-weighted image (B) and axial T2-weighted image (C) show the continuity of the cerebellar white matter (white
arrows) across the midline. Slightly more rostral T2-weighted image (D) shows posterior pointing (white arrow) of the fourth
ventricle.

Malformations Associated with Generalized the elevation of the tentorium. In addition, wide variation can be seen
Developmental Disorders that Significantly Affect in families with the same genetic mutation, ranging from mild vermian
the Brainstem and Cerebellum hypoplasia to mega cisterna magna to varying severities of true Dandy-
Walker malformation (824,877). Of interest, one of the causative genes,
Mesenchymal-Neuroepithelial Signaling Defects Associated known as FOXC1 (at chromosome 6p25.3), is expressed only in the
with Mid-Hindbrain Malformations—The Dandy-Walker mesenchyme (developing leptomeninges) overlying the cerebellum and
Complex not in the cerebellum itself (824). This observation suggests (a) that
The Dandy-Walker malformation and related disorders (cerebellar abnormal mesenchymal development in the posterior fossa results in
hypoplasia, mega cisterna magna, and retrocerebellar arachnoid cysts) malformations of both the cerebellum and the overlying leptomenin-
are a group of disorders that have generated considerable confusion ges, (b) that the mesenchyme interacts with the developing cerebellum
and controversy (827,873–875). The Dandy-Walker malformation during embryogenesis, and (c) that mutations causing maldevelopment
consists of an enlarged posterior fossa with a high position of the ten- of both the cerebellum and its overlying mesenchyme are likely neces-
torium, hypoplasia or agenesis of the cerebellar vermis, and a dilated, sary for development of a true Dandy-Walker malformation. Thus,
cystic-appearing fourth ventricle that fills nearly the entire posterior retrocerebellar arachnoid cysts and enlargement of the cisterna magna
fossa (Fig. 5-135) (874,876). However, there is a wide variation in the (called mega cisterna magna, an enlarged posterior fossa secondary to
severity of the vermian hypoplasia, the size of the fourth ventricle, and an enlarged cisterna magna, but a normal cerebellar vermis and fourth

Barkovich_Chap05.indd 477 5/6/2011 9:00:21 PM


478 Pediatric Neuroimaging

ventricle, Fig. 5-136) are a part of the Dandy-Walker spectrum from of two of these four risk factors identifies patients with borderline or
an embryogenesis perspective. However, in the absence of mass effect lower intelligence 94% of the time (878).
(causing hydrocephalus) or associated cerebral/cerebellar dysgen- On imaging studies, the classic Dandy-Walker malformation is
esis, these findings seem to be of little, if any, clinical significance. If identified by hypoplasia or absence of the cerebellar vermis, hypopla-
a brain malformation or hydrocephalus is present, patients often have sia of the cerebellar hemispheres, an enormous fluid-filled fourth ven-
developmental delay (874,876,878–880). The degree of developmental tricle, and a large posterior fossa with high tentorium and elevation
delay seems to be related to the level of control of hydrocephalus, to of the torcular Herophili (Fig. 5-135) (884). The brainstem is typi-
the extent of associated supratentorial anomalies (878,881,882), and cally compressed against the clivus. The cerebellum may be markedly
to the degree of cerebellar dysgenesis, manifested as lobulation of the (Fig. 5-135), moderately (Fig. 5-137), or mildly (Fig. 5-138) hypoplas-
vermis; normal lobulation is associated with good intellectual outcome tic and the vermis markedly (Fig. 5-135), moderately (Fig. 5-137), or
whereas some have found that abnormal vermian lobulation is asso- mildly (Fig. 5-138) rotated. The mega cisterna magna may be differen-
ciated with poor intellectual outcome (880,883). Seizures, hearing or tiated by the presence of a normal (in size, lobulation, and orientation)
visual difficulties, systemic abnormalities, and other CNS abnormali- vermis. Both of these should be differentiated from the Blake pouch
ties are associated with poor intellectual development; the presence cyst, in which the inferior aspect of the fourth ventricle herniates

FIG. 5-135. Prenatal and postnatal images of a classic Dandy-Walker malformation. Sagittal (A) and axial (B) fetal MR
images show a high tentorium (white arrows), high torcular Herophili (white arrowhead), hypoplastic cerebellum (black
arrow), and large cisterna magna (C) that expands the posterior fossa. Sagittal (C) and axial (D) postnatal MR images show
that the posterior fossa and cisterna magna (C) have both enlarged, hydrocephalus has developed (note big third ventricle
and shunt catheter [small black arrows in C] and large temporal horns in D). The hypoplastic vermis (large black arrows) has
been pushed further upward. The heterogeneity in the lateral ventricles is a result of intraventricular hemorrhage.

Barkovich_Chap05.indd 478 5/6/2011 9:00:22 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 479

FIG. 5-136. Mega cisterna magna. Sagittal T1-weighted image shows a large posterior fossa (white arrows in A) with
enlargement, but no displacement, of leaves of the falx cerebelli (black arrows in B) and normally formed cerebellum,
indicating that this is not an arachnoid cyst or a Dandy-Walker malformation.

through the foramen of Magendie into the vallecula and retrovermian this group, the cerebellar hemispheres may appose each other inferiorly
cistern (826,827,875). However, no precise MRI characteristic allows after the posterior fossa cyst has been shunted; midline sagittal images
separation of the mega cisterna magna from a Blake pouch cyst or from may then simulate the appearance of an intact vermis (Fig. 5-139).
a mild Dandy-Walker malformation, as the walls of the cyst cannot Axial or coronal images will show the cerebellar hemispheres in appo-
be adequately identified, even with thin section steady-state imaging sition without an intervening vermis.
techniques. In view of the observation that all malformations in this
group are associated with abnormal development of the posterior fossa Malformations of Neuronal and Glial Proliferation
mesenchyme and that abnormal neurodevelopment is more related to that Prominently Affect the Brainstem and Cerebellum
parenchymal anomalies than to the cyst or hydrocephalus, this distinc- Cerebellar Tubers (Tuberous Sclerosis) Tuberous sclerosis is dis-
tion may be academic. cussed in Chapter 6 (Phakomatoses).
Associated anomalies are common: hydrocephalus in 70% to 90%,
callosal anomalies in as many as 30%, PMG or gray matter heteroto- Lhermitte-Duclos Disease/Cowden Syndrome (Dysplastic Cer-
pia in 5% to 10%, and occipital cephaloceles in up to 16% (878,879). ebellar Gangliocytoma) Also known as diffuse hypertrophy of the
Syringohydromyelia may be present if the cyst obstructs CSF circula- cerebellar cortex, dysplastic cerebellar gangliocytoma was first reported
tion through the foramen magnum (885). With any of the anomalies in by Lhermitte and Duclos in 1920 (886). Depending upon the source, it

FIG. 5-137. Sagittal T1-weighted image shows a moderate Dandy-Walker FIG. 5-138. Sagittal T1-weighted image shows a malformation in the
malformation with only moderate vermian hypoplasia and moderate Dandy-Walker spectrum with mild vermian hypoplasia combined and
posterior fossa enlargement. severe cisternal enlargement.

Barkovich_Chap05.indd 479 5/6/2011 9:00:23 PM


480 Pediatric Neuroimaging

FIG. 5-139. Dandy-Walker malformation after shunting of the posterior fossa cyst. Sagittal T1-weighted image (A) shows the
corpus callosum is hypogenetic with only the genu present. The posterior fossa is enlarged as a result of a high tentorium. On first
glance, the vermis looks intact. However, the vermis is hypoplastic (arrows mark the lower limit) with cerebellar hemispheres in
the midline beneath it. The axial T2-weighted image (B) at the level of the inferior fourth ventricle shows the cerebellar hemi-
spheres in apposition (arrows) without an intervening vermis.

may be classified as a tumor, a malformation, or part of a phakomato- Enhancement has been reported after intravenous administration
sis syndrome. Patients may become symptomatic at any age as a result of paramagnetic contrast (899), but it appears to be uncommon
of mass effect from the lesion with resultant intracranial hypertension (Fig. 5-141). Mass effect from the lesion can cause cerebellar tonsillar
(887–889). Cerebellar signs are usually mild or absent (890). Alterna- herniation, often described as a Chiari I malformation, and associated
tively, the anomaly may be discovered incidentally at autopsy or during syringomyelia (900). Diffusion-weighted imaging shows diffusivity sim-
imaging for an unrelated condition (887–889). Associated anomalies ilar to cerebellar cortex (153,901,902), while perfusion imaging shows
may include megalencephaly, heterotopia, microgyria, polydactyly, increase in relative cerebral blood volume, relative cerebral blood flow,
partial gigantism, macroglossia, and multiple visceral hamartomas and and mean transit time (900). Proton MR spectroscopy with intermedi-
neoplasms (890,891). The visceral lesions are nearly identical to those ate (135 milliseconds) echo time shows rather characteristic findings of
seen in Cowden disease (a multiple hamartoma syndrome character- elevated lactate and slightly reduced NAA (by about 10%), myo-inositol
ized by mucocutaneous manifestations of trichilemmomas, related fol- (by 30%–80%), and choline (by 20%–50%) (153,900,902,903). The
licular malformations, and distinctive hyalinizing, mucinous fibromas MRS, therefore, is a very useful technique to help distinguish this local-
and oral papillomas [892]), suggesting an association between the two ized dysplasia from neoplasm, in which choline is nearly always elevated
disorders; this suggestion is supported by reports of families with evi- (see Chapter 7). FDG PET and Tl-201 SPECT show increased uptake of
dence of both syndromes (893,894). Cowden disease is an autosomal tracer and, therefore, are not good tests to use for this distinction (902).
dominant disorder, caused by mutation of the tumor suppressor gene
PTEN at chromosome 10q23.3 [895]), that is associated with increased Microcephaly with Disproportionate Brainstem/Cerebellar
frequency of malignancies and other lesions of the breast, thyroid, Hypoplasia
colon, and pelvic adnexa (896). Other CNS manifestations of Cowden Cerebellar Hypoplasia due to CASK Mutations A familial disorder
disease include macrocephaly, meningiomas and hearing loss. Not sur- consisting of microcephaly and mental retardation with abnormal cere-
prisingly, in view of this association, familial cases of Lhermitte-Duclos bral cortical development and profound pontocerebellar hypoplasia has
syndrome have been reported (891,896). recently been associated with heterozygous loss-of-function mutations
Pathologically, Lhermitte-Duclos disease consists of a sharply mar- of a gene called CASK, located at chromosome Xp11.4 (904). The pro-
ginated region of enlarged cerebellar cortex. Typically, a portion of one tein product, CASK, is thought to have several functions during devel-
cerebellar hemisphere is involved; the process may extend into the ver- opment, including enhancing transcriptional activity of TBR1, a protein
mis or, rarely, the contralateral hemisphere (717). Microscopically, one that regulates expression of the protein Reelin (an important substance
sees a thick layer of ganglion cells replacing the granular layer of the in neuronal migration and cortical lamination of the cerebrum and cer-
cerebellar cortex, a thick hypermyelinated marginal layer, and a thin ebellum [148,905]). Indeed, neuropathologic analysis of a patient with a
Purkinje cell layer (717). hemizygous CASK mutation showed a severely hypoplastic cerebellum,
CT scans show the presence of a nonspecific hypodense cerebellar a small brainstem, and abnormalities of cortical layering in the cerebrum
mass (897). MRI shows a sharply marginated cerebellar mass with T1 and cerebellum. MRI shows profound cerebellar and brainstem hypopla-
and T2 prolongation; coursing through the mass are curvilinear struc- sia (Fig. 5-142), often with blurring of the cerebral cortical-white matter
tures of gray matter intensity that appear to be the cerebellar cortex (Fig. junction and abnormal cerebral sulcation (904). A distinctive feature is
5-140). In apparent contradiction of pathological findings, the cortical the normal-to-large appearing corpus callosum (Fig. 5-142), which is an
ribbon appears of normal or slightly less than normal thickness (898). unusual finding in microcephaly.

Barkovich_Chap05.indd 480 5/6/2011 9:00:25 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 481

FIG. 5-140. Lhermitte-Duclos syndrome. Sagittal T1-weighted image (A) shows hypointensity (black arrows) of the posterior
left cerebellar hemisphere. Axial T1-weighted (B) image shows the hypodense region has mass effect, displacing fourth ventricle
(open white arrow) and medulla (solid black arrows).

Malformations of Neuronal Migration that Prominently Affect Diffuse Molar Tooth Type Dysplasias Associated with Defects
the Brainstem and Cerebellum in Ciliary Proteins (Joubert Syndrome and Related Disorders/
These disorders, including lissencephaly, malformations due to gaps in Syndromes with “Molar Tooth” Malformation)
the pial limiting membrane, heterotopia, and PMG, were discussed in The concept of molar tooth dysplasias has evolved from that of a
section “Malformations of Cortical Development” of this chapter. Not rare cerebellar malformation presenting in children with episodic
unexpectedly (because the factors determining cell proliferation and hyperpnea, abnormal eye movements, ataxia, and mental retardation
cell migration are similar supra- and infratentorially), cerebellar hyp- (906–909) to a much more complicated entity encompassing multi-
oplasia is often present in such malformations; thus, the midbrain and ple syndromes. Recent work has shown that this brainstem/cerebellar
hindbrain should be carefully examined in all such patients. malformation (called by some the “molar tooth midbrain-hindbrain

FIG. 5-141. Lhermitte-Duclos syndrome. Axial T2-weighted image (A) shows heterogeneous hyperintensity
in the cerebellum with mass effect (white arrows) upon the pons. Postcontrast T1-weighted image (B) shows
heterogeneous, pial enhancement of the malformation.

Barkovich_Chap05.indd 481 5/6/2011 9:00:25 PM


482 Pediatric Neuroimaging

FIG. 5-142. Cerebellar hypoplasia


secondary to mutation of CASK. Sagit-
tal T1-weighted image (A) and coronal
T2-weighted image (B) show moder-
ate cerebellar and pontine hypoplasia.
The cerebrum has mildly diminished
white matter and some blurring of
the cortical-white matter junction. A
small periventricular nodular hetero-
topion is present (white arrow) in the
left trigone.

malformation” because axial images through the midbrain have the


appearance of a tooth) is seen with many associated anomalies that
TABLE 5-7 Joubert Syndrome and
encompass many syndromes; these are called, as a group, Joubert
syndrome and related disorders (JSRD) (235,910–915). The genetic Related Disorders
basis of JSRD appears to be highly heterogeneous (234,914–917).
Most of the related syndromes seem to be the result of mutations to Primary criteria
genes encoding ciliary proteins (912–915,917), which are important Neurological signs Hypotonia/ataxia
in a wide range of functions, including ciliogenesis, body axis forma- Developmental delay
tion, renal function, brain development, and ocular development. Oculomotor apraxia
Therefore, mutations that affect these structures cause a wide range
of disorders (918). Radiologic hallmark Molar tooth sign
The classification of JSRD has recently been revised (235). Zaki Occasional features Mental retardation
et al. (235) divided these malformation syndromes into four separate (seen in all forms) Breathing abnormalities
disorders: JS, COACH (Cerebellar vermis hypoplasia, Oligophrenia, Postaxial polydactyly
Ataxia, Ocular Coloboma, Hepatic fibrosis) syndrome, CORS (Cere- Mild retinopathy
bro-Oculo-Renal Syndrome), and Oculo-Facial-Digital Syndrome
Secondary criteria
Type VI (OFD-VI syndrome). Patients with all JSRD syndromes have
hypotonia, ataxia, developmental delay (cognitive and motor), oculo- Joubert syndrome Mild retinopathy
motor apraxia, and the “molar tooth sign” on brain MRI; occasional Mild nephropathy
features (seen in all forms) include mental retardation, postaxial poly- COACH syndrome Hepatic fibrosis
dactyly, mild retinopathy, and PMG (235,919). Other defining charac- (at least one) Coloboma (choroidal or retinal)
teristics are listed in Table 5-7 (235).
Cerebello-oculo-renal Ocular signs (at least one)
The genetic bases of these various disorders are being elucidated,
syndrome (CORS) Retinopathy (Typically congenital
although they are complex. Nine genetic loci associated with JSRD have
blindness)
been identified at chromosomes 9q34.3 (JBTS1, gene is unknown),
Coloboma
11p11.2-q12.3 (JBTS2, gene is unknown), 6q23 (JBTS3, AHI1, encod-
Renal signs (at least one)
ing Jouberin), 2q13 (JBTS4, NPHP1), 12q21 (JBTS5, CEP290), 8q22
Cysts
(JBTS6, TMEM67), 16q12 (JBTS7, RPGRIP1L), 3q11.2 (JBTS8,
Nephronophthisis
ARL13B), and 4p15.3 (JBTS9, CC2D2A) (234,916–918,920,921). Of
Renal failure
interest, AHI1 is involved in normal axon outgrowth and decussation
(922), which probably relates to the lack of decussation of many white Oro-facial-digital Oro-facial signs (at least one)
matter tracts in molar tooth malformations (see below). The multiplic- syndrome Type VI Cleft lip/palate
ity of causative genes implies that the molar tooth midbrain-hindbrain Tongue tumors
malformation may be the result of several different processes; there- Notched upper lip
fore, identification of the molar tooth sign does not, in itself, allow a Digital signs (at least one)
diagnosis to be made. Mesaxial polydactyly
Few neuropathology studies of affected individuals have been Preaxial polydactyly
reported. Friede reported a small, dysplastic cerebellar vermis with Bifid digits
midline clefting, dysplasias and heterotopia of cerebellar nuclei, near
Adapted from Zaki MS, Abdel-Aleem A, Abdel-Salam GMH, et al. The molar
total absence of the pyramidal decussation, and anomalies in the struc-
tooth sign: a new Joubert syndrome and related cerebellar disorders classification
ture of the inferior olivary nuclei, descending trigeminal tract, solitary system tested in Egyptian families. Neurology 2008;70:556–565, with permission.
fascicle, and dorsal column nuclei (717). Yachnis and Rorke (923)

Barkovich_Chap05.indd 482 5/6/2011 9:00:27 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 483

reported absence of decussation of the superior cerebellar peduncles diagnosis will depend on whether other anomalies are found and,
and central pontine tracts, as well. Saito et al. found that “the midline ultimately perhaps, on the genetic analysis.
structures of the brainstem are disordered both structurally and func- Patients with JSRD are frequently referred for fetal MRI after rou-
tionally” in JS (924). tine fetal sonography shows an enlarged retrovermian CSF space (a
The imaging features of the molar tooth malformation are char- “large cisterna magna”). These patients should be carefully scrutinized
acteristic (908). Sagittal images show a small vermis that is situated for a small vermis (the midline cleft is difficult to see in the fetus), a
abnormally high such that the fastigium of the fourth ventricle is at narrow midbrain-pons junction (Fig. 5-144A and B) and the pres-
the upper pons or at the pontomesencephalic junction, rather than ence of a molar tooth sign on axial images, and the presence of large,
its normal position at the middle or low-middle pons. Thin sagittal horizontal superior cerebellar peduncles on coronal (Fig. 5-144C) or
images will often show that the midbrain-pons junction is abnormally parasagittal images (925). Detection of these imaging features should
thin (Fig. 5-143). The vermian folial pattern is abnormal (Fig. 5-143). strongly suggest the diagnosis of JSRD and should initiate a search for
Coronal images typically show a cleft (Fig. 5-143B) in the superior associated findings such as renal cysts, microphthalmia (associated
vermian midline. The small size of the dysmorphic vermis results in a with retinal dysplasia), ocular colobomas, polydactyly, hamartomas of
triangular-shaped mid-fourth ventricle (Fig. 5-143C) and a “bat-wing” the tuber cinereum, or tumors of the tongue.
shaped superior fourth ventricle on axial images. Inferiorly, below the
small vermis, the two cerebellar hemispheres come into apposition in Localized Brain Malformations that Significantly Affect
the midline (Fig. 5-143C). The superior cerebellar peduncles do not the Brainstem and Cerebellum
cross in the dorsal midbrain; they are usually large, nearly horizontal Malformations Affected Multiple Levels of Mid-Hindbrain
and can be clearly seen as they extend between the midbrain and cer- Horizontal Gaze Palsy with Progressive Scoliosis In the early
ebellum, surrounded by CSF (Fig. 5-143). The midbrain is small in its 1970s, Dretakis and Konodoyannis (926,927) and Crisfield (928)
anterior-posterior diameter, particularly in the midline (Fig. 5-143A), reported a group of patients with congenital scoliosis associated with
probably because of the absence of the decussation of the superior cer- horizontal gaze palsy. This disorder was subsequently found to be
ebellar peduncles. The characteristic appearance of the midbrain on autosomal recessive, caused by mutations of the ROBO3 gene, located
axial images, with the enlarged superior cerebellar peduncles and the at chromosome 11q23-q25 (929,930). The ROBO3 protein has been
absence of their decussation, gives the classic “molar tooth sign” on shown to be important for midline crossing of axons in the brainstem
axial images (Fig. 5-143D). Diffusion tensor tractography confirms the and spinal cord (930). Affected patients have congenital horizontal
absence of decussation of the superior cerebellar peduncles (872). gaze palsy: no horizontal smooth pursuit, saccades, optokinetic nys-
From the imaging perspective, it is important to remember two tagmus, or vestibuloocular responses, but vertical motions are normal.
facts: first, multiple syndromes have a molar tooth posterior fossa mal- Convergence movements may be markedly reduced. Scoliosis devel-
formation; and second, these groups of patients cannot be differen- ops during childhood and has variable severity (931). Diagnosis can be
tiated by neuroimaging alone. Therefore, all patients with the molar made by MRI, which shows characteristic anomalies of the brainstem.
tooth malformation should be screened for supratentorial anomalies Midline sagittal images show a small pons, but the diagnosis is estab-
(hypothalamic hamartoma, PMG or other malformations of cortical lished from axial images, which show a dorsal midline pontine cleft
development), ocular, hepatic, and renal disease (235). The ultimate and both dorsal and ventral midline clefts of the medulla (Fig. 5-145).

FIG. 5-143. Molar tooth malformation characteristic of Joubert syndrome and related disorders. Sagittal T1-weighted
image (A) shows the dysplastic, small cerebellar vermis (black arrows) that lies abnormally high. Note the abnormal folial
pattern. The isthmus (white arrow) is abnormally narrow. Coronal T1-weighted image shows the large superior cerebellar
peduncles (white arrows) and the vermian cleft (white arrow). Axial T1-weighted images (C and D) show the triangular
fourth ventricle (C), resulting from absence of the inferior vermis, and the “molar tooth” appearance of the midbrain (D)
secondary to the narrow isthmus (small white arrow) and the large superior cerebellar peduncles (larger white arrows).

Barkovich_Chap05.indd 483 5/6/2011 9:00:27 PM


484 Pediatric Neuroimaging

FIG. 5-143. (Continued)

FIG. 5-144. Fetal MR images of a molar tooth malformation


shows a small vermis (black arrows) and large cisterna magna
(C) on the sagittal image (A), narrow isthmus (black arrows)
on the axial image (B), and large horizontal superior cerebellar
peduncles (black arrows in C).

Barkovich_Chap05.indd 484 5/6/2011 9:00:28 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 485

FIG. 5-145. Horizontal gaze palsy with progressive scoliosis. Midline


sagittal T2-weighted image of the brain (A) shows a small brainstem, in
particular a small pons, with abnormal dorsal concavity (black arrows).
Axial T2-weighted images (B and C) show the midline brainstem cleft
(black arrows) resulting from a lack of axonal crossing of the midline.

The appearance of the medulla has been described as looking like a these disorders is CFOEM2, an autosomal recessive disorder in which
butterfly (Fig. 5-145C) (932). The cerebellum and midbrain appear affected individuals are born with bilateral exotropia and ptosis due to
normal, while spine images show progressive scoliosis with no under- mutations of the PHOX2A gene at chromosome 11q13 (938). Diagno-
lying spinal cord lesion. Diffusion tensor tractography shows no sis is confirmed by MRI, which shows a normal appearing brainstem,
decussation of rostro-caudally directed axonal bundles (ventral or but very small or absent oculomotor nerves (939). Very thin sections
dorsal) (933,934), which is concordant with neurophysiological stud- using steady-state imaging techniques (constructive interference in
ies (933,935). Images of the spine show progressive scoliosis. steady state [CISS], fast imaging employing steady-state acquisition
[FIESTA]) to optimally identify the cranial nerves should be used in
Congenital Fibrosis of Extraocular Muscles Congenital fibrosis of these cases.
the extraocular muscles (CFOEM) is one of a group of congenital neu-
romuscular diseases that are currently called congenital cranial dysin- Midbrain Malformations
nervation disorders. They include congenital, nonprogressive sporadic Tectal Enlargement Two groups of patients have been described in
or familial abnormalities of cranial musculature that result from devel- which striking enlargement of the midbrain tectum was identified (9).
opmental abnormalities of one or more cranial nerve with primary or The most common of these is patients with the oculocerebrocutaneous
secondary muscle dysinnervation (936). Three phenotypes of CFOEM (also called OCCS and Delleman) syndrome, a disorder characterized
have been described, all of which are the result of maldevelopment by a colobomatous ocular malformation, skin appendages (predomi-
of the oculomotor nerve (cranial nerve III) and its corresponding nantly located near the orbit), and cerebral malformations consist-
motoneurons; the precise phenotype depends upon which of the five ing of unilateral frontal PMG with PVNH, agenesis of the corpus
subnuclei of CN III are affected (819,937). The best characterized of callosum (sometimes with interhemispheric cyst), large dysmorphic

Barkovich_Chap05.indd 485 5/6/2011 9:00:29 PM


486 Pediatric Neuroimaging

tectum, absent cerebellar vermis, and small cerebellar hemispheres ing from the interpeduncular cistern posteriorly to the aqueduct that
(246,247,940). Milder cases of OCCS may have normal orbits and cere- seems nearly continuous with the third ventricle (941,942). Indeed, the
bral hemispheres; they are identified by enlarged inferior colliculi and thalami seem to extend inferiorly into the cerebral peduncles, as if the
absent cerebellar vermis (Fig. 5-146). Another group of patients with diencephalic-midbrain junction were inferiorly displaced and resulting
tectal enlargement was characterized by an enormously enlarged quad- in a widened midbrain (10). The aqueduct may be elongated anteriorly,
rigeminal plate, but no other brain anomalies (9). The mechanism by as in an aqueductal “pouch” that appears to be an inferior extension
which these tectal anomalies develop is not known (9). of the third ventricle into the midbrain. In our experience, cerebellar
hypoplasia is nearly always seen in congenital cases.
Midbrain Clefts Midbrain clefts are malformations of unknown cause
that result in oculomotor dysfunction, internuclear ophthalmoplegia, Midline Dorsal Clefts: Trisomy 14 The authors have seen a single
and ptosis, probably due to involvement of the oculomotor nuclei brain study of a patient with trisomy 14 in which a dorsal cleft was
and median longitudinal fasciculus (941,942). In severe cases, ataxia, present in the midbrain, giving the appearance on sagittal images of
dysarthria, and deafness may be present (942,943). It is likely that this a markedly widened aqueduct. Oculomotor dysfunction was present.
abnormality can be acquired due to intracranial hypotension, and this The embryogenesis of this malformation is unknown.
is by far the most common cause (944). Therefore, all patients with this
appearance should be evaluated for intracranial hypotension. Some cases Malformations of Rh1 Including Cerebellar Malformations
may be developmental (941,942,945), as many patients are initially Cerebellar Nodular Heterotopia with Overlying Dysgenesis
discovered in childhood and have associated brain anomalies (10). In Although abnormal clusters of gray matter in the cerebellar white mat-
either case, imaging shows a midline cleft in the mesencephalon, extend- ter may be found incidentally, they usually coexist with other cerebellar

FIG. 5-146. Oculocerebrocutaneous (Delleman) syndrome, mild


case. Sagittal T1-weighted image (A) shows marked dorsal elongation
of the inferior colliculi (white arrow) and a dysmorphic-appearing
midline cerebellum. Axial T1-weighted images (B and C) show the
dysmorphic tectum (white arrows in B) and absence of the vermis
(C). In more severe cases, the corpus callosum is absent and polymi-
crogyria is present in a cerebral hemisphere. (These images courtesy
Dr. Junichi Takanashi, Chiba, Japan.)

Barkovich_Chap05.indd 486 5/6/2011 9:00:30 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 487

FIG. 5-147. Cerebellar heterotopia with


occipital dysplasia. Coronal T2- weighted
image (a) shows a large cluster of hetero-
topic neurons (white arrows) in the deep
right cerebellar hemisphere; the overlying
cortex has abnormal sulcation. Axial T2
weighted image through the lower cere-
brum (b) shows enlargement and abnormal
sulcation (white arrows) of the right occipi-
tal lobe. Note the hypoplastic left ocular
globe (white arrowheads).

malformations, most commonly abnormal foliation of the overlying affected patients are typically mildly affected (autistic features, speech
cerebellar cortex (717). Symptoms seem to arise from accompanying delay, ocular apraxia) or asymptomatic (845,949); the diagnosis may
malformations and not from the heterotopia themselves, but the source be made incidentally when imaging is performed for unrelated rea-
of the symptoms is difficult to determine due to poor understanding sons. When focal, cerebellar foliation anomalies are typically isolated
of the functions of the cerebellum (946–948). Imaging shows variably anomalies. These anomalies are thought to result from disruption of
sized nodules of gray matter intensity within the cerebellar white mat- normal cerebellar cortical development by a genetic cause, such as a
ter, usually associated with abnormal overlying foliation (Fig. 5-147). mutation in a gene that alters the proliferation and migration of cer-
In our experience, the occipital cortex is nearly always dysmorphic in ebellar cortical neurons (particularly granule cells), or by a destructive
these patients (Fig. 5-147B). event in utero: fetal infection (especially with cytomegalovirus), prena-
tal hemorrhage, hypoxia-ischemia (which seems to particularly affect
Cerebellar Foliation Anomalies (Cerebellar Cortical Dysgene- the Purkinje cells), or exposure to toxins such as alcohol or radiation
sis) Often referred to as cerebellar PMG or (in pathology literature) as (845,949,950).
cerebellar heterotaxias, cerebellar cortical dysgenesis describes a group Focal hemispheric dysgenesis is usually identified by abnormal
of conditions in which the folial pattern of the cerebellar cortex is dis- contour of the affected hemisphere or by lack of the normal arboriza-
turbed, usually in association with diminished volume of the affected tion of the cerebellar white matter (Figs. 5-148 and 5-149). (Normally,
portion of the cerebellum. In the absence of supratentorial anomalies, the cerebellar fissures are radially oriented from the deep cerebellar

FIG. 5-148. Abnormal vermian foliation. Normal sagittal T1-weighted image (A) shows the primary fissure and prepyramidal
fissures separating the vermis into three sections of similar size. Sagittal T1-weighted image (B) shows abnormal foliation of the
vermis, diagnostic of vermian dysgenesis. Note that no normal primary fissure or prepyramidal fissure can be identified.

Barkovich_Chap05.indd 487 5/6/2011 9:00:31 PM


488 Pediatric Neuroimaging

FIG. 5-149. Two cases of focal cerebellar dysgenesis. Coronal T1-weighted images show (A) focal cortical thinning and
abnormal foliation in the right lateral posterior cerebellum (black arrows) and (B) focal left cerebellar hypoplasia with a
vertical cleft (white arrows).

nuclei on coronal images and the white matter bundles follow that typically PMG or cobblestone cortex (see section “Malformations of
same orientation.) Abnormal fissure orientation (Fig. 5-149A) should Cortical Development” in this chapter) (4,717). When the dysgenesis
raise suspicion for focal dysgenesis; loss of volume or a frank cleft is unilateral and the affected hemisphere is small, the anomaly is better
may accompany the abnormal fissuration (Fig. 5-149B) (845). Focal classified as cerebellar hemisphere hypoplasia (discussed later in this
vermian dysgenesis is best identified on sagittal images by abnormal section).
contour or location of the fourth ventricle, or by abnormal foliation;
normally, the primary fissure and the prepyramidal fissure are easily Duplication of Cerebellar Hemisphere
seen on midline sagittal images, roughly dividing the vermis into thirds The authors are aware of only two cases of duplication of the cerebel-
(Fig. 5-148A); an inability to identify them (Fig. 5-148B) should sug- lar hemisphere, one of which was associated with ipsilateral internal,
gest a malformation. Occasionally, diffuse (Fig. 5-150) malformations middle, and external ear duplication (952). In one patient, the affected
of the cerebellar cortex are encountered without associated supraten- patient had no cerebellar signs or symptoms, while the second had a
torial dysplasias (951). Diffuse cerebellar cortical dysgenesis is usu- calvarial protuberance in the occipital region, generalized hypotonia,
ally associated with malformations of cerebral cortical development, depressed deep tendon reflexes, and mild truncal ataxia. MRI in the

FIG. 5-150. Diffuse cerebellar cortical dysgenesis. Axial (A) and coronal (B) T1-weighted image show diffusely abnormal
foliation, with abnormally deep, nearly vertical fissures rather than normal fissures radiating outward from the deep cerebellar
white matter.

Barkovich_Chap05.indd 488 5/6/2011 9:00:32 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 489

sagittal and coronal planes makes the diagnosis if the supernumerary ventral and middle pons are absent. Jissendi-Tchofo et al. (954) propose
hemisphere and its peduncles can be identified. The embryologic cause three potential mechanisms, including abnormal axonal navigation,
of this malformation is not known. abnormal neuronal migration, and a combination of the two.

Pons Malformations Pontine Dysgenesis with Dorsal or Ventral Clefts Dorsal and ven-
Pontine Tegmental Cap Dysplasia Patients with this pontine teg- tral longitudinal clefts are seen in the pons in a number of conditions;
mental cap dysplasia present with cranial neuropathies; the eighth cra- they are rarely isolated malformations (9). Nearly all affected patients
nial nerve is most commonly affected, followed by the seventh and fifth have a combination of cranial neuropathies and long tract signs. The
cranial nerves. A swallowing disorder may be present, suggesting ninth clefts are believed to result from a defect in axonal navigation across
nerve involvement. Affected children may have gross deficits in motor the pontine midline, particularly a defect in ventral pontine crossing of
and cognitive function, suggesting supratentorial involvement, as well axons from the middle cerebellar peduncles; as many of the molecular
(953,954). The diagnosis is made by MRI, which shows a small ventral signals that guide axons across the midline are similar in the cerebrum
pons in association with a round or “beak-shaped” dorsal pontine protu- and the brain stem, it is not surprising that many affected patients have
berance (the tegmental cap, Fig. 5-151). The middle cerebellar peduncles anomalies of the corpus callosum (Fig. 5-152) (9). Ventral pontine
are small to nearly absent. Color fractional anisotropy maps show that clefts are also very common in patients with MEB and WWSs (see sec-
the tegmental cap is a white matter structure, composed of transversely- tion “Malformations of Cortical Development” earlier in this chapter)
oriented axons that seem to connect to the middle cerebellar peduncles (9) and are seen in horizontal gaze palsy with progressive scoliosis (see
(Fig. 5-151F) and that the normal ventral transverse pontine fibers in the above in this section) (932).

FIG. 5-151. Pontine tegmental cap dysplasia. Sagittal T1-weighted images


show ventral pontine hypoplasia and the appearance of a rounded (white
arrows in A) or beak-like (white arrows in B) “cap” over the dorsal pons. Axial
(C) and coronal (D) T1-weighted images show the “cap” (white arrows) forming
a rather flat anterior border to the fourth ventricle. Normal color FA map (E) of
the pons shows the red decussation of the middle cerebellar peduncles (white
arrows), blue corticospinal and corticopontine tracts (white arrowheads), red
transverse pontine fibers to pontine nuclei (black arrowheads), blue dorsal lon-
gitudinal tracts (black arrows) and green middle cerebellar peduncles (m). FA
map of pontine tegmental cap dysplasia (F) shows absence of the ventral decus-
sation of the middle cerebellar peduncles, a single ventral group of longitudinal
pontine fibers (blue fibers, white arrows), and the tegmental cap (red fibers, white
arrowheads), whose red color indicates transverse orientation of the fibers. The
middle cerebellar peduncles (green fibers, black arrows) are very small. Color
coding: red, transverse; blue, supero-inferior; green, anteroposterior.

Barkovich_Chap05.indd 489 5/6/2011 9:00:34 PM


490 Pediatric Neuroimaging

FIG. 5-151. (Continued)

FIG. 5-152. Brainstem cleft with callosal anomaly. Midline sagittal T1-weighted image (A) shows callosal hypogenesis
with only a small anterior corpus callosum (black arrows). The brainstem appears very thin. Axial T2-weighted image
(B) shows a large midline dorsal and ventral pontine cleft; on other images, the cleft continued into the medulla.

Barkovich_Chap05.indd 490 5/6/2011 9:00:35 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 491

FIG. 5-153. Dorsal medullary cap malformation. (A) Sagittal T2-weighted image shows nearly complete corpus
callosum agenesis and a dorsal gray matter intensity medullary mass (black arrow). (B) Axial T1-weighted image
shows the medullary cap remains of gray matter intensity, suggesting it is composed of ectopic neurons. Other images
showed hippocampal anomalies and abnormalities of the cerebellar nuclei in this patient. (These images courtesy
Dr. Susan Blaser, Toronto.)

Medulla Malformations seen and others have reported patients with cerebellar hypoplasia after
Medullary Tegmental Caps with Callosal Agenesis/Hypogen- incidentally noted intrauterine or early postnatal cerebellar hemor-
esis Rarely, dorsal medullary protuberances are identified on MRI in rhage (Fig. 5-154) (10,845). On MRI, the affected cerebellar hemi-
infants being imaged for multiple anomalies of the musculoskeletal sphere ranges in size from nearly completely absent to slightly small.
(clinodactyly, rocker bottom feet), genitourinary, or gastrointestinal The contour of the hemisphere may be smooth or irregular (with or
systems. On sagittal images, a dorsal (tegmental) cap is seen at the level without clefts). Signal intensity of the affected hemisphere is usually
of the medulla; absence or severe hypogenesis of the corpus callosum normal. Supratentorial findings range from normal to ventriculom-
is common in these patients (Fig. 5-153). The nature and cause of the egaly to schizencephaly and other cortical malformations.
“cap” are not clear at present.

Combined Hypoplasia and Atrophy in Putative Prenatal


ANOMALIES OF THE CRANIOCERVICAL
Onset Degenerative Disorders JUNCTION (THE CHIARI MALFORMATIONS)
Pontocerebellar Hypoplasias In 1891, Chiari (955) described three malformations of the hind-
The pontocerebellar hypoplasias are discussed in Chapter 3, under the brain, all of which were associated with hydrocephalus. These three
heading of “Cerebellar Atrophy.” malformations will be discussed in the order in which they were
described.
Mid-Hindbrain Malformations with Congenital Disorders of
Glycosylation
Chiari I Malformations
The CDG are discussed in Chapter 3, under the heading of “Cerebellar
Atrophy.” The Chiari I malformation is defined as caudal cerebellar tonsillar ecto-
pia; that is, caudal extension of the cerebellar tonsils below the foramen
Other Metabolic Disorders with Cerebellar or Brainstem magnum. Affected children typically present clinically with occipital
Hypoplasia or Disruption or posterior cervical headaches (especially when straining or cough-
These disorders are discussed extensively in the last section of Chap- ing), lower cranial nerve palsies, otoneurologic disturbances (such as
ter 3. disequilibrium, tinnitus, or vertigo), or dissociated anesthesia of the
extremities secondary to syringohydromyelia (956,957). Syringomy-
Cerebellar Hemisphere Hypoplasia elia is present in about 10% at the time of diagnosis (958). Impaired
Unilateral cerebellar hypoplasia is a rare anomaly. It is typically found oropharyngeal function is reported to be common in children less than
when imaging a patient with developmental delay or evaluation of a 3 years old (959). Several cases have been reported in which sleep apnea
syndrome such as Möbius or Goldenhar syndromes; occasionally, it is was attributed to the Chiari I malformation (960–962). In infants and
found incidentally when imaging for unrelated events such as trauma nonverbal children, headaches may be manifested as irritability or
or headaches (843–845). Most authors believe that it is much more crying, often associated with arching (hyperextension) of the neck
commonly acquired than genetic, as it is often associated with cer- (963). Older age at time of diagnosis is associated with increased risk
ebellar clefts or cortical malformation (10,845,949). Indeed, we have of headache and significant neurologic symptoms (958). Nystagmus,

Barkovich_Chap05.indd 491 5/6/2011 9:00:37 PM


492 Pediatric Neuroimaging

FIG. 5-154. Unilateral cerebellar hypoplasia secondary to prenatal cerebellar injury. Coronal fetal MR image
(A) shows heterogeneity (white arrows), likely hemorrhage, in the left cerebellar hemisphere. Postnatal coronal
T2-weighted image (B) confirms left cerebellar hypoplasia (black arrows) but shows no evidence of blood. (These
images courtesy Dr. Guido Gonzales, Santiago, Chile.)

particularly downbeat nystagmus, is reported to be strongly associated syndrome and Pfeiffer syndrome, can develop Chiari I malformations
with lesions of the foramen magnum, particularly with the Chiari I because the suture closure (particularly the lambdoid sutures) results in
malformation (964). a small posterior fossa and skull base, possibly exacerbated by increased
The Chiari I malformation is likely the result of several different venous sinus pressure (973–975). Other syndromes that result in a
processes. The most common type of Chiari I malformation appears small skull base (primarily chondrodystrophies), can also result in ton-
to be the result of an abnormally small bony posterior fossa, as (a) sillar ectopia/Chiari I malformations (976,977).
the clivus is often short or abnormally horizontal in affected patients Another group of patients with Chiari I malformation are those
(Figs. 5-155 and 5-156) (965,966), (b) the straight sinus is more vertical with acquired deformities of the foramen magnum that lead to basilar
than normal (Fig. 5-157), (c) a strong correlation exists between poste- invagination (978). These patients, nearly always adults, also present
rior fossa volume and degree of cerebellar ectopia (967), and (d) a high with headaches, cranial neuropathies, or syringohydromyelia. A fourth
incidence of craniovertebral anomalies has been noted (966,968,969) subgroup, actually mislabeled as Chiari I malformations, is composed
in affected patients. Up to 90% of affected patients have median basi- of patients with myelomeningoceles and mild hindbrain anomalies
lar invagination (Figs. 5-156 and 5-158), with 20% showing additional consisting primarily of tonsillar ectopia (2% of myelomeningocele
paramedian invagination. Many of the patients with basilar invagina- patients in Emery’s series of 100 autopsies of patients with myelom-
tion and shortened clivus also have C-1 assimilation (969). There is eningoceles had tonsillar ectopia as their only hindbrain anomaly
remodeling of the inferior surface of the foramen magnum that leads [979]). The patients in this subgroup have myelomeningoceles at birth,
to bony indentation between the assimilated C-1 and the superior fac- often have supratentorial anomalies such as hypogenesis of the corpus
ets of C-2. The foramen magnum is, thus, in an abnormal position that callosum, mild tectal beaking, and fenestrations of the falx, and prob-
results in an irreducible posterior odontoid invagination (Fig. 5-156) ably should be classified as Chiari II malformations with mild hind-
(970). The odontoid invagination in combination with the abnormal brain manifestations.
clivus, compounded by the tonsillar herniation and the abnormal posi- Two other groups deserve mention. The first is patients with long
tion of the inferior medulla, leads to progressive neural compromise standing “compensated” hydrocephalus or pseudotumor cerebri who,
(Fig. 5-156) (969). in fact, have chronic tonsillar herniation; indeed, up to 12% of patients
However, this theory of causation applies only to part of the group with longstanding pseudotumor cerebri may develop chronic tonsil-
that we identify by imaging as having the Chiari I malformation. lar herniation (980). The second is patients with intracranial hypoten-
A number of other subgroups of patients can be identified with tonsil- sion secondary to chronic CSF leaks (the “sagging brain”) (981,982).
lar extension through the foramen magnum and resultant compression Both of these groups of patients can present with headaches that are made
of the neural structures and CSF spaces. One cause of such malforma- worse by activity and, hence, may be considered as candidates for fora-
tions may be effects ventriculoperitoneal shunts that are placed for men magnum decompression, which is precisely the wrong therapy. The
treatment of infantile hydrocephalus (956,971). It is likely that these group with chronic increased intracranial pressure can be identified by
patients have premature closure of their cranial sutures because shunt- looking for signs of hydrocephalus (large anterior recesses of the third
ing of CSF reduces the volume of their brain; as a result, the forces ventricle, commensurately enlarged temporal horns in the absence of
required to keep the sutures open disappear. When the brain subse- large Sylvian fissures, see Chapter 8) or pseudotumor cerebri (dilated
quently grows enough to fill the cranial vault, the calvarium cannot optic nerve sheaths, compressed venous structures, empty sella) on the
grow quickly enough to keep up with brain growth; as a result, the imaging studies. Those with intracranial hypotension can be identified
brain grows downward with the tonsils ultimately protruding through by the character of their headache (made worse by standing up, better
the foramen magnum. Similar tonsillar herniation can result from cal- by lying down), associated clinical signs and symptoms (nausea, vom-
varial thickening in dysplasias of bone (972). Premature closure of mul- iting, cranial neuropathies, vertigo, hyperacusis, neck pain/stiffness)
tiple cranial and facial sutures due to genetic causes, such as Crouzon (944,983,984) and certain specific imaging characteristics (Fig. 5-159):

Barkovich_Chap05.indd 492 5/6/2011 9:00:38 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 493

FIG. 5-155. Chiari I malformation with associated syringohydromyelia. A. The cer-


ebellar tonsil (white asterisk) is enlarged, compressed (pointed), and extends more
than 1.5 cm below the bottom of the foramen magnum. Notice that the upper cervi-
cal spinal cord is compressed (white arrow) between the odontoid and the cerebellar
tonsil. B. Sagittal T1-weighted image of cervical spine shows cervical hydromyelia
(white arrows). C. Axial T2-weighted image shows that the central canal of the cord
is dilated, confirming hydromyelia.

FIG. 5-157. Chiari I malformation. Sagittal T2-weighted image shows


FIG. 5-156. Chiari I malformation. Sagittal T1-weighted image shows compressed, low-lying cerebellar tonsils (white arrows). The clivus is nor-
platybasia and shortened clivus. Note the odontoid process pointing poste- mal, but the posterior fossa is small because of a steep tentorium cerebelli,
riorly and distorting the cervicomedullary junction. manifested by the nearly vertical straight sinus (large white arrows).

Barkovich_Chap05.indd 493 5/6/2011 9:00:39 PM


494 Pediatric Neuroimaging

FIG. 5-158. Chiari I malformation with horizontal clivus. Sagittal T1-weighted image (A) shows an abnormally
horizontal clivus (white arrows) and low, pointed cerebellar tonsils (black arrows) with little subarachnoid space
around the tonsils, medulla, and upper cervical spinal cord. Axial T2-weighted image (B) at the foramen magnum
level shows the medulla (m) being compressed (white arrows) by the low-lying tonsils (t).

FIG. 5-159. Tonsillar ectopia secondary to


intracranial hypotension. Sagittal T1-weighted
image (A) shows low-lying cerebellar tissue (white
arrowheads), flattening of the ventral pons (black
arrows) against the clivus, an enlarged pituitary
gland (white arrow), and sagging floor of the
third ventricle. Axial T2-weighted image (B)
shows some narrowing of the lateral ventricles
and excessive fluid (white arrows) in the subdural
space surrounding the cerebral hemispheres.
Coronal T1-weighted image (C) shows smooth,
uniform enhancement (white arrows) of all dural
surfaces.

Barkovich_Chap05.indd 494 5/6/2011 9:00:40 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 495

the brainstem and third ventricle “sag” inferiorly, the pituitary gland is slightly greater tonsillar ectopia than adults or children younger than
upwardly convex, the dura is thickened and shows marked enhance- 5 years old. Asymptomatic tonsillar ectopia of 6 mm should not be
ment after administration of intravenous contrast, and the dural considered pathologic in children between the ages of 5 and 15 years
venous sinuses are enlarged (944,981,982). In very severe, longstanding (988,990). The dens should also be evaluated on the sagittal images,
cases, the midbrain may appear swollen and flattened (944). In these as abnormally increased posterior inclination of the dens is associated
patients, the site of the CSF leak (often a spinal arachnoidal diverticu- with an increased incidence of syringohydromyelia (991).
lum) should be identified and the leak treated (983,984). The CSF leak Although routine MRI studies are often sufficient to make the
can be diagnosed by the finding of extrathecal CSF on spinal imag- diagnosis of Chiari I malformation, CSF flow studies are sometimes
ing (MRI first with myelogram to follow if MRI is equivocal); initial useful in patients in whom either the clinical history or imaging study
treatment of choice is epidural blood patch (983,984). As a result of is atypical or borderline. Flow studies will show decreased CSF flow
another, iatrogenic, cause of intracranial hypotension, Chiari I malfor- at the level of the foramen magnum, decreased CSF flow through the
mations can develop as a consequence of lumbar to peritoneal shunting vallecula and increased motion of the brainstem and cerebellar ton-
of cerebrospinal fluid (985,986); the tonsillar herniation in this situa- sils in patients with symptomatic Chiari I malformations (Fig. 5-160)
tion, which is similar in pathophysiology to spontaneous intracranial (992–994). Of note, patients with normal CSF flow studies are rarely
hypotension, may reverse after removal of the shunt. symptomatic from their Chiari I malformation (995,996) and nearly
In general, tonsillar ectopia should not be considered pathologic all patients with Chiari I malformations and syringohydromyelia (995)
unless the tonsils are compressed (pointed) and the CSF spaces at the or occipital headaches (996) have abnormal CSF flow studies. In addi-
foramen magnum and C-1 are effaced. Mild cerebellar tonsillar ectopia tion to the qualitative findings, quantitative analysis of CSF motion
(<5 mm below a line from the basion to the opisthion on a sagittal shows prolonged CSF systole in the prepontine cistern, reduced sys-
midline image in adults, and with no compression of the cervicomed- tolic velocities and duration of CSF systole in the cervical subarach-
ullary junction or the subarachnoid space at that level) appears to be noid space just below foramen magnum, increased CSF velocity at the
of no clinical significance in most patients (987–989). Indeed, Milhorat foramen magnum, and increased duration of CSF systole in the cervi-
et al. (966) found that the tonsils were at least 5 mm below the fora- cal subarachnoid space at the C-2 to C-3 level (most pronounced in
men magnum in 91% of patients with symptomatic Chiari I malfor- patients with syringes) (994). Some improvement of all of the above
mations. Thus, it appears that when the tonsils extend more than 5 mm flow characteristics is seen after successful foramen magnum decom-
below the foramen magnum and the retrocerebellar cisterns and those pression (993–996).
surrounding the craniocervical junction are obliterated on MRI, clini- In all patients with Chiari I malformations, the spine should be
cal symptoms are more likely to develop (966). The extent of asymp- imaged to look for concurrent syringohydromyelia (Fig. 5-160E) which
tomatic tonsillar descent below the foramen magnum appears to be has an estimated incidence of 20% to 65% (966,989,997,998). Syringo-
age dependent. Children between the ages of 5 and 15 years old have hydromyelia is discussed in Chapter 9.

FIG. 5-160. Normal and abnormal CSF flow studies at the foramen magnum. A–D. Normal CSF flow. Images during CSF systole show
abundant downward flow (high signal intensity) through the anterior and posterior foramen magnum and the foramen of Magendie.
Note that there is little apparent brainstem or cerebellar motion. E. Sagittal T2-weighted image shows horizontal clivus (white arrows),
vertical straight sinus (white arrowheads), severe tonsillar herniation, and cervicothoracic syringohydromyelia (black arrows). F–I.
Reduced CSF flow and increased tonsillar motion in a patient with a Chiari I malformation. During CSF systole (F and G), the column of
CSF flow is compressed (black arrowheads) at the level of the posteriorly pointing odontoid. Increased downward motion (hyperintensity,
black arrowheads) of the brainstem and cerebellar tonsils is present. As CSF diastole begins (H), hypointensity of anterior CSF flow is
noted but is again reduced at the odontoid. Low signal intensity develops in the brainstem and cerebellum (black arrows in I), indicating
increased upward motion of those structures.

Barkovich_Chap05.indd 495 5/6/2011 9:00:42 PM


496 Pediatric Neuroimaging

FIG. 5-160. (Continued)

Barkovich_Chap05.indd 496 5/6/2011 9:00:42 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 497

Chiari II Malformations etiology (1005), emphasizing the importance of gene-gene and gene-
environment interactions in the origins of these defects. Defects of the
The Chiari II malformation is part of a complex malformation cytoskeleton, cell cycle and molecular regulation of cell viability are
involving the hindbrain, spine, and mesoderm of the skull base and prominent among the mice with neural tube defects (1005). In addi-
spinal column (999). Virtually all patients with Chiari II malformations tion, many transcriptional regulators and proteins that affect chroma-
have myelomeningoceles (some define the Chiari II malformation as tin structure are necessary for neural tube closure (1005), although
the hindbrain deformity seen in patients with myelomeningoceles and, the downstream molecular pathways regulated by these proteins are
indeed, the initial descriptions were of patients with myelomeningo- unknown. Some key signaling pathways involved in the formation of
celes [955,1000]). Often, the myelomeningocele is discovered in utero myelomeningoceles have been identified, including over-activation of
as a result of prenatal sonography or elevation of alpha-fetoprotein on SHH signaling and loss of function in the planar cell polarity path-
amniocentesis. After postnatal closure of the myelomeningocele (usu- way, retinoid signaling, and inositol signaling (1005). These processes
ally within the first 48 hours of life), more than 80% of affected patients involved in neural tube closure are critical to the development of focal
develop hydrocephalus (1001); it is in the evaluation of the hydroceph- expansions of the central canal of the developing neural tube that will
alus that the radiologist is most often exposed to these patients in the eventually form the cerebral vesicles and ventricles (1006). If the pos-
postnatal period (although imaging is also performed prenatally by terior neuropore fails to close, the cerebral ventricles will fail to expand
fetal sonography and fetal MRI). Less commonly, the infant develops sufficiently for a normal sized posterior fossa to form and for the
swallowing difficulties, stridor, apneic spells, weak cry, or arm weak- thalami to separate normally; as a result, a pressure gradient develops
ness and is imaged to look for compression of the brainstem at the between the intracranial (higher pressure) and intraspinal (lower pres-
foramen magnum or C-1 level (1002). Patients with brainstem dys- sure) spaces. The pressure gradient causes posterior fossa structures
function have a significantly greater mortality than those without, even to stretch and herniate downward. This theory, proposed by McLone
if decompressive surgery is performed (1003). Epilepsy occurs in 17% and Knepper (1006), is supported by results of fetal myelomeningo-
of patients with myelomeningoceles, almost always a result of CNS cele repair, which show improvement of the posterior fossa malfor-
pathology beyond the manifestations of the Chiari II malformation: mation, in addition to improvement of hydrocephalus (Fig. 5-161)
encephalomalacia/stroke, PVNH, or calcifications (1004). (1007,1008).
Neural tube closure is a complex process, requiring a number of This same theory (1006) also explains the characteristics of the
cell biological functions. Mutations of more than 200 genes have been hindbrain findings, which are best conceptualized as resulting from a
found to cause neural tube defects in mice; however, the pattern of normal sized cerebellum developing in an abnormally small posterior
occurrence in humans suggests a multifactorial polygenic or oligogenic fossa with a low tentorial attachment in the setting of a rostral-to-caudal

FIG. 5-161. Chiari II malformation before (at 22 post conceptual weeks) fetal repair of myelomeningocele and after
birth. A and B. Fetal MR images. Sagittal image (A) shows ventriculomegaly associated with hypogenesis of the corpus
callosum (small white arrows point to hypogenetic corpus), small crowded posterior fossa with cerebellum (small black
arrows) herniating through foramen magnum, and lumbosacral spina bifida (large black arrows). Axial image (B) shows
marked ventriculomegaly.

Barkovich_Chap05.indd 497 5/6/2011 9:00:44 PM


498 Pediatric Neuroimaging

FIG. 5-161. (Continued) C–E. Postnatal images. Sagittal


T1-weighted image (C) shows the hypogenetic callosum (small
white arrows). The cerebellum has returned to almost normal
after in utero closure of the myelomeningocele. The quadrigemi-
nal plate (large white arrows) is slightly beaked inferiorly and
posteriorly. Axial T2-weighted image (D) shows that the ventricle
size has returned to nearly normal after the myelomeningocele
repair. Axial T1-weighted image shows an incidental nodule of
heterotopic gray matter (arrow) in the wall of the left ventricular
trigone. Our experience indicates that about 15% of patients with
myelomeningoceles have periventricular nodular heterotopia.

pressure gradient. The result is that the cerebellum is “squeezed” out 70% of patients with the Chiari II malformation (979,999). The cer-
of the posterior fossa as it grows. The cerebellum is indented superi- ebellum sometimes extends anterolaterally into the cerebellopon-
orly by the tentorium and inferiorly by the foramen magnum or, more tine and cerebellomedullary angles, wrapping around the brainstem
commonly, the posterior arch of C-1. The pons and fourth ventricle (Fig. 5-163). The fourth ventricle is low in position (typically below
are stretched inferiorly and narrowed in their anterior-posterior diam- the pons), vertically oriented, and narrowed in its anterior-posterior
eters (Figs. 5-162 and 5-163). The medulla is also stretched inferiorly diameter (Figs. 5-162 and 5-163). The cerebellar vermis is usually par-
and extends below the foramen magnum. The cervical spinal cord is tially herniated into the cervical spinal canal (Figs. 5-162 and 5-163).
stretched inferiorly as well. The dentate ligaments, which attach to the The herniated cerebellum often degenerates; when this degeneration
lateral aspects of the spinal cord and hold it in place, allow a variable is severe, virtually no cerebellum may be present (Fig. 5-164) (999).
amount of caudal displacement of the cervical spinal cord; however, Occasionally, the fourth ventricle can herniate posteriorly and infe-
the ligaments eventually restrict caudal displacement of the cord. If riorly, behind the medulla and beneath the vermis; this condition is
the medulla is “sucked” down further than the dentate ligaments will known as “an encysted fourth ventricle” (999). The fourth ventricle
allow the spinal cord to move, a characteristic cervicomedullary kink may also be isolated or “trapped” (see Chapter 8) as a result of a com-
is formed (Figs. 5-162 and 5-163). This kink is seen in approximately bination of aqueductal narrowing (or scarring) and diminished CSF

Barkovich_Chap05.indd 498 5/6/2011 9:00:45 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 499

FIG. 5-162. Severe Chiari II hindbrain malformation. Sagittal T1-weighted image (A) shows a severe hindbrain anomaly. The quadrigeminal plate (small
white arrows) is stretched posteriorly and inferiorly, the cerebellum is small and stretched inferiorly. The fourth ventricle (small white arrowhead) is at
the craniocervical junction. A tongue of cerebellar tissue (black arrows) extends inferiorly into the cervical subarachnoid space. The cervicomedullary
kink (large white arrowhead) lies just ventral and inferior to the cerebellar tissue. Note the abnormal posterior extension of the third ventricle (large white
arrow). Axial T2-weighted images (B and C) show periventricular nodular heterotopia (black arrows) and a large massa intermedia (white arrow), as well
as ventriculomegaly. Note that the temporal cortex seems to have too many gyri that are too small; this is stenogyria due to the shunted hydrocephalus.

flow through the fourth ventricular outflow foramina or the basilar The mesencephalic tectum (quadrigeminal plate) is often distorted
cisterns (Fig. 5-165). in patients with the Chiari II malformation, probably secondary to
The fourth ventricle may be very small in patients with Chiari II compression by the temporal lobes combined with the rostral-caudal
malformations (Fig. 5-162); therefore, the isolated fourth ventricle may pressure gradient; the result is that the tectum is stretched posteri-
not look enlarged on casual observation. Consequently, a “normal- orly and inferiorly (“beaked” tectum, Figs. 5-162 and 5-164) (1009).
sized” fourth ventricle in a patient with a Chiari II malformation should Another finding that may be present is a posterior concavity of the
initiate a search for hydrocephalus. When affected patients have hydro- petrous bones and, less commonly, clivus (Fig. 5-165), probably result-
cephalus or an isolated fourth ventricle, the spine should be examined ing from pressure effects of the cerebellum and brainstem in the small
because of a high incidence of associated syringohydromyelia (Fig. posterior fossa (1010). This “scalloping” of the petrous bones may be
5-165). Conversely, when worsening syringohydromyelia develops in difficult to appreciate on MRI.
patients who have had myelomeningocele repairs (almost all of whom Supratentorial anomalies are commonly associated with Chiari
will have a Chiari II malformation), the head should be studied to look II malformations. Abnormalities of the corpus callosum are seen in
for worsening hydrocephalus or a trapped fourth ventricle. 70% to 90% of affected patients, usually consisting of hypoplasia or

FIG. 5-163. Typical Chiari II malformation. Sagittal T1-weighted image (A) shows mild enlargement of the inferior colliculi (white arrow), narrowing
and inferior displacement of the fourth ventricle (white arrowhead), and the cerebellar tongue and cervicomedullary kink in the upper cervical spine.
The corpus callosum is thin and lacks an inferior genu. Axial T2-weighted images (B and C) show cerebellar tissue (white arrows) extending anteriorly
around the brainstem due to the small posterior fossa.

Barkovich_Chap05.indd 499 5/6/2011 9:00:46 PM


500 Pediatric Neuroimaging

FIG. 5-164. Chiari II malformation with near absence of the cerebellum. (A) Sagittal T1-weighted MR images show callosal hypo-
genesis, a very small posterior fossa, elongation of the tectum (small white arrows) and only a small remnant of the cerebellum (in the
cervical subarachnoid space, large white arrow). In (B), the occipital lobe (white arrowhead) can be seen extending down to the foramen
magnum.

hypogenesis (absence of the splenium and absence of the rostrum) dysgenesis. Prominence of the occipital horns and of the posterior
(Figs. 5-161 and 5-163) (57); callosal hypogenesis may, however, be third ventricle is common and can remain even after shunting (Fig.
more severe (Fig. 5-165). PVNH are present in 10% to 15% of affected 5-162) (1011–1013), possibly related to the abnormal corpus callosum
children (Fig. 5-162). Enlargement of the caudate heads and massa and resulting from a deficiency of white matter tracts in the posterior
intermedia is common (1011). Another common anomaly, possibly aspect of the cerebrum. Also after shunting, the medial walls of the
due to chronic hydrocephalus, is fenestration of the falx cerebri, with ventricular trigones often appear dysplastic and a large CSF-contain-
interdigitation of gyri across the interhemispheric fissure at the sites ing structure may appear between the atria and occipital horns of the
of fenestration; this is a nonspecific finding seen in all patients with lateral ventricles. The gyral pattern is often abnormal in the temporal
chronic hydrocephalus and in many with congenital mesenchymal and occipital lobes after shunting, having the appearance of multiple
small gyri (Figs. 5-162 and 5-165). This pattern is not PMG, because
the cortex is of normal thickness but may be secondary to decompres-
sion of the expanded (by hydrocephalus) cortex, abnormal interaction
of the developing cerebral cortex with the overlying leptomeninges, or,
much less likely, to cortical dysgenesis of the hemisphere medial to the
atria and occipital horns. This appearance has been called stenogyria
(717,1014).
Analysis with DTI and fiber tracking shows abnormalities of the
fornices and cingula, which were reported as discontinuous or dys-
plastic in 69% to 77% of patients (1015). (These abnormalities may
be associated with memory and learning deficits, although this result
needs to be confirmed.) The cause of these white matter anomalies
may be genetic or mechanical (secondary to hydrocephalus) (1015).
When very young patients with the Chiari II malformation are
imaged on CT, irregularity of the surfaces of the inner and outer table
of the skull is frequently seen. This lacunar skull or Luckenschädel
appearance is explained by McLone and Knepper as being the result of
incomplete expansion of the calvarium in utero, with resultant disorga-
nization of the collagenous outer meninges from which the membra-
nous calvarium forms (1006). It is of no diagnostic significance in the
era of CT and MRI, and it seems to normalize by age 6 months.
The multiple findings described above are present in a variable
FIG. 5-165. Chiari II malformation with isolated fourth ventricle. Sagit-
tal T1-weighted image shows a large fourth ventricle (white arrows) with a proportion of patients (Table 5-8). It is unusual to see all of the cere-
very narrow aqueduct and decompressed lateral ventricles. Note multiple bral and cerebellar manifestations of the Chiari II malformation in a
small gyri in the occipital lobe, compatible with stenogyria. The corpus cal- single patient. The hindbrain deformity can be extremely severe, with
losum is hypogenetic. The clivus is very thin, presumably due to pressure near-total absence of the cerebellum or it can be relatively mild with
erosion. Note the cervical syringohydromyelia (white arrowheads). only cerebellar tonsillar ectopia. The patients with mild hindbrain

Barkovich_Chap05.indd 500 5/6/2011 9:00:48 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 501

ANOMALIES OF THE MESENCHYME


TABLE 5-8 Frequency of Brain and (MENINGES AND SKULL)
Spine Anomalies in Patients
Cephaloceles and Other Calvarial and Skull Base
with Chiari II Malformations
Defects
Anomaly Frequency Definition, Causes, and Classification
Myelomeningocele Always Definition and Causes

Hydrocephalus Almost always The term cephalocele refers to a defect in the skull and dura with
extracranial extension of intracranial structures. Cephaloceles are
Dysplastic tentorium Almost always divided into four types (1018,1019). Meningoencephaloceles are her-
Small posterior fossa Almost always niations of CSF, brain tissue, and meninges through the skull defect.
Meningocele refers to herniation of the meninges and CSF only. Atretic
Luckenschädel Almost always
cephaloceles are forme fruste of cephaloceles consisting of dura, fibrous
Caudal displacement of brainstem Usually tissue, and degenerated brain tissue; they are most common in the pari-
Cervicomedullary kink Usually etooccipital area. Glioceles consist of a glial-lined cyst containing CSF.
In all of these conditions, the skull defect and herniation most com-
Upward cerebellar herniation Usually
monly occur in the midline. Occasionally, cephaloceles may develop in
Large massa intermedia Usually adults, due to gradual thinning of bone, most commonly in the walls of
Elongated cranial nerves Usually the sphenoid sinus or mastoid air cells. Ultimately, dura, leptomeninges,
and sometimes brain may herniate through the thinned bone causing
Tectal beak Usually an acquired cephalocele. These can become symptomatic (usually hear-
Callosal hypogenesis Usually ing loss or recurrent otitis and otorrhea) and require repair (1020).
The cause of congenital cephaloceles has not been fully deter-
Syringohydromyelia ∼50%
mined. Some argue that cephaloceles result from a failure of neuru-
Malformations of cortical development Occasionally lation (failure of primary neural tube closure) (1021–1024). These
Aqueductal stenosis Occasionally authors note that the most common sites of cephalocele and myelocele
formation correlate well with either failure of closure of the sites of
primary neural tube closure (1021), failure of the closure to propa-
gate from the site of primary closure (1022), or abnormal location of
deformities are often classified as having the Chiari I malformation; closure (1023). Others argue that the disorders of neurulation should
however, the presence of supratentorial anomalies and myelomenin- result in cerebral tissue with disorganized neuroepithelial patterning,
gocele suggests that these patients belong in the Chiari II classification. such as exencephaly, rather than the typically well-developed neural
It should be noted that the term “Arnold-Chiari malformation” is only structures seen in cephaloceles (4,1025). These authors believe that
properly applied to the Chiari II malformation. cephaloceles are the result of a postneurulation event in which brain
tissue herniates through a defect in the mesenchyme that will become
cranium and dura (1025,1026). In support of this theory, Gluckman
Chiari III Malformations et al. (1025) showed that in a chick model there is a critical time point
The Chiari III malformation was originally defined as a condition (at the end of the embryonic period) during postneurulation rapid
characterized by herniation of posterior fossa contents (the cerebel- brain growth when encephaloceles are prone to occur; neural tissue
lum and, sometimes, brainstem) through a posterior spina bifida at the herniates through a mesenchymal defect resulting in decompression
C-1 to C-2 level (955). A variable amount of cerebellar tissue remains of the primitive ventricle. This theory best explains the distorted,
in the posterior fossa. Rather than a variant of Chiari malformation, stretched appearance seen in the brain adjacent to the calvarial defect.
in the authors’ opinion, this condition should be considered a high However, it is most likely that cephaloceles can be the end result of
cervical myelocystocele (see Chapter 9 for a discussion of myelocys- several developmental disorders. Those extending through the skull
toceles). Using this definition, Chiari III is an extremely rare condi- base, which is formed of endochondral bone, may be caused by either
tion; the author has seen only three such cases. More recently, many failure of induction of the bone resulting from faulty closure of the
neurosurgeons have begun to classify all low occipital encephaloceles neural tube (1027,1028) or failure of the basilar ossification centers to
or those with an occipito-cervical encephalocele with a small poste- unite (1029). The calvarium and cranial dura are derived from mesen-
rior fossa and a caudally displaced brainstem (1016) or herniation of chyme that migrates from the neural crests and the paraxial mesoderm
posterior fossa contents through a low occipital and/or upper cervical (1030,1031). Cephaloceles through the calvarium may be secondary to
osseous defect (Fig. 5-166) (1017). Patients diagnosed by these more defective induction of the bone, focal dysgenesis of the dura, pressure
recent criteria are more common; therefore, the diagnosis of Chiari III erosion of the bone by an intracranial mass or cyst (1027,1032,1033),
malformation has become more common in the past 10 years. or failure of closure of the neural tube (1021,1022).
Affected patients usually present with difficulty in breathing and
swallowing as well as with long tract signs (that may be secondary to Classification of Cephaloceles
the disorganization of brainstem nuclei), seizures and developmental Cephaloceles are named for the location of the bone defects through
delay (1017). Definitive diagnosis is established by MRI, which shows which they course. The categories of cephaloceles include the (a)
the cerebellum, brainstem, or cervical spinal cord herniating through occipito-cervical (involving the occipital bone, foramen magnum, and
a high cervical spina bifida (Fig. 5-166). Treatment is surgical repair; posterior arches of upper cervical vertebrae, and often called Chiari
outcome is guarded (1017). III malformations as discussed in the previous section), (b) occipital,

Barkovich_Chap05.indd 501 5/6/2011 9:00:49 PM


502 Pediatric Neuroimaging

FIG. 5-166. Chiari III malformation. Sagittal T1-weighted image (A) and axial T2-weighted image (B) show
cerebellar and brainstem tissue (arrows) herniating through dorsal osseous defects in the lower occipital bone
and the upper cervical vertebrae.

(c) parietal, (d) frontal, (e) temporal (along the superior surface of the to answer these questions (Figs. 5-167 to 5-169). CT better depicts the
petrous ridge), (f) frontoethmoidal (between nasal bones and ethmoid bony changes (Fig. 5-169A), but routine MRI studies (Figs. 5-167 and
bone), (g) sphenomaxillary (through orbital fissures into pterygo- 5-168) will show the amount of brain tissue versus fluid in the cepha-
palatine fossa), (h) spheno-orbital (through defect in sphenoid bone locele in addition to demonstrating associated cerebellar cortical dys-
or optic canal/orbital fissure into orbit), (i) nasopharyngeal (through plasia, venous sinus anomalies, dorsal interhemispheric cysts, callosal
ethmoid, sphenoid, or basiocciput into nasal cavity or pharynx), and anomalies, gray matter heterotopia, tonsillar and brainstem ectopia, or
(j) lateral (along coronal or lambdoid sutures) (1034). A complete dis- Dandy-Walker malformations, all of which have higher frequencies in
cussion of cephaloceles in all of these locations is beyond the scope of patients with cephaloceles (1018,1027,1033,1039). The integrity and
this book. In this section, cephaloceles in the four most common and, the course of the dural venous sinuses are best demonstrated by the use
therefore, most clinically relevant locations will be discussed. Nasal of MR venography (1038,1040) acquired at the time of the MRI study
dermoids and nasal gliomas are included in the section on frontoeth- (Figs. 5-167 and 5-170). Noncontrast 2D time of flight (TOF) acquisi-
moidal cephaloceles, as the clinical presentation can be very similar; tion in the coronal plane is optimal for the venogram (1040).
moreover, dermoids and cephaloceles may have a common etiology
(1029,1035). Finally, this section includes a discussion of calvarial der- Frontoethmoidal Cephaloceles, Nasal Dermoids, and Nasal
moids and other masses that present as calvarial masses in childhood, Gliomas
as well as congenital calvarial defects, both of which can mimic cepha- Embryology. Frontoethmoidal cephaloceles, nasal gliomas, and nasal
loceles. dermoids are included in the same section because all three can pres-
ent as congenital midline nasal masses (229). Moreover, all three are
Occipital Cephaloceles The occipital bone is the most common believed to have a similar embryological derivation, resulting from a
location for cephaloceles in the white populations of Europe and North lack of normal regression of a projection of dura that extends through
America, accounting for approximately 80% of cephaloceles in that the embryologic foramen cecum, between the developing nasal cartilage
group. With Western populations becoming more ethnically mixed, and nasal bone (Fig. 5-171) (229,1029). If the dural projection remains
however, frontal cephaloceles are being seen much more frequently adherent to the skin and pulls it inward, a small dimple will develop on
and are now, overall, as common as occipital ones (1036). As with all the surface of the nose. The dimple is the orifice of a dermal sinus tract
cephaloceles, neurodevelopmental outcome is related to the size of that can extend superiorly along the path of the dural projection for a
the cephalocele, the amount of tissue present in the sac, and the pres- variable distance, sometimes all the way into the cranial vault through
ence of associated anomalies (up to 50%) (1036,1037). Supratentorial the foramen cecum (Fig. 5-172). Dermal or epidermal cysts can develop
and infratentorial structures are involved with equal frequency. Not anywhere along the dermal sinus tract. Cephaloceles presumably
uncommonly, supratentorial structures, infratentorial structures, and result from herniation of intracranial tissues into the dural projection
the tentorium are all included within the cephalocele. The tentorium through the foramen cecum (Fig. 5-173) (1029). Nasal gliomas (nasal
cerebelli may be dysplastic (1038). The diagnosis is usually obvious cerebral heterotopia) are accumulations of dysplastic brain tissue in the
clinically, and imaging studies are ordered to answer two major ques- nasal cavity or subcutaneous tissue that are separate from intracranial
tions: (a) are other severe brain anomalies present? and (b) do the dural contents (Fig. 5-174). They are postulated to result from herniation of
venous sinuses (superior sagittal sinus, straight sinus, and transverse brain tissue into the dural projection with subsequent regression of the
sinuses) course within the cephalocele? MRI is the modality of choice more superior portion of the projection (229,1029).

Barkovich_Chap05.indd 502 5/6/2011 9:00:49 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 503

FIG. 5-167. Occipital cephalocele with gray matter heterotopia. A. Sagittal T1-weighted image shows a small pos-
terior fossa with brainstem and cerebellum stretched posteriorly toward an occipital bone defect. A mostly cystic
mass (white arrows) extends through the defect. B. Axial T2-weighted image shows the vermis stretched posteriorly
through the calvarial defect. A small amount of vermis extends through the calvarial defect. C. Axial T1-weighted
image shows multiple subependymal nodules (white arrowheads) of gray matter in the walls of the lateral ventricles.
D. Sagittal view of a 2D TOF venogram shows no venous anomalies.

Clinical and Imaging Aspects. The frontoethmoidal region is anteriorly by the frontal process of the maxilla, and posteriorly by the
the most common location of encephaloceles in southeastern Asia lacrimal bone and the lamina papyracea of the ethmoid bone (1043).
(1018,1027,1041–1043) and, with the extensive migration of people Many frontoethmoidal cephaloceles involve both the nasoethmoidal
from southeastern Asia to North America, they have become roughly and naso-orbital regions (1041). Clinical presentation of frontoeth-
as common as occipital cephaloceles in North America (1036). They moidal cephaloceles, nasal dermal cysts, and nasal gliomas is similar,
are subdivided by location into three subtypes. Nasoethmoidal cepha- usually nasal stuffiness or a nasal mass (1028,1042). If examination
loceles are the most common of the frontoethmoidal region (1044), reveals a nasal dimple, the diagnosis of dermal sinus is established and
extending through a defect between the nasal bones and the nasal carti- an imaging study is required to search for an associated (epi)dermal
lage. In the nasofrontal group, the defect is located between the frontal cyst and intracranial communication of the sinus, which puts the child
and nasal bones. The defect in naso-orbital cephaloceles is bordered at risk for intracranial infection (Fig. 5-175). If no dimple is seen, an

Barkovich_Chap05.indd 503 5/6/2011 9:00:49 PM


504 Pediatric Neuroimaging

FIG. 5-168. Occipital cephalocele.


A. Sagittal T1-weighted image shows
a large occipital cephalocele. Both
supratentorial and infratentorial
brain tissues are distorted as they
enter the sac along with CSF from
cisterns and the fourth ventricle. B.
Axial T2-weighted image shows the
supratentorial and infratentorial
structures stretching into the cepha-
locele sac.

FIG. 5-169. Small occipital infratentorial cephalocele.


(A) Surface rendering of the calvarium from a CT scan shows a
tiny opening (black arrow) in the midline just above the nuchal
muscular insertion line. On sagittal T2-weighted image (B), the
lesion (white arrow) appears to be filled with CSF but is obscured
by chemical-shift artifacts. The posterior fossa looks otherwise
normal. (C) A T1-image shows the cephalocele (white arrows) as
a CSF-intensity lesion coursing through subcutaneous fat.

Barkovich_Chap05.indd 504 5/6/2011 9:00:51 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 505

FIG. 5-170. Use of MR venography to demonstrate dural sinuses in cephalocele sac. Sagittal T1-weighted image (A) shows the occipital cephalocele. 2D
TOF venogram (B) shows dural venous sinuses (arrows) in the cephalocele sac. This is important information for the surgeon.

FIG. 5-171. Embryology of the frontoethmoidal


region. Nasal dermoids, frontoethmoidal cephaloce-
les, and nasal gliomas all are believed to have similar
embryologic derivations. Frontal cephaloceles presum-
ably result from lack of closure of the fonticulus fronta-
lis (A) with herniation of brain tissue or dura through
the calvarial opening. Normally, a projection of dura
extends through the embryologic foramen cecum (B),
between the developing nasal cartilage and nasal bone.
Normal regression leads to a normal skull base and
frontonasal region, as in (C). Lack of regression leads to
frontonasal pathology. (Reprinted from Barkovich AJ,
Vandermarck P, Edwards MB, Cogen PH. Congenital
nasal masses: CT and MR imaging features in 16 cases.
Am J Neuroradiol 1991;12:105–116, with permission.)

Barkovich_Chap05.indd 505 5/6/2011 9:00:52 PM


506 Pediatric Neuroimaging

FIG. 5-172. Formation of nasal dermal sinuses. If the dural projection (Fig. 5-171B)
remains adherent to the skin, a small dimple will develop on the surface of the nose. The
dimple is the orifice of a dermal sinus tract that can extend superiorly along the path of
the dural projection for any distance, including all the way into the cranial vault through
the foramen cecum. Dermoids or epidermoids may develop anywhere along the tract.
(Reprinted from Barkovich AJ, Vandermarck P, Edwards MB, Cogen PH. Congenital
nasal masses: CT and MR imaging features in 16 cases. Am J Neuroradiol 1991;12:105–
116, with permission.)

FIG. 5-173. Formation of frontoethmoi-


dal cephaloceles. Cephaloceles presumably
result from herniation of intracranial tissues
into the dural projection through the fon-
ticulus frontalis (A) or foramen cecum (B).
(Reprinted from Barkovich AJ, Vandermarck
P, Edwards MB, Cogen PH. Congenital nasal
masses: CT and MR imaging features in 16
cases. Am J Neuroradiol 1991;12:105–116,
with permission.)

FIG. 5-174. Formation of nasal gliomas (nasal cerebral heterotopia). These accumulations
of dysplastic brain tissue in the nasal cavity or subcutaneous tissue are postulated to result
from herniation of brain tissue into the dural projection (Fig. 5-171B) with subsequent
regression of the more superior portion of the projection. (Reprinted from Barkovich AJ,
Vandermarck P, Edwards MB, Cogen PH. Congenital nasal masses: CT and MR imaging
features in 16 cases. Am J Neuroradiol 1991;12:105–116, with permission.)

Barkovich_Chap05.indd 506 5/6/2011 9:00:54 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 507

FIG. 5-175. Nasal dermal sinus with epidural dermoid cyst. Axial CT scan (A) shows an enlarged, unclosed foramen cecum (white
arrow). Midline sagittal reformation of CT scan (B) shows an anterior frontoethmoidalnasal bony canal (black arrows) corresponding
to the dermal sinus tract, with remodeling of surrounding bone. Midline sagittal T2 MR image (C) shows the encysted epidural por-
tion of the dermal tract (t), the naso-ethmoidal tract (white arrows) leading from the dimple (white arrowhead) and adjacent brain
edema and abscess (black arrows). Axial T1contrast-enhanced image (D) shows that the dermal tract, epidural cyst, and intracerebral
abscess all enhance. Surgery demonstrated that the dura was intact, but inflamed, with adjacent infection of the leptomeninges and
brain; pathology demonstrated presuppurative cerebritis.

imaging study is usually ordered to establish the presence or absence of midline of the anterior cranial fossa is still not ossified. Ossification of
a connection of the mass with intracranial contents. the midline cribriform plate and crista galli begins around age 2 months
CT evaluation of the nasofrontal region is often difficult in the neo- and shows a steady increase to age 14 months, after which little change
nate, as the anterior skull base is largely cartilaginous at birth. There- is seen. The percentage of ossified skull base steadily increases over the
fore, it is crucial to understand the evolution of the CT appearance of first 2 years of life. By 24 months, 84% of the anterior skull base is com-
this region in the normal neonate and infant. In the normal neonate, pletely ossified, except for a cartilaginous gap anteriorly in the region of
no calcified bone is seen in the region of the crista galli and cribriform the foramen cecum. The knowledge of this development is important
plate; the absence of ossification should not be misinterpreted as a sign of when evaluating for the presence of cephalocele in an infant (1045). If
a cephalocele. Ossification begins in the lateral aspect of the roof of the a cephalocele is suspected, an MRI should be obtained.
ethmoidal labyrinth and spreads toward the midline. By age 6 months, If thin section (preferably ≤2 mm), contiguous T1- and T2-weighted
50% of the anterior skull base has ossified; however, the anterior images can be obtained, MRI is the modality of choice in the evaluation

Barkovich_Chap05.indd 507 5/6/2011 9:00:56 PM


508 Pediatric Neuroimaging

FIG. 5-176. Nasal dermal sinus with intracranial dermoid. Axial CT after intrathecal injection of iodinated contrast (A) shows a filling
defect (arrows) superior to the foramen cecum. Sagittal reformation (B) shows erosion of the adjacent inner table of the frontal bone
by the dermoid. (Reprinted from Barkovich AJ, Vandermarck P, Edwards MB, Cogen PH. Congenital nasal masses: CT and MR imaging
features in 16 cases. Am J Neuroradiol 1991;12:105–116, with permission.)

of nasal dermal sinuses (229). Although CT better shows the relationship hyperintense compared to brain (Figs. 5-175 and 5-177) (1046). Care
of the dermal sinus to specific bones as it courses through the nasal sep- should be taken not to mistake normal marrow in the crista galli or the
tum, MRI better shows associated intracranial (epi)dermal cysts (Figs. nasal process of the frontal bone (see Fig. 2–19B) for a dermoid tumor.
5-175B, C and 5-177) and does not use ionizing radiation; moreover, FLAIR images are more sensitive than T1-spin echo or FSE/TSE in the
intrathecal contrast is not necessary for MR evaluation. If MRI is not detection of inclusion tumors but do not show the relationship to the
available or is contraindicated, CT should be performed using no more calvarium as well. 3DFT spoiled gradient-echo images (MP-RAGE,
than 1.0 mm slice thickness; subarachnoid infusion of iodinated contrast SPGR, 3DFLASH, T1-FAST, TFE) can be used to obtain thinner slices but
helps to detect intracranial masses (Fig. 5-176). MRI should be obtained may be degraded by susceptibility artifact from interfaces of intracranial
using both T1 and T2 weighting and 1 to 2 mm image thickness in the contents with bone and air. To avoid this artifact, the echo time should
sagittal, coronal, and axial planes. Fast spin-echo/turbo spin-echo T2 be as short as possible. Finally, the administration of a gadolinium-
images are preferable, as artifact from the different magnetic suscepti- compound intravenously may demonstrate the enhancement associ-
bilities of air, bone, and brain are minimized; thin section volumetric ated with infection (Fig. 5-175D). Because the dermal tract is open at
fast spin-echo images are ideal, as there is no interslice gap and the data the skin, infection is a common complication of dermal sinuses/cysts in
can be reformatted in any plane. (Epi)dermal cysts will be isointense to this location like in the spine and posterior fossa.

FIG. 5-177. Nasal dermoids with intracranial extension. A and B. Sagittal T1-weighted image with fat suppression shows a hypointense
mass (black arrow) expanding the foramen cecum and extending into the epidural space. Axial T2-weighted image (B) shows the intracra-
nial hyperintense mass (black arrows) just anterior to the crista galli. C. Sagittal T1-weighted image in a different patient shows a glabellar
mass (large arrows) and a (epi)dermal cyst (small arrows) extending from the glabellar region superiorly through the foramen cecum into
the intracranial cavity. D–F. Sagittal T1- (D) and T2- (E) weighted images from another patient show a glabellar mass (m) that extends
through an enlarged fonticulus frontalis into the extradural space; the foramen cecum (arrow) is normal. Axial T1-weighted image (F)
shows the heterogeneous mass extending posteriorly (white arrows) along the sinus tract.

Barkovich_Chap05.indd 508 5/6/2011 9:00:57 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 509

FIG. 5-177. (Continued)

MRI is superior to CT in the evaluation of cephaloceles and nasal Anomalies and Syndromes,” “Intracranial Lipomas” and Fig. 5-181)
gliomas because it directly shows the extension of brain tissue through (1018,1048,1049). Hypertelorism is common in all cephaloceles in this
the bone defect (Figs. 5-177 to 5-179) in cephaloceles and the lack group and in nasal gliomas because of separation of midline structures
thereof in nasal gliomas (Fig. 5-180) (1047). Imaging in the sagittal by the herniating brain tissue.
plane is optimal for this determination. The signal intensity of nasal
gliomas is similar to that of both epidermoid tumors and brain tis- Parietal Cephaloceles and Atretic Cephaloceles Parietal cepha-
sue (1047); therefore, anatomic factors are more important than signal loceles are uncommon, comprising approximately 10% of cephaloceles
characteristics in making a distinction between these malformations. (1018). They are usually associated with significant underlying brain
In difficult cases, acquisition of CT images through the defect after anomalies and, consequently, have poor prognoses. Among the most
intrathecal contrast administration can be helpful to determine whether commonly associated anomalies are the Dandy-Walker malformation,
the soft tissue mass is continuous with the subarachnoid space; sagittal callosal agenesis with interhemispheric cyst, WWS, and holoprosen-
reformations of ≤1 mm images are optimal. MRI is also superior to cephaly (1032,1034,1039,1050). Because of the proximity of parietal
CT in showing the presence or absence of associated brain anomalies cephaloceles to the superior sagittal sinus, it is particularly important
(Fig. 5-179), the presence of which has profound prognostic implica- to locate the position of the dural venous sinuses with respect to the
tions. Anomalies of the corpus callosum, interhemispheric lipomas, and cephalocele in these patients (Fig. 5-182). Neurosurgical repair is much
malformations of cerebral cortical development frequently accompany more difficult when the sinus is located within the cephalocele.
nasofrontal encephaloceles. Frontoethmoidal encephaloceles are not A high percentage of parietal cephaloceles, and indeed of all cepha-
uncommonly seen in association with craniofacial anomalies, callosal loceles, fall into the category of atretic cephaloceles (1032,1050), small
dysgenesis, or lipomas, often in the form of midline craniofacial dysra- defects in the skin and calvarium that are postulated to be a result
phisms (also known as frontonasal dysplasia, see sections “Associated of a persistence of midline neural crest cells that prevent the mutual

Barkovich_Chap05.indd 509 5/6/2011 9:00:58 PM


510 Pediatric Neuroimaging

FIG. 5-178. Frontoethmoidal cephalocele. Off-midline sagittal T2-weighted image (A) shows a rounded, well demarcated high
signal mass (white arrows) just lateral to the foramen cecum, anterior to the olfactory bulb. Coronal T2-weighted image (B) shows
that the mass is contained within the right nasal cavity and communicates with the intracranial space through a narrow canal (white
arrowheads) in the ethmoid bone.

induction of ectoderm and mesoderm (1051,1052). Atretic parietal cephaloceles have a very high incidence of associated anomalies (100%
cephaloceles usually present as small (5–15 mm), hairless midline in Yokota’s series [1032]), including porencephalies, interhemispheric
masses near the vertex (1032). A sharply marginated calvarial defect cysts, and callosal agenesis (1018). Venous anomalies, particularly fen-
is present below the skin lesion, through which the cephalocele com- estration of the superior sagittal sinus and persistence of an embry-
municates with the intracranial cavity via a strand of connective tissue onic falcine sinus (sometimes associated with absence of the vein of
(Fig. 5-183). The calvarial defect may be too small to see on standard Galen and straight sinus) are seen in most affected patients (Fig. 5-183)
spin-echo MR images; thin section (1–2 mm) spoiled gradient-echo (1053–1055).
(T1-weighted with gradient spoiler or T2 steady-state images such as Atretic parietal-occipital or occipital cephaloceles (Fig. 5-184) usu-
CISS/FIESTA [1053]) or fast spin-echo MR images or thin CT images ally present as nodular, small (<15 mm) masses just above the exter-
may be necessary to establish the communication. Atretic parietal nal occipital protuberance (1032). They enter the calvarium through a

FIG. 5-179. Fronto-orbital cephalocele. A. CT soft tissue remodeling of the face shows a huge left lateronasal and infraorbital skin-covered mass.
B. CT, bone surface rendering of the floor of the anterior cranial fossa shows a wide defect (black arrows) anterior to the ethmoids in the location
where the foramen cecum should have been.

Barkovich_Chap05.indd 510 5/6/2011 9:00:59 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 511

FIG. 5-179. (Continued) C. MR venogram, seen from above, demonstrating displacement of


the facial veins (white arrows); note venous anomalies such as the prominent occipital sinus
(O) on the right. D. Off-midline sagittal reformation of CT scan, showing the wide frontoeth-
moidal canal. E. Midline sagittal T2 image with the brain herniating (white arrows) through an
enlarged foramen cecum. F. Off-midline sagittal T2-weighted image demonstrating the CSF-
filled cephalocele (C) inferior to the orbit; note the diffuse periventricular gray matter hetero-
topia (black arrows).

small calvarial defect and then penetrate the dura immediately below may be apparent on imaging studies, as well (Fig. 5-185D), sometimes
the torcular Herophili, which is typically located higher than usual. associated with the “morning glory syndrome” (1057). Diminished
The tracts typically terminate in the falx cerebri or tentorium cerebelli. visual acuity and hypothalamic-pituitary dysfunction are common
Patients with atretic occipital cephaloceles appear to have a low inci- because the inferior third ventricle, hypothalamus, and optic chiasm
dence of associated anomalies and a good prognosis for normal devel- are stretched as they extend into the sac (Fig. 5-185) (1027,1028).
opment (1032). The diagnosis of nasopharyngeal cephalocele can be made by plain
film or imaging studies. On a submental vertex plain film, a hole of
Nasopharyngeal Cephaloceles Nasopharyngeal cephaloceles are variable diameter with well-defined sclerotic margins is seen in the
also very uncommon. They are important, however, as they are occult sphenoid, ethmoid, or, rarely, basi-occipital bone. Imaging studies
cephaloceles; they are not obvious on clinical examination. In con- demonstrate a large sac of CSF extending through this bone defect
trast to cephaloceles in other locations, which are obvious on physical into the nasopharynx. In large encephaloceles, erosive changes will be
examination and thus diagnosed at birth, nasopharyngeal cephaloce- seen in the hard palate. When the defect is in the ethmoid or sphenoid
les are often discovered toward the end of the first decade of life. The bone, the third ventricle, hypothalamus, pituitary gland, optic nerves,
usual clinical presentation may be persistent nasal stuffiness or exces- and optic chiasm may all be contained within the encephalocele sac
sive “mouth-breathing” by the child secondary to obstruction of the (Fig. 5-185) (1027,1028). Correct characterization of these lesions by
nasopharynx (1027,1028) or spontaneous CSF rhinorrhea (1056). Clin- the radiologist is crucial. If the encephalocele is misdiagnosed as a soft
ical examination will reveal a nasal or pharyngeal mass that increases in tissue mass, the ensuing biopsy may be devastating.
size with Valsalva maneuver. Associated intracranial and ocular anoma-
lies are common. Agenesis of the corpus callosum is seen in approxi- Fetal Imaging of Cephaloceles As in other malformations, the
mately 80% of affected patients (Fig. 5-185). Hypoplasia of the optic diagnosis of cephaloceles in fetuses primarily depends on ultrasound,
discs is seen on ophthalmologic exam; retinal dysplasia and colobomas with MRI being indicated to provide a better depiction of the lesion

Barkovich_Chap05.indd 511 5/6/2011 9:01:00 PM


512 Pediatric Neuroimaging

FIG. 5-180. Nasal glioma. T1 (A) and T2 (B) off-midline


images show a mass (m) in the nose that is of the same sig-
nal as the brain. Axial T1-weighted image (C) with contrast
again shows a similar appearance to the brain. No com-
munication is seen between the mass and the intracranial
cavity (in contrast to Fig. 5-178).

FIG. 5-181. Midline craniofacial dys-


raphism. Axial CT (A) shows lipoma
(open curved arrows) with peripheral
calcification (solid arrows) extend-
ing through a frontal calvarial defect.
Sagittal T1-weighted image (B) shows
the interhemispheric lipoma (arrows)
extending from the posterior frontal
region down through the crista galli
into the nasofrontal area. The corpus
callosum is absent.

Barkovich_Chap05.indd 512 5/6/2011 9:01:01 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 513

FIG. 5-182. Parietal cephalocele. Midsagittal T2-weighted


image (A) shows a cephalocele at the posterior margin of
the parietal lobes, containing mostly CSF with some tissue
(black arrow) at the level of the skull defect. The junction
of the falx cerebri, the tentorium and the falx cerebelli is
malformed, extending too far dorsally and the straight
sinus correspondingly elevated. The superior colliculi and
superior vermis appear “pulled” toward the defect. The
corpus callosum is abnormally thin and short. Sagittal
reformat of a CT venogram (B) demonstrates the presence
of venous structures (white arrows) within the cephalo-
cele. Sagittal view of an MR TOF venogram (C) demon-
strates the abnormal sino-venous pattern but fails to show
the venous content of the cephalocele.

FIG. 5-183. Atretic parietal cephalocele. Sagittal T2-weighted image (A) shows a small subcutaneous mass (white arrows)
in the parietal region. No continuity is seen of the brain tissue with the mass. Sagittal reformation from 2D TOF MR veno-
gram (B) shows abnormality of the dural venous sinuses, with a persistent falcine sinus (white arrows) and absence of the
normal straight sinus.

Barkovich_Chap05.indd 513 5/6/2011 9:01:02 PM


514 Pediatric Neuroimaging

FIG. 5-184. Atretic parietal cephalocele. Midsagittal T2 image (A) shows a midline bony defect; just external to the defect, some
fluid is present in the small cephalocele (white arrow). A CSF channel at the level of the parietooccipital sulci extends from the suprav-
ermian cistern to the defect. The straight sinus is elevated and corresponds in location to a falcine sinus. Anteroposterior view of an
MR venogram (B) shows fenestration of the superior sagittal sinus, which courses on either side of the atretic cephalocele.

and to identify any other CNS malformations. The features that are When the mass is located at the anterior fontanelle, the most common
looked for are the same in fetuses as in neonates (Figs. 5-186 to 5-188); diagnosis is dermoid. Dysembryoplastic dermoid tumors (commonly
MRI is used in our practices because it demonstrates more details and referred to as dermoid “cysts” even though they are usually solid and
may allow diagnosis of a more specific disorder better than ultrasound filled with keratin) comprise about 20% of all scalp lesions (1059).
(Fig. 5-187). Also, as fetal MRI is used for prognostication, it should be Dysembryoplastic dermoids are simple benign teratomatous tumors;
remembered that cephaloceles are not static disorders: they are sub- they differ from the dermal sinuses, tracts and cysts that result from a
mitted to the combined influence of brain growth and CSF forces, and failure of the separation of the skin from the neural tube (in the spine
the neonatal abnormality may be more severe than the abnormality or posterior fossa) or from the dura (in the frontonasal area) and are
assessed at midgestation (Fig. 5-188). better described as inclusion cysts. Patients with calvarial dermoids
typically present in the first few months of life with a soft or firm mid-
Fontanelle Dermoids and Other Calvarial Masses of Child- line solitary mass over the calvarium. The most common location is
hood A number of different types of lesions can present as masses on the anterior fontanelle, although they often occur along other cranial
the head in children and mimic cephaloceles on clinical exam (1058). or facial sutures or within the diploic space of the calvarium; they may

FIG. 5-185. Sphenoidal cephalocele. Axial CT image (A) shows large, sharply marginated hole (arrows) in the sphenoid bone. Sagittal CT refor-
mation (B) shows the large bony defect with CSF (arrows) herniating within it. Sagittal T1-weighted image (C) reveals the encephalocele extending
down through the sphenoid bone into the nasopharynx and impression upon the posterior hard palate (small arrows). The dorsum sella (curved
arrow) remains intact. The optic chiasm (open white arrow) runs through the encephalocele. The corpus callosum is absent. Axial T1-weighted
image (D) shows the cephalocele defect (white arrows) and a small left globe (black arrows) with retinal detachment.

Barkovich_Chap05.indd 514 5/6/2011 9:01:04 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 515

FIG. 5-185. (Continued)

occur practically anywhere. Size and imaging characteristics are variable. (see Chapter 7); rarely melanotic neuroectodermal tumors of infancy
In infants, ultrasound with a high resolution (7–10 MHz) transducer is (see Chapter 7) develop in the cranial vault in infants, typically in the
an excellent first study, showing a sharply defined mass with an echo- region of the anterior fontanel or other sutures (1063).
genic capsule. Over the first year of life, the mass may diminish in size Solitary infantile myofibromatosis of the skull is a benign tumor
and become increasingly echogenic (1060). CT shows a soft tissue mass that is composed of fascicles, whorls, and nodules of spindle-shaped
that is typically hypodense, well defined, round, and causes minimal cells with myofibroblastic features (1064). Radiologically, they appear
scalloping of adjacent bone. MRI shows a well-defined mass that is as solitary lytic lesions of calvarium that show increased uptake on
99m
typically isointense to immature brain but may be isointense to CSF Tc-methylene diphosphonate bone scan. On CT, these sharply defined
(Fig. 5-189) (1061); the same appearance is seen when identified in the masses originate in the calvarium or dura, have soft tissue attenuation,
fetus (Fig. 5-190). Separation from the underlying intracranial space is and show marked enhancement after administration of iodinated con-
easily identified in both the coronal and sagittal planes. trast. On MRI, they exhibit long T1 and T2 relaxation times compared
Martinez-Lage et al. (1058) and Yoon and Park (1062) reported the with gray matter and otherwise show similar features to those seen on
frequency of various pathologic entities that presented as masses on the CT (1065,1066). Prognosis is excellent after excision (1067).
head in childhood and adolescence. They found that the incidence var- Sinus pericranii are dilated extracranial veins that communicate
ies depending upon the age of the patient at the time of presentation with the dural sinuses by way of transcalvarial emissary veins. On
(Table 5-9). They found that MRI was best single radiologic exam to clinical examination, they appear as soft, bulging masses in the scalp
evaluate the patient prior to surgery. Malignant tumors were rare and that are usually less than 1.5 cm in diameter and are most commonly
included mainly rhabdomyosarcomas and metastatic neuroblastoma located close to the midline in the frontal region (1068). Rarely, they

FIG. 5-186. Occipital cephalocele associated with a Dandy-Walker cyst in a 21-week-old fetus. A. Sagittal midline
image shows the huge fourth ventricular cyst (4), a considerably elevated tentorium, expanded squamous occipital
bone and posterior herniation of the cephalocele (C). B. Axial image demonstrates the herniated CSF-filled cephalocele
(c) extending through the cranial defect.

Barkovich_Chap05.indd 515 5/6/2011 9:01:05 PM


516 Pediatric Neuroimaging

FIG. 5-187. Atretic occipital cephalocele in a 23-week-


old fetus. A. The sagittal midline image reveals the small
bone defect (black arrow); it also shows posterior dis-
placement of the upper brainstem. B. Axial image shows
the defect (white arrow) in the tentorial dura and in the
calvarium. C. A higher cut demonstrates a cerebral cleft
with irregular ependymal lining (black arrows), either gray
matter heterotopia or ependymal destruction.

FIG. 5-188. Evolution of an occipital cephalocele. Sagittal image (A) in a 20-week-old fetus shows the huge CSF-filled cephalocele (c) with
a relatively small solid component (black arrow) at the bony defect. Note also the reduced volume of CSF. Sagittal postcontrast T1 image of
the same child during the neonatal period (B) still shows a mostly CSF-filled cephalocele, but the solid component has become larger and
includes cerebellar as well as cerebral tissue. Note the prominent venous channels in the sac.

Barkovich_Chap05.indd 516 5/6/2011 9:01:06 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 517

FIG. 5-189. Dermoid of the anterior fontanelle. Sagittal T1-weighted image


shows a low intensity mass (arrows) overlying the anterior fontanel, sepa-
rated from intracranial contents by intact calvarium.

occur in lateral frontal and temporal locations (1069). Diagnosis is carotid arteries will often show an underlying vascular anomaly, most
made clinically (the lesions are soft and spontaneously reduce when commonly a developmental venous anomaly but, occasionally, an arte-
intracranial pressure is reduced) and patients usually require no treat- riovenous fistula (1071).
ment (1068). Imaging studies will show a soft tissue mass that enhances
after contrast infusion and is accompanied by scalloping of the outer Imaging Appearance of Surgically Repaired Cephaloceles A
table of the skull (1070); intracranial vascular anomalies, in particu- tract of CSF is almost always seen extending into the deep portion of
lar developmental venous anomalies, are frequently present (1071). the brain from the cephalocele sac (Fig. 5-192). This CSF tract per-
The calvarial defect can sometimes be seen on a thin section CT or sists after surgical repair and is very helpful in identifying patients
MR image; sagittal images may show the relationship to the under- who have had prior repairs of encephaloceles. Another helpful finding
lying dural venous sinus (Fig. 5-191A) (1058,1070). The best way to on imaging studies is distortion of the brain resulting from stretch-
confirm the diagnosis is with the use of Doppler sonography, which ing of the brain tissues toward the site of the encephalocele. This dis-
will show flow within the lesion as well as communication with an tortion probably results from in utero extrusion of non-myelinated
underlying venous sinus (Fig. 5-191B). Angiographic analysis is no brain tissue through the calvarial defect. Unmyelinated brain tissue is
longer necessary. If performed, injection of the internal and external extremely soft and is easily deformed. The brain tissue tends to retain

FIG. 5-190. Dermoid of the scalp, 22-week-old fetus. A. Off-midline sagittal image showing a well demarcated, bright rounded
mass (black arrow) in the scalp at the level of the anterior fontanelle. B. Same case, coronal image, shows the mass (black arrow) just
to the side of the fontanelle. Note the similarity to Fig. 5-189.

Barkovich_Chap05.indd 517 5/6/2011 9:01:08 PM


518 Pediatric Neuroimaging

TABLE 5-9 Calvarial Masses in Childhood (by Age)


First year of life Epidermal cysts
Cephalohematomas (see Chapter 4) Malignant tumors
Dermoids and epidermoids (see Chapter 7) Leptomeningeal cysts (see Chapter 4)
Hemangiomas Angiomas
Tumors (see Chapter 7) Atretic cephaloceles
Metastatic neuroblastoma Sinus pericranii
Ewing sarcoma Ages 8–17 y
Leukemic masses Fibrous dysplasia
Infantile myofibromatosis Dermoids and epidermoids
Melanotic neuroectodermal tumor of infancy Osteomas
Sinus pericranii Langerhans cell histiocytosis
Atretic cephaloceles Sinus pericranii
Ages 1–7 y Angiomas
Dermoids and epidermoids Plexiform neurofibromas (see Chapter 6).
Langerhans cell histiocytosis (see Chapter 7)
From (1058) and (1062).

the configuration it held at the time of myelination, so the distorted small emissary veins. However, in a small number of patients (about
tail of tissue pointing toward the calvarial defect remains throughout 1 in 20,000), parietal foramina are enlarged, remaining up to 5 cm in
the patient’s life (979). diameter; these are known as giant parietal foramina or foramina pari-
etalia permagna, and they are often associated with mutations of the
Other Congenital Calvarial Defects Other than those associated homeobox genes MSX2 (located at chromosome 5q34–35, known as
with cephaloceles, congenital defects of the cranial vault are rare. The PFM1) or ALX4 (located at chromosome 11p11–12, known as PFM2)
most common are parietal foramina and defects associated with cutis (1074–1076). Both are inherited in an autosomal dominant manner.
aplasia congenita. They are more common in Asians than Europeans and represent the
most common type of congenital skull defect (1074). Affected children
Parietal Foramina. Parietal foramina are oval, usually paired defects are typically asymptomatic, although bulging of the scalp at the defect is
that are parasagittally located in the mid to posterior parietal bones often noted during crying. A syndrome has been described that includes
(1072). Parietal foramina may appear unilateral in early infancy, but craniofacial dysostosis and mental retardation, attributed to deletion
the bilaterality only becomes apparent with development of the ridge of the short arm of chromosome 11 (1077). Another report associates
of bone along the sagittal suture (Fig. 5-193A) (1073). The overlying craniosynostosis, delayed fontanelle closure, parietal foramina, imper-
skin and hair are normal. Normal bone growth never occurs within forate anus, and skin eruption; it is known as CDAGS (1078).
the defects (1072). Most parietal foramina are smaller than 1 mm in In addition to these syndromes, giant parietal foramina are worthy
diameter by the time the calvarium matures and serve as conduits for of note in this neuroimaging text because they may be mistaken for

FIG. 5-191. Sinus pericranii. Sagittal postcontrast T1-weighted image (A) in a child with a soft mass near the midline over the parietal convexity has an
enhancing lesion (white arrow) that seems to communicate with the superior sagittal sinus through a small calvarial defect. Doppler sonogram through
the lesion (B) shows that the mass communicates with the superior sagittal sinus (SS) through an emissary vein (e).

Barkovich_Chap05.indd 518 5/6/2011 9:01:09 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 519

cephaloceles on fetal imaging and because they are associated with


rather characteristic intracranial anomalies. Fetal sonography and fetal
MRI may show focal bulging of cerebrospinal fluid beyond the margins
of the calvarium in the parasagittal parietal bone (1079). This should
not be mistaken for a cephalocele, as it will resolve over time. Recogni-
tion of the known associated anomalies may aid the correct diagnosis.
The tentorium has a high insertion with a consequent large posterior
fossa. As a result of the tentorial anomaly, one or more medial occipi-
tal gyri herniate inferiorly into the tentorial incisura, giving a charac-
teristic appearance on MR studies (Fig. 5-193B). Finally, the straight
sinus is underdeveloped, resulting often in the presence of persistent
embryonic venous structures such as falcine sinuses (Fig. 5-193B) and
persistent median prosencephalic veins (1075,1076). Coronal or sag-
ittal images may show the parietal cortex appearing to protrude into
the calvarium (Fig. 5-193C). Associated malformations may include
craniosynostosis, vertebral anomalies, digital anomalies, clavicular
hypoplasia, and absence of the acromion process.

FIG. 5-192. Postoperative occipital cephalocele. Sagittal T1-weighted Calvarial Defects associated with Cutis Aplasia. Cutis aplasia con-
image shows stretching of the cerebellum toward a tract (open arrows) of genita is the name given to the absence of a portion of skin at birth, in
CSF that leads to the site of prior cephalocele repair. a localized or widespread area (1080). It most commonly (60%–70%)

FIG. 5-193. Parietal foramina. Surface reformation


of CT scan (A) shows large defects (black arrows) in
the parietal bones of an infant. Sagittal T1-weighted
image (B) shows an anomalous connection (white
arrows) of the straight sinus and the superior sagit-
tal sinus. Venous anomalies are commonly present
and are likely related to the presence of the foramina.
Coronal T2-weighted MR scan (C) shows parietal
lobe and blood vessels (white arrows) herniating into
the calvarial defects.

Barkovich_Chap05.indd 519 5/6/2011 9:01:10 PM


520 Pediatric Neuroimaging

drug or toxin exposure (fetal hydantoin syndrome) (1084). Rarely, no


cause is found (1085).

Intracranial Lipomas
Intracranial lipomas are most commonly found incidentally, when
imaging is performed for reasons unrelated to their discovery, although
they can rarely be associated with epilepsy, particularly when they are
located over the surface of the cerebral cortex (1086,1087). Lipomas are
malformations; they are believed to result from abnormal differentia-
tion of the meninx primitiva, the undifferentiated mesenchyme that
surrounds the developing brain (1088–1090). Normally, the meninx
primitiva differentiates into the leptomeninges and the subarachnoid
space. For some, as yet unknown, reason, some areas of meninx dif-
ferentiate into fat in some patients, forming intracranial lipomas. As
a result of their formation from the meninx, intracranial lipomas are
almost always located in the subarachnoid space. As they are mal-
formations, not neoplasms, lipoma cells do not multiply (they will
hypertrophy, like other normal fat cells, when patients gain weight or
receive steroid therapy) and almost never exert mass effect on adja-
cent structures. Moreover, because lipomas are, essentially, maldiffer-
entiated subarachnoid space, blood vessels and cranial nerves often
course through them. Therefore, surgical treatment is rarely indicated
and has high morbidity. The most common locations for intracranial
FIG. 5-194. Calvarial defect associated with cutis aplasia congenita. Axial lipomas are the deep interhemispheric fissure (dorsal to the corpus
postcontrast CT image shows parietal calvarial defect (arrows) with abnor-
callosum, 40%–50%), the quadrigeminal plate/supravermian cisterns
mal thinning of the overlying skin.
(20%–30%), the suprasellar/interpeduncular cisterns (10%–20%),
the cerebellopontine angle cisterns (∼10%), and the Sylvian cisterns
presents as a solitary defect on the scalp, but multiple lesions sometimes (∼5%) (1087,1090). Occasionally, lipomas are found on the surface of
occur. The majority of scalp lesions develop on the vertex just lateral the cerebrum (1086). In such situations, blood vessels course through
to the midline. Most are sporadic, but some familial cases have been the lipoma and the underlying cortex is dysgenetic with poorly defined
reported and some teratogens have been postulated to be responsible cortical lamination, fusion of the molecular layer, disrupted basal lam-
(1080). The lesions are noninflammatory and well-demarcated; they ina, prominent astrocytosis, and abnormal synaptic profiles (1086).
may be circular, oval, linear, or stellate, and range in size from 0.5 cm to Interhemispheric lipomas (commonly called “callosal lipomas” or
as large as 10 cm. If they form early in gestation, these lesions may heal “lipomas of the corpus callosum” because they are situated adjacent
before delivery and appear as an atrophic, membranous, or parchment- to the corpus) are almost always associated with hypogenesis or agen-
like scar with associated alopecia, whereas less mature defects present esis of the corpus callosum. These malformations can be large and
at birth as ulcerations, occasionally involving the dura or meninges. lobulated in appearance (these most typically are seen in the anterior
About 20% of cases involving the scalp have associated calvarial defects callosal region) or thin and curvilinear (these are typically seen in
(Fig. 5-194), probably due to the same forces that caused the cutaneous the posterior callosal region, curving around the most posterior part
lesion (1081). The skull lesions do not heal by spontaneous regenera- of the corpus). Encephaloceles and cutaneous lipomas can be asso-
tion (1082). ciated, as well, usually in the frontal region. In fact, several midline
Isolated congenital defects of the calvarium with intact brain, developmental disorders appear to have lipomas of the interhemi-
scalp, and meninges are rare (1082). These defects usually occur in the spheric fissure as a part of the syndrome. Many of these are associated
midline and are of variable size (ranging from 1 to 10 cm [1073]). They with midline facial clefts, basilar cephaloceles, and retinal dysplasia,
probably represent either a fusion defect or a congenital failure of ossi- a condition known as midline craniofacial dysraphism or frontona-
fication of a portion of a calvarial bone. As a result of their location, sal dysplasia (see Fig. 5-181 in section of this chapter on frontona-
these calvarial defects may expose the sagittal sinus as well as the dura sal cephaloceles) (93). When interhemispheric lipomas and callosal
mater and, therefore, such defects increase the possibility of serious anomalies are associated with median cleft lip or palate and cuta-
complications such as meningeal infection and hemorrhage; mortality neous polyps of the face, a diagnosis of Pai syndrome can be made
rates of up to 12% have been reported (1074). Bone regeneration rarely (1091,1092).
occurs without surgical intervention (1073,1082). Perlyn et al. (1074) Skull radiographs of patients with intracranial lipomas may be
have classified these along with other congenital calvarial defects and normal or, in large interhemispheric lipomas, may reveal punctate or
discuss potential therapies. curvilinear midline calcifications with adjacent lucent (fat density)
regions (75). On CT, lipomas are sharply demarcated areas of marked
Persistence of the Anterior Fontanelle. The range of normal closure hypodensity in affected cisterns (Fig. 5-195). Lobulated interhemi-
of the anterior fontanelle is generally regarded to be 4 to 26 months spheric lipomas may extend inferiorly into the choroid plexus of the
(1083); more than 95% are closed by the age of 19 months. Delayed ventricles, anteriorly in front of the callosal genu, posteriorly behind
closure may be associated with increased intracranial pressure, bone the hypoplastic body or splenium, or dorsally into the interhemi-
diseases (vitamin D rickets, hypophosphatasia, osteogenesis imper- spheric fissure. Calcification is often present in lobulated interhemi-
fecta, cleidocranial dysostosis), endocrine disorders (athyrotic hypo- spheric lipomas, most commonly within a fibrous capsule surrounding
thyroidism), chromosomal abnormalities (trisomy 13, 18, or 21), and the lipoma (1093). The calcification may be curvilinear, located at the

Barkovich_Chap05.indd 520 5/6/2011 9:01:11 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 521

FIG. 5-195. Large, lobular interhemispheric lipoma. Axial CT scan


(A) through the lipoma shows a lucent area (large white arrows) in the inter-
hemispheric fissure immediately superior to the corpus callosum. Small
peripheral calcifications (small white arrows) are seen within the wall of the
lipoma. Sagittal T1-weighted image shows a large bulky interhemispheric
lipoma (large white arrows). (B) Note that the corpus callosum (small white
arrows) is hypogenetic with no anterior genu, rostrum, or splenium. Coronal
T1-weighted image (C) shows the lipoma extending into the velum inter-
positum and then through the choroidal fissures into the lateral ventricles.

periphery of the lipoma (Fig. 5-195A) or, less commonly, nodular and lipomas from ectopic posterior pituitary glands (Fig. 5-199, also see
within the center of the lipoma. Calcification is less common in lipo- section on “Anomalies of the Hypothalamic-Pituitary Axis” of this
mas located elsewhere. The full extent of the lipoma and associated chapter). Lipomas in the suprasellar (Fig. 5-199) and quadrigeminal
callosal hypogenesis can only be fully appreciated on MRI. plate/supravermian cisterns (Fig. 5-200) are typically small and may
The MR appearance of a lipoma is that of a hyperintense mass on not show obvious chemical shift artifact. In contrast, lipomas in the
T1-weighted sequences, becoming hypointense on conventional spin- interhemispheric fissure (Fig. 5-195), sylvian fissure (Fig. 5-198), and
echo T2-weighted images as the TE increases (Figs. 5-195 and 5-196) cerebellopontine angle (Fig. 5-201) may be quite large. Chemical-shift
and when fat suppression is applied. On fetal MRI, lipomas are isointense artifact may be difficult to differentiate from signal voids due to rapid
to slightly hypointense compared with surrounding brain (Fig. 5-196). flow in arteries; this is a particularly important issue in sylvian fissure
Smaller lipomas typically have no adjacent brain malformations, but dys- lipomas (Fig. 5-198) and interhemispheric lipomas. Fat suppression is
plastic brain tissue often accompanies large ones; for example, the cer- particularly important to make this distinction and to identify cranial
ebellar vermis is sometimes small and dysmorphic when adjacent to a nerves coursing through the lipomas (Fig. 5-201), as sylvian fissure
large supravermian/collicular lipoma (1087). When lipomas are large, it lipomas are sometimes associated with aneurysms of the middle cere-
is easy to see associated chemical shift artifact (areas of hypointensity and bral vessels (1094).
hyperintensity on opposite sides of the lipoma in the frequency encod- Interhemispheric lipomas may appear large and lobulated or thin
ing direction, seen best on conventional T2-weighted images acquired and curvilinear. Lobulated interhemispheric lipomas tend to be located
with a short bandwidth), resulting from the different chemical shifts more anteriorly (Figs. 5-195 and 5-196), may wrap around the ante-
of water and fat protons (Figs. 5-197 and 5-198). The presence of the rior and posterior ends of the corpus callosum, and sometimes extend
chemical shift artifact should alert the reader that the lesion is fat, and not through the choroidal fissure and into the stroma of the choroid plex-
blood. Application of a fat suppression sequence will make the lipoma uses of the lateral ventricles (Fig. 5-195). Curvilinear lipomas are thin
hypointense to gray matter (Figs. 5-198 and 5-199), confirming the and typically extend posteriorly behind the corpus (Fig. 5-197), some-
diagnosis. Fat suppression is useful to differentiate small hypothalamic times wrapping around and coursing into the velum interpositum. The

Barkovich_Chap05.indd 521 5/6/2011 9:01:11 PM


522 Pediatric Neuroimaging

FIG. 5-196. Fetal and postnatal MRI of a lobulated


interhemispheric lipoma. Fetal sagittal (A) and axial
(B) images show an intermediate intensity mass
(white arrows) in the region of the corpus callosum.
The corpus callosum itself is poorly seen. Postnatal
sagittal T1-weighted image (C) shows the lipoma
(white arrows) immediately dorsal to the hypoge-
netic corpus callosum (white arrowheads).

FIG. 5-197. Small, linear interhemispheric lipoma with chemical-shift artifact. Sagittal T1-weighted image (A) shows a
linear hyperintense mass (large arrows) above a hypogenetic corpus callosum (note the small splenium). Axial T2-weighted
image (B) shows marked chemical-shift artifact manifested as hypointensity (black arrows) posterior to the hyperintense
lipoma. This is a result of mismapping of the fat protons due to their chemical shift being several parts per million different
than that of water protons.

Barkovich_Chap05.indd 522 5/6/2011 9:01:12 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 523

FIG. 5-198. Sylvian fissure lipoma mimicking an aneurysm. Axial T1-weighted image
(A) shows a hyperintense lesion (white arrows) in the sylvian fissure in the region of
the middle cerebral artery trifurcation. Axial T2-weighted image (B) shows heterogene-
ity (white arrow) in the lesion, with curvilinear hyperintensity in the anterior portion
of the lesion, representing mismapped fat protons, and hypointensity in the posterior
portion of the lesion, representing absence of signal due to mismapping of fat protons.
This suggests that the lesion is a lipoma, as the anteroposterior plane was the frequency
encoding plane. Coronal T1-weighted image with fat suppression (C) shows absence of
signal in the lesion (white arrows), confirming that it is a lipoma.

FIG. 5-199. Hypothalamic, interpeduncular cistern lipoma: value of fat suppression. Sagittal T1-weighted image (A) shows the
small lipoma (white arrow). Postcontrast sagittal T1-weighted image with fat suppression (B) shows hypointensity (white arrow)
at the site of the lipoma. Note that the fat in the calvarium and clivus have also become hypointense.

Barkovich_Chap05.indd 523 5/6/2011 9:01:13 PM


524 Pediatric Neuroimaging

FIG. 5-200. Quadrigeminal plate/supravermian lipomas. Sagittal T1-weighted images shows a small (white arrow in A) and a large
(white arrows in B) lipoma located between the inferior colliculus and the superior cerebellar vermis.

adjacent corpus callosum is essentially always hypogenetic, with lipoma inner and outer walls consist of sheets of arachnoid cells that join
extending around the posterior portion of the corpus; no callosal fibers with normal arachnoid at the margins of the cyst (1095–1098). True
are seen dorsal or posterior to the lipoma (Figs. 5-195 to 5-197) (1090). arachnoid cysts differ from cysts caused by trauma or inflammation
When affected patients have associated facial dysmorphism, computed (sometimes called arachnoid loculations, acquired arachnoid cysts, or
tomography is extremely helpful for the evaluation of the bony cranio- secondary arachnoid cysts) in that the latter are merely loculations of
facial anomalies and the planning of surgical reconstruction. cerebrospinal fluid surrounded by arachnoidal scarring (1096,1098).
Ultrastructural studies have shown that the cells lining true arachnoid
Arachnoid Cysts cysts contain specialized membranes and enzymes for secretory activ-
ity (1099). When true arachnoid cysts expand, therefore, the mecha-
Pathologic Characteristics nism appears to be an accumulation of CSF secreted by cells in the cyst
Arachnoid cysts are congenital lesions of the arachnoid membrane wall rather than by osmotically induced filtration or a ball-valve mech-
that expand by CSF secretion. Light and electron microscopic stud- anism. The precise differences between arachnoid cysts that expand
ies have conclusively demonstrated that congenital arachnoid cysts over time and those that remain stable in size have yet to be worked
(also called true arachnoid cysts) are intra-arachnoid in location; their out (1098,1100,1101).

FIG. 5-201. Cerebellopontine angle lipoma enveloping cranial nerve. Axial CT scan (A) shows a hypodense mass (white arrows) in the right cerebel-
lopontine angle. Postcontrast axial T1-weighted image (B) shows the lesion (white arrows) to be hyperintense. Fat-suppressed postcontrast T1-weighted
image (C) shows suppression of the high signal intensity (white arrow), confirming that it is a lipoma. Note the cranial nerve (white arrowhead) coursing
through the lipoma.

Barkovich_Chap05.indd 524 5/6/2011 9:01:15 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 525

Arachnoid cysts usually arise within and expand the margins of symptoms, the physical and neurological signs and symptoms reflect
CSF cisterns that are rich in arachnoid. The sylvian fissures are the most their anatomical distribution and, in particular, their effect on CSF
common location for arachnoid cysts, accounting for up to 50% of flow. Small cysts, up to several centimeters in size, are asymptomatic
these lesions. The cerebellomedullary/cerebellopontine angle cisterns and their discovery is almost always incidental. When middle cra-
are the next most common location (∼15%), followed by the cerebral nial fossa cysts are large, they are often discovered as a result of cra-
convexity (∼13%), suprasellar region (∼11%), retrocerebellar cisterns nial asymmetry (Fig. 5-202); occasionally middle cranial fossa cysts
(∼10%), interhemispheric fissure (∼5%), quadrigeminal plate cistern may be associated with seizures, headaches, or, rarely, hemiparesis
(∼5%), and “other” locations (∼12%) (1101,1102). (1096), but the actual relationship of these symptoms to the cyst is
questionable (1101). The incidence of hydrocephalus associated with
Clinical Characteristics arachnoid cysts is low, and current estimates of less than 10% (1104)
Arachnoid cysts usually occur sporadically. They have a higher inci- are much higher than the authors’ experience would suggest. When
dence in individuals with autosomal dominant polycystic kidney dis- hydrocephalus develops, it is usually associated with large supraten-
ease, being reported in as many as 8.1% of patients with this disorder torial cysts, suprasellar cysts, posterior fossa cysts, or quadrigeminal
(1103), and in Aicardi syndrome (87). Most cysts, if not associated cistern cysts. Resultant symptoms include intracranial hypertension,
with other anomalies, remain clinically silent and are only detected macrocephaly, or visual loss (see Chapter 8). Suprasellar cysts may also
incidentally (1104). In the rare cases in which arachnoid cysts produce cause hypothalamic-pituitary dysfunction and the bobble-head doll

FIG. 5-202. Large middle cranial fossa arachnoid cyst in a neurologically asymptomatic child with plagiocephaly.
Coronal T1-weighted image (A) and axial T2-weighted image (B) show the cyst (C) to be isointense with CSF. The
left hemicalvarium is expanded (white arrows), the left cerebral hemisphere is compressed, and the midline (inter-
hemispheric fissure, septum pellucidum) is shifted from left to right. Axial FLAIR (C) and average diffusivity (D)
images confirm that the mass is composed of CSF.

Barkovich_Chap05.indd 525 5/6/2011 9:01:16 PM


526 Pediatric Neuroimaging

syndrome (1096,1097,1105). When hydrocephalus is present, it may is straightforward using diffusion imaging, FLAIR, or magnetization
be communicating or non-communicating and may result from either transfer imaging. If these techniques are not available, cisternography,
defective CSF resorption or mechanical obstruction to egress of CSF using intrathecal water-soluble contrast (Iohexol 180 mg of iodine
from the ventricles (1106). After treatment of the cyst, cognitive and per mL, 3 mL injected intrathecally via lumbar puncture) may be neces-
neurological outcome are excellent (1107). sary to differentiate epidermoids from arachnoid cysts. After intrathe-
cal injection of contrast, the patient is scanned after being placed prone
Imaging Characteristics in the head-down position (30°) for 2 minutes. A sharp border between
Arachnoid cysts appear on CT and MRI as sharply marginated, homo- the lesion and surrounding CSF is seen with arachnoid cysts whereas
geneous, unilocular masses that have signal identical to CSF on both irregular interdigitation into the interstices of the mass is noted with
CT and MRI. The use of FLAIR and diffusion sequences (Fig. 5-202) epidermoids. Cystic astrocytomas are more easily differentiated. Intra-
can help to confirm the diagnosis of cyst. On transfontanelle sonog- venous administration of paramagnetic contrast will show an enhanc-
raphy, they are sharply marginated, sonolucent masses with a well- ing solid tumor component in cystic astrocytomas; arachnoid cysts do
defined posterior border and enhanced through transmission. Fetal not enhance and do not have mural nodules.
MRI shows the cyst as a CSF-intensity mass that displaces adjacent Cysts of the middle cranial fossa differ somewhat in appearance,
parenchyma with a very sharp margin between the parenchyma and depending upon size. Approximately 20% are small, biconvex or semi-
the mass (Fig. 5-203). circular lesions with little mass effect and no expansion of bone (1097).
Suprasellar arachnoid cysts can expand in all directions from the These small cysts can be missed on CT because of bone artifact and
suprasellar cistern. They may extend inferiorly into the sella turcica, volume averaging; indeed, many CSF collections in the anterior middle
laterally into the middle cranial fossa, and posteriorly into the inter- cranial fossa are not cysts at all, but focal enlargements of the sylvian
peduncular and prepontine cisterns. As the cyst expands superiorly, it fissure. Neither small cysts nor enlargements of the fissures are gener-
fills the space previously occupied by the third ventricle and pushes the ally considered to be of clinical significance.
third ventricle superiorly (Fig. 5-204). In this process, it can disrupt Approximately 50% of middle cranial fossa cysts are of moder-
the pituitary stalk and compress the hypothalamus. In very large cysts, ate size (Fig. 5-205). They occupy the anterior and middle portions of
the superior pole of the cyst can obstruct both foramen of Monro, the temporal fossa and frequently displace the tip of the temporal lobe
causing hydrocephalus; it may even bulge further superiorly between posteriorly, superiorly, and medially. They cause a characteristic flat
the leaves of the septum pellucidum (Fig. 5-204). The posterior pole of medial border of the sylvian fissure and open the lips of the fissure lat-
the cyst may displace the pineal gland inferiorly and cover the entrance erally. The temporal fossa is usually mildly expanded by these lesions.
of the aqueduct. The cyst can stretch the optic nerves and chiasm, as Approximately 30% of middle cranial fossa cysts are large, occupy-
well. Extension into the middle cranial fossae may be seen. MRI usu- ing nearly the whole temporal fossa, sometimes extending to the fron-
ally easily makes diagnosis, with the midline sagittal image showing the tal fossa and convexity (Fig. 5-202). These large cysts cause substantial
cyst displacing the floor of the third ventricle superiorly (Fig. 5-204). mass effect with expansion of the hemicranium, compression of adja-
The major differential diagnoses of suprasellar arachnoid cysts are cent brain and midline shift (Fig. 5-202). In neonates, they cause mac-
epidermoids and cystic astrocytomas. By standard MR and CT tech- rocephaly and plagiocephaly, which is often the reason the children are
niques, epidermoids are usually less sharply marginated than arach- imaged. When located anteriorly, they can extend into the orbit via the
noid cysts and are more heterogeneous; however, there are cases in optic canal. Subdural and intracystic hemorrhages, which may follow
which differentiation by standard spin-echo MRI is impossible. As dis- trauma or occur spontaneously, sometimes complicate middle cranial
cussed in the section on “Epidermoids” in Chapter 7, differentiation fossa arachnoid cysts. The bleeding is probably caused by the disruption

FIG. 5-203. Fetal MRI of a cerebellopontine angle arachnoid cyst. Parasagittal (A) and axial (B) images show the sharply margin-
ated, CSF-intensity cyst (white arrows) situated in the left cerebellopontine angle and displacing adjacent parenchyma. The fourth
ventricle (white arrowhead) is slightly compressed and displaced to the right.

Barkovich_Chap05.indd 526 5/6/2011 9:01:17 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 527

FIG. 5-204. Suprasellar arachnoid cyst. Sagittal T1-weighted image (A) shows the large suprasellar cyst extending upward to com-
press the third ventricle and downward into the prepontine cistern. The optic chiasm (curved white arrow) is displaced superiorly and
anteriorly. Note that the anterior wall of the cyst extends further anteriorly than the anterior cerebral arteries. The anterosuperior
surface of the pons (black arrows) is distorted. The inferior wall of the cyst (small white arrows) can be seen in the prepontine cistern.
The lateral ventricles have been decompressed by a ventriculoperitoneal shunt. Axial T1-weighted reformatted image (B) shows the
cyst (short black arrows) extending into the interpeduncular cistern, displacing the cerebral peduncles. T1-weighted images reformat-
ted in the coronal plane (C and D) show the cyst extending laterally into the right middle cranial fossa and displacing the uncus and
parahippocampal gyrus (small arrows in C) and extending upward to obstruct the foramina of Monro. Arrows in (D) point to the cyst
walls at the foramina.

of cortical veins, which frequently traverse the cyst near its periphery. pression of the cyst into the subdural space, perhaps through a weak
These veins bleed readily with slight manipulation at the time of sur- portion of the cyst wall (1111).
gery (1108). Concurrent subdural or intracystic hemorrhage should Approximately 25% of arachnoid cysts occur in the posterior
be carefully ruled out in any patient with signs of arachnoid cyst and fossa (Fig. 5-207). These lesions are usually large (>5 cm) at the time
acute progression of symptoms. Specific attention should be paid to of presentation (1097,1106,1112). Although the cysts can occur any-
the temporal fossa in any patient with clinical signs consistent with an where in the posterior fossa, the cerebellopontine angle and inferior
arachnoid cyst and radiographic evidence of a temporal fossa hem- or dorsal midline (Fig. 5-207) are the most common locations. The
orrhage. Similarly, the possibility of concurrent subdural hematoma major importance of these lesions is in their differentiation from the
should be considered whenever a sylvian fissure cyst appears to be cysts of the Dandy-Walker complex. This distinction is made via a
separated from the inner table of the calvarium (1097). Middle cranial combination of clinical and radiological features. Patients with retro-
fossa arachnoid cysts may rupture into the subdural space spontane- cerebellar arachnoid cysts are most commonly asymptomatic. If they
ously, or after trauma as a result of preexisting or traumatic communi- rapidly enlarge, affected children may present with signs of cerebellar
cation (Fig. 5-206) (1109,1110); this results in formation of a subdural dysfunction (ataxia, dysdiadochokinesis) secondary to compression of
hygroma. Rarely, spontaneous disappearance of arachnoid cysts has the cerebellum by the cyst. In contrast patients with the Dandy-Walker
been reported; this presumably results from spontaneous decom- malformation usually present in infancy either with hydrocephalus or

Barkovich_Chap05.indd 527 5/6/2011 9:01:18 PM


528 Pediatric Neuroimaging

FIG. 5-205. Moderate sized middle cranial fossa arachnoid cyst. Parasagittal T1-weighted image (A) shows the hypointense cyst
(white arrows) displacing the anterior portions of the frontal and temporal lobes posteriorly. The T2-weighted axial image (B) shows
only moderate mass effect with minimal midline shift when compared to the large cyst in Fig. 5-202.

developmental delay (873). Radiologically, differentiation is made by as the walls of cysts (1113). Analysis of CSF flow, using flow-sensitive
determining whether the cyst is an enlarged fourth ventricle (Dandy- steady-state free procession imaging or phase-contrast studies, is
Walker malformation), is in communication with the fourth ventricle sometimes helpful in determining whether there is free communica-
(Dandy-Walker complex, but not the true Dandy-Walker malforma- tion between the cyst and the fourth ventricle; however, flow can be
tion), or whether it is isolated from the surrounding subarachnoid space complex and such studies may be misleading. The most reliable meth-
(arachnoid cyst). Magnetic resonance cisternography with 3D steady- odologies, in particularly difficult cases, are CT cisternography and
state imaging such as CISS or FIESTA can depict thin membranes such ventriculography. These methods will always allow determination as to
whether the cyst communicates with the subarachnoid space (cyst fills
after lumbar injection of iso-osmolar nonionic contrast and tilting the
patient’s head down in the supine position), the ventricular system (the
cyst fills after injection of iso-osmolar nonionic contrast into the lat-
eral ventricle and tilting the head up), or is an isolated arachnoid cyst
or loculation (cyst doesn’t fill on either injection). Large retrocerebel-
lar cysts may be associated with anomalies of the dural venous sinuses
(Fig. 5-207).

Therapy
By far, most arachnoid cysts are asymptomatic and require no treat-
ment. Symptomatic arachnoid cysts can be treated either by fenestra-
tion or by cyst-peritoneal shunting. After shunting, the cyst appears
completely or nearly completely decompressed. After adequate treat-
ment of the cyst, the associated hydrocephalus usually resolves as well.

Craniosynostosis
Nonsyndromic Craniosynostosis
Diagnosis of Craniosynostosis
Craniosynostosis can be broadly grouped into two categories. Primary
craniosynostosis refers to premature fusion of one or more cranial
sutures as a developmental error. The cause is thought to be a develop-
mental anomaly of the skull base. Secondary synostosis refers to pre-
mature sutural closure resulting from other causes such as intrauterine
FIG. 5-206. Spontaneous rupture of a middle cranial fossa arachnoid cyst compression of the skull, the effect of teratogens, or lack of brain
into the subdural space. Noncontrast CT shows a large right middle cranial growth. Craniosynostosis may be seen both in otherwise normal indi-
fossa arachnoid cyst (C) with an enlarged frontal subdural space (SD). The viduals and as a part of syndromes associated with other developmen-
patient has hydrocephalus. tal anomalies (1114–1117). Overall, about 15% of cases are syndromic,

Barkovich_Chap05.indd 528 5/6/2011 9:01:19 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 529

FIG. 5-207. Retrocerebellar and infracerebellar arachnoid cysts. A–C. Retrocerebellar


cyst. Sagittal T1-weighted (A) and axial T2-weighted (B) images show a retrocerebellar
CSF collection (arrows) that is isointense with CSF and is causing scalloping of the inner
table of the occipital bones. Maximum intensity projection from a 2D TOF MR veno-
gram (C) shows an abnormally high bifurcation of the superior sagittal sinus. D and E.
Infracerebellar cyst. Axial SE T2-weighted (D) and coronal FLAIR (E) images show an
infravermian arachnoid cyst (white arrows), superiorly displacing the cerebellar vermis
and laterally displacing the inferomedial aspects of the cerebellar hemispheres.

Barkovich_Chap05.indd 529 5/6/2011 9:01:20 PM


530 Pediatric Neuroimaging

that is, associated with other anomalies of the brain or body. Of the and language deficits (1123). Isolated metopic synostosis accounts
remaining 85%, 75% to 80% are simple nonsyndromic, involving only for 18% of simple NSC cases (1118) and causes a prominent, wedge-
one suture, and 20% to 25% are multisuture nonsyndromic (1118). shaped forehead (trigonocephaly, Fig. 5-209); underlying syndromes,
The precise cause of premature sutural closure has not been fully such as 11q23 deletion, 9p deletion, Opitz C syndrome, and various
clarified. In the normal patient, sutures close when the growth of the aneuploidies, are seen in up to 30% of affected patients (1124). Isolated
brain slows. It is known that early slowing or cessation of brain growth unilateral coronal (20%–30% of NSC, with 60%–75% of those affected
results in early closure of the cranial sutures. An interesting theory, being female, Fig. 5-210) or lambdoid (<5% of NSC, Fig. 5-211) synos-
called the microspicule hypothesis, has been proposed based on find- tosis causes cranial asymmetry (plagiocephaly). Considering only
ings in an animal model (1119). The microspicule hypothesis contends craniosynostosis in which a molecular diagnosis is made, the most
that microspicules of bone are continually growing across open sutures common form is Muenke syndrome, followed by Saethre-Chotzen syn-
but are fractured by calvarial expansion due to the normal forces of drome (1117). Of note, the most common cause of plagiocephaly is not
growth. If the normal forces causing expansion of the calvarium dis- synostosis, but “positional flattening” resulting from the neonate being
appear, due to either bone anomaly or lack of cranial growth, the positioned almost exclusively in the supine position with the head usu-
microspicules are not fractured and the sutures begin to close, result- ally tilted to the same side, only bottle-feeding with the bottle on the
ing in premature synostosis (1119). More recent molecular biologic same side, and slow motor development (1125).
studies suggest that interactions of fibroblast growth factors (FGFs) Multiple suture involvement is seen in about 5% of patients with
and their receptors (FGFRs) influence the balance between the pro- NSC (1126). Clinically, they are divided into two groups; those with
liferation of osteoprogenitor cells and the differentiation of new bone premature fusion of two sutures and those with complex craniosynos-
in the membranous bones of the calvarium; these processes take place tosis due to involvement of more than two sutures (1126). Patients
at the sutures (1117). When the balance is shifted from proliferation with two-suture fusion have similar courses to those with monosu-
of progenitors to production of bone, the sutures appear to close. All tural fusion (normal development, normal intelligence, and normal
of the genes that have been found to be causative in craniosynostosis intracranial pressure) other than a higher rate of reoperation (25% vs.
syndromes (FGFR1, FGFR2, FGFR3, TWIST1, and MSX2) seem to par- 5%) (1118,1121,1127). In contrast, those with complex craniosynos-
ticipate in the molecular pathways involved in this process (1117). Still tosis have a high incidence of increased intracranial pressure, Chiari I
another study suggests that the formation of the membranous bone malformation, and developmental delay, while the rate of reoperation
of the calvarium is related to the BMP pathway, which is initiated in is 37% (1118). Indeed, it seems likely that mutations and diagnosis
the skin ectoderm in the dorsal midline. It appears that expression of of specific syndromes are significantly higher among complex cran-
a BMP inhibitor (noggin) maintains the patency of the cranial sutures iosynostosis than among simple ones (1128). Of NSC patients with
until some event related to slowing of brain growth causes the expres- multiple suture involvement, 40% to 50% have oxycephaly secondary
sion of noggin to decrease (1120). As molecular biologic techniques to bilateral coronal and lambdoid synostosis, 30% to 40% have brachy-
improve, the precise factors involved in the development and subse- cephaly secondary to bilateral coronal synostosis, and about 20% are
quent closure of the cranial sutures are slowly being elucidated. “unclassified” secondary to multiple assorted sutural synostoses. It is
When isolated to a single suture, craniosynostosis is usually spo- important to remember that some cases of single or dual suture synos-
radic and presents as an abnormally shaped head. Isolated sagittal tosis may be genetic. This is particularly true for coronal synostoses;
synostosis accounts for about 60% of simple nonsyndromic cranio- the recurrence risk for bilateral coronal synostosis is 5% to 7% (1129).
synostosis (NSC) cases (1121) and causes a long, narrow head shape Bilateral lambdoid synostosis should raise suspicion for rhomb-
(scaphocephaly or dolichocephaly, Fig. 5-208); this disorder is easy to encephalosynapsis (see earlier section in this chapter on cerebellar
diagnose clinically and probably does not need radiologic evaluation malformations). Craniosynostosis syndromes are discussed in the fol-
unless it is atypical (1122). Of note, affected patients may have speech lowing section.

FIG. 5-208. Scaphocephaly secondary to sagittal suture


synostosis. Axial CT image (A) shows an elongated, narrow
head secondary to sagittal synostosis. Note the ridge of bone
(white arrow) along the prematurely closed suture. Superior
view (B) of 3D reformation shows the closed sagittal suture
with ridge (large black arrows) and the open metopic (white
arrows), coronal (large black arrowheads), and lambdoid
(small black arrowhead) sutures.

Barkovich_Chap05.indd 530 5/6/2011 9:01:22 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 531

FIG. 5-209. Trigonocephaly. Axial


CT shows (A) a pointed frontal region
(arrows) secondary to premature clo-
sure of the metopic suture. 3D sur-
face reformation of the calvarium (B)
shows a ridge of bone (black arrows)
along the prematurely closed suture.

Radiology of Synostosis request 3D reconstructions of the calvarium (derived from CT scans,


Isolated sagittal, metopic, or unilateral coronal synostosis is diagnosed see Figs. 5-208 to 5-211) in order to plan their surgery. Neuroimaging
clinically and confirmed by plain films of the skull; cross-sectional is also performed to look for underlying brain anomalies; therefore,
imaging studies are not indicated to assess the sutures. The characteris- both soft tissue CT images (or MRI) to look at the brain and bone
tic findings on skull films are available in any standard text on pediatric algorithm CT images to look at the calvarium should be photographed
radiology. Briefly, premature sagittal synostosis causes scaphocephaly or archived. Infants with trigonocephaly may have anterior midline
and ridging along the sagittal suture (Fig. 5-208), premature metopic anomalies (Opitz C syndrome, Jacobsen syndrome, holoprosencephaly,
synostosis causes trigonocephaly and ridging along the metopic suture or many others [1126,1128]). Those with unilateral posterior plagio-
(Fig. 5-209), and premature coronal synostosis shows plagiocephaly cephaly (unilateral occipital flattening) may have an underlying brain
and an uplifting of the lateral orbital roof (Fig. 5-210), giving the anomaly or brain damage (either anatomic or secondary to metabolic
“Harlequin eye” appearance. disease). Both brain anomalies and brain damage can cause reduced
Although experienced surgeons may treat single suture synos- infant mobility and result in extended periods of lying on their backs,
tosis without the assistance of imaging studies, many surgeons now primarily on one side. This secondary flattening, resulting from lack of

FIG. 5-210. Plagiocephaly secondary to unilateral coronal suture synostosis. Bone window image from axial CT scan (A)
shows plagiocephaly; the right hemicalvarium is smaller than the left. The left coronal suture (white arrow) is open, but no
right coronal suture can be seen. The sagittal suture (white arrowhead) is displaced to the right. Anterior view of 3D reforma-
tion of the calvarium (B) shows the open left coronal suture (small black arrows) and fusion of the right (black arrowheads).
Note the elevation of the right orbital roof (large black arrows), the so-called Harlequin eye sign of coronal synostosis.

Barkovich_Chap05.indd 531 5/6/2011 9:01:22 PM


532 Pediatric Neuroimaging

FIG. 5-211. Plagiocephaly secondary to unilateral lambdoid suture synostosis. Bone window from axial CT scan
(A) shows flattening of the right occipital region (white arrowheads) and bulging on the left (white arrow) secondary
to closure of the left lambdoid suture. The ear is in normal position or slightly posterior. Postero-inferior view of 3D
reformation of calvarium (B) shows better the bulging of the left occiput (white arrow) ipsilateral to the synostosis.
The right lambdoid suture (black arrows) is open.

infant movement, is usually not associated with synostosis. Sawin et al. the position of the ipsilateral ear (as above, with lambdoid synostosis,
(1130) have noted that occipital flattening (occipital plagiocephaly) is the ipsilateral ear is posteriorly positioned—pulled back by sutural
commonly associated with benign enlargement of the subarachnoid fusion—whereas the ear is anteriorly positioned—pushed forward—
spaces (typically frontal, sylvian, and basal cisterns), usually without in infants with positional flattening of the occiput) (1137). CT should
ventriculomegaly (see Chapter 8 for a discussion of benign enlarge- be obtained if surgery is contemplated, as it more accurately depicts the
ment of the subarachnoid spaces); they postulated that increased cranial sutures; in particular, 3D reformations should be constructed
compliance of the skull in the presence of large subarachnoid spaces from the CT data (Figs. 5-208, 5-210, and 5-211). 3D reformations of
predisposes the patients to positional flattening. Other predisposing CT images of the calvarium are the definitive method of making the
factors for posterior plagiocephaly include supine positioning during radiologic diagnosis of suture synostosis, but they should be acquired
early infancy (overwhelmingly most common), torticollis, prematurity, with as low a dose of radiation as possible while performing a technically
and developmental delay (1131). It is important to emphasize that this adequate study (see Chapter 1).
positional flattening of the skull is usually not associated with any neuro- It is important to recognize that sutures may appear falsely closed
logic abnormality, but is merely a result of laying supine for long periods of when the plane of imaging is parallel to the suture. When the reforma-
time, which is common because supine positioning of infants seems to tions are constructed from axial images, the upper part of the lamb-
reduce the incidence of sudden infant death syndrome (1132). Features doid suture is the most common area to falsely appear closed. Certain
that are useful in helping to identify nonsynostotic occipital plagio- compensatory changes in calvarial shape are seen with occipital plagio-
cephaly are forward displacement of the ipsilateral ear (with lambdoid cephaly of any etiology. The anterior portion of the ipsilateral middle
synostosis the ipsilateral ear is posteriorly displaced) and ipsilateral cranial fossa is enlarged. The orbit is elevated and there is increased lat-
frontal bossing (1133). In the absence of underlying neurologic abnor- eral convexity of the squamous portion of the ipsilateral temporal bone.
mality, the vast majority of cases of occipital plagiocephaly are non- Addition abnormalities found in isolated coronal synostosis include
synostotic and improve once the children no longer lay on the same expansion of the ipsilateral inferior occiput (below the suture), narrow-
side of their head all the time (1131,1133–1135). If necessary, the child ing of the sphenopetrosal angle, decreased size of the ipsilateral frontal
may wear a helmet during sleep (1134–1136). fossa, flattening and posterior displacement of the ipsilateral frontal cal-
The radiologic evaluation of children with unilateral occipital pla- varium, compression of the ipsilateral anterior cranial hemifossa, and
giocephaly is evolving. In most cases, when the children are develop- medial, posterior, and superior displacement of the orbital roof (1138).
ing normally, no radiologic studies are necessary. A follow-up clinical Anomalies of venous drainage can occur with multisutural synos-
exam after instructing parents to change head positions in the crib or, tosis of both syndromic and nonsyndromic causes (1139,1140). If sur-
in older infants, using molding helmets, will usually yield a confident gical intervention is being contemplated, it is essential to evaluate the
diagnosis (1134). Plain skull films are not helpful in either ruling out intracranial venous sinuses with venography (MR venography is the
underlying brain abnormality or in diagnosing lambdoid synostosis. study of choice because of the absence of ionizing radiation) (1141).
If the children have any neurologic abnormalities, an MRI should Abnormal venous pressure may be associated with increased intrac-
probably be obtained, as MRI is the most sensitive method for detect- ranial pressure (or frank hydrocephalus) and consequences of this
ing brain abnormalities. MRI can also provide information regarding elevated pressure should be contemplated and addressed in advance

Barkovich_Chap05.indd 532 5/6/2011 9:01:23 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 533

(974,1139). Moreover, in the setting of increased intracranial venous anomalies of other bones, usually the hands and, particularly, fingers.
pressure, transcalvarial emissary veins may enlarge around the cranial Approximately 15% of all craniosynostosis cases present with associ-
sutures (974); surgeons should be alerted to this possibility. ated anomalies involving mainly the face and limbs and are consid-
ered to be syndromic (1118,1128). More than 180 syndromes manifest
Abnormal Fontanelle Closure craniosynostosis and at least half of them seem to follow Mendelian
The anterior fontanelle is almost always seen on axial CT scans of patterns of inheritance (London Dysmorphology Database, http://
neonates and infants. In general, the fontanelle should not close until www.lmdatabases.com/about_lmd.html). From an embryological per-
the age of about 12 months, and should be closed by about 18 months. spective, genetic studies show that some calvarial cells are increased in
Premature closure of the fontanelle is sometimes a sign in impaired number in children with FGFR2 mutations and that these cells have
brain growth; in such cases, it is associated with microcephaly and an increased maturation rate, leading to increased subperiosteal bone
with premature suture fusion. Delayed closure of the anterior fon- matrix formation and premature ossification of the calvarium during
tanelle is usually associated with syndromic craniosynostosis, in fetal development (1142).
which the inability of the calvarium to grow at the sutures is partially Although several different syndromes are defined on the basis of
compensated by the persistent patency of the fontanelle, allowing clinical features, a precise classification of affected patients is difficult
the brain to grow upward (turricephaly). Other conditions in which because of considerable overlap (1128). In addition, patients with
the fontanelle closure is delayed include athyrotic hypothyroidism, mutations of the same gene can have different phenotypic manifes-
bone dysplasias (achondrogenesis Type II, campomelic dysplasia, tations and vice versa (1143–1146). Identical FGFR2 mutations have
cleidocranial dysostosis, hypophosphatasia, osteogenesis imperfecta, been found in patients carrying the diagnosis of Crouzon, Pfeiffer,
pycnodysostosis, thanatophoric dysplasia, rickets), and chromosomal and Jackson-Weiss craniosynostosis syndromes, suggesting that these
anomalies (Down syndrome, Trisomy 13p, Aase syndrome, progeria, entities may represent a clinical spectrum of the same disorder, with
Russell-Silver syndrome, Triploidy syndrome, Trisomy 9p, Trisomy 18, the differences resulting from the effect of genetic modifiers (1126).
and Zellweger syndrome). In addition, an identical FGFR2 mutation has been found in unrelated
patients with Pfeiffer and Apert syndromes (1147). Moreover, the same
phenotype can be caused by mutations in different genes, as in cases of
Syndromic Craniosynostosis
Pfeiffer syndrome due to FGFR1 and FGFR2 mutations (1145). Thus,
Background as in many genetic disorders, the precise phenotype of the affected
Syndromic craniosynostosis includes those disorders in which the patient is mostly likely related to the effects of both epigenetic factors
face and the skull develop abnormally, typically in conjunction with and gene mutation upon the function of the protein product; many

TABLE 5-10 Inherited forms of craniosynostosis


Syndromes/OMIM
(McKusick) Number Site of Mutation Classification Essential Features Inheritance
Apert, Apert- Chromosome Acrocephalosyndactyly I, II Craniosynostosis, severe syndactyly of Autosomal
Crouzon/101200 10q26 (FGFR2) hands and feet, down-turned mouth, dominant
hypertelorism
Nonsegmentations of C-spine
Venous anomalies
Chiari I malformations
Callosal, septal ageneses
Limbic anomalies
Saethre- Chromosome Acrocephalosyndactyly III Craniosynostosis, facial asymmetry, low Autosomal
Chotzen/101400 7p21.2 hair line, ptosis, deviated nasal septum, dominant
(TWIST) syndactyly of second, third fingers
Normal cognition
Muenke/602849 Chromosome Nonsyndromic coronal Craniosynostosis, thimble-like middle Autosomal
4p16.3 (FGFR3) craniosynostosis phalanges, coned epiphyses, carpal dominant,
and tarsal fusions usually
Commonest syndromic cause of paternal
craniosynostosis, usually unilateral or
bilateral coronal
Pfeiffer, Chromosome Acrocephalopolysyndactyly Craniosynostosis, malformed enlarged Autosomal
Noack/101600 10q26 (FGFR2) I, V thumb and great toe, soft tissue syndac- dominant
Chromosome tyly of second, third digits, polydactyly,
8p11 (FGFR1) stenosis/atresia of external auditory
Others (?) canal, normal intelligence

(Continued)

Barkovich_Chap05.indd 533 5/6/2011 9:01:24 PM


534 Pediatric Neuroimaging

TABLE 5-10 Inherited forms of craniosynostosis (Continued)


Syndromes/OMIM
(McKusick) Number Site of Mutation Classification Essential Features Inheritance
Carpenter, Summitt, unknown Acrocephalopolysyndactyly Craniosynostosis, preaxial polydactyly, Autosomal
Goodman, Sakati- II, III, IV soft tissue syndactyly, malformed ears, recessive
Nyhan/201000 obesity, hypogenitalism, variable mental
retardation
Crouzon, craniofacial Chromosome Craniosynostosis with other Craniosynostosis, maxillary hypoplasia, Autosomal
dysostosis/123500 10q26 (FGFR2) somatic abnormalities shallow orbits with proptosis, bifid dominant
uvula or cleft palate
Crouzon with Chromosome Chiari I malformations
acanthosis nigrans 4p16.3 (FGFR3) Venous anomalies
Jugular venous stenosis
Craniosynostosis, Unknown Craniosynostosis with other Craniosynostosis and fibular aplasia Autosomal
fibular aplasia, somatic abnormalities recessive
Lowry/218550
Craniosynostosis, Chromosome Craniosynostosis with other Craniosynostosis, varus deformity of Autosomal
midface hypopla- 10q26 (FGFR2) somatic abnormalities great toes and tarsal fusion dominant
sia, foot defects, Chromosome
Jackson- 8p11 (FGFR1)
Weiss/123150
Craniosynostosis, Unknown Craniosynostosis with other Craniosynostosis, cleft lip and palate, Autosomal
mental retardation somatic abnormalities choroidal coloboma, mental retardation recessive
clefting/218650
Craniosynos- Chromosome Craniosynostosis with other Synostosis of one or more sutures, Autosomal
tosis, radial 8q24.3 somatic abnormalities bilateral radial aplasia recessive
aplasia, Baller- (RECQL4)
Gerold/218600
Craniosynostosis, Chromosome Craniosynostosis with other Synostosis of multiple sutures, mandibu- Autosomal
arachnodactyly 15q21.1 (FBN1) somatic abnormalities lar and maxillary hypoplasia, multiple dominant
hernia, Shprintzen- abdominal hernias, exophthalmos,
Goldberg/182212 mental retardation
Craniosynostosis Chromosome Craniosynostosis (variable) Synostosis ranges from metopic ridging to Autosomal
Type 2 (Boston 5q34–5q35 with digital abnormali- cloverleaf skull dominant
Type Craniosynos- (MSX2) ties (short metatarsals)
tosis)/604757
X-linked hypo- Chromosome Sagittal synostosis with Sagittal synostosis, dolichocephaly. Occa- Autosomal
phosphatemic Xp22.2–22.1 growth retardation, sionally multiple sutures recessive
rickets/307800 (PHEX) rachitic/osteomalacic
bone disease, hypophos-
phatemia, and renal
defects in phosphate
resorption

craniosynostosis syndromes are allelic and have overlapping features. with both coronal sutures (and, sometimes, both lambdoid sutures),
Nonetheless, it is useful to have some method to classify these disor- the membranous bone of the calvarium expands between the sutures
ders, and Table 5-10 is included for this purpose. but not at them, resulting in a characteristic lobulated skull con-
Of the many types of suture synostosis patterns, bilateral coronal figuration known as cloverleaf skull or Kleeblattshädel (Fig. 5-213).
synostosis (Fig. 5-212) is the most common combination, but com- Multiple synostosis syndromes may be isolated or associated with
bined bilateral coronal and lambdoid synostosis or a combination syndactyly, polydactyly, fusion of cervical vertebrae, anomalies of
of bilateral coronal synostosis and sagittal synostosis can be seen, the middle ear, or other somatic anomalies (Table 5-10) (1114–
among others. When the sagittal suture is synostotic in conjunction 1116,1128,1148,1149).

Barkovich_Chap05.indd 534 5/6/2011 9:01:24 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 535

FIG. 5-212. Turricephaly secondary to bilateral coronal synostosis.


Sagittal T1-weighted image (A) shows the “tower head” appearance
with the frontal lobe extending upward (white arrows) through the
late-closing anterior fontanelle and the anterior corpus callosum
(black arrows) being pulled up, as well. Axial T2-weighted images
show lateral expansion of the temporal lobes (white arrows in B) and
shortening of the frontal lobes (white arrows in C) secondary to the
shortened anterior cranium. Lateral view of 3D reformation of cal-
varium (D) shows complete closure of coronal suture, while superior
view (E) shows the persistently open fontanelle (black arrows).

Barkovich_Chap05.indd 535 5/6/2011 9:01:24 PM


536 Pediatric Neuroimaging

FIG. 5-213. Kleeblattschädel with venous anomaly and hemorrhage in a neonate with no identified genetic disorder. Sagittal
T1-weighted image (A) shows an extremely small posterior fossa, leading to compression and inferior displacement of posterior
fossa structures. The head is towering and the ventricles are enlarged secondary to hydrocephalus. Posterior fossa is small and
posterior fossa structures are crowded, resulting in severe herniation of posterior fossa structures through the foramen mag-
num. Axial noncontrast CT scan (B) shows hypertelorism, lateral bulging of the middle cranial fossae, and an area of mixed
hypodensity (solid arrows) and slight hyperdensity (open arrows), indicating a hemorrhage, in the right temporal lobe. Coronal
T1-weighted image (C) shows the right temporal lobe hemorrhage (white arrows), the markedly enlarged ventricles, and the
bulging of the middle cranial fossa. The configuration of the calvarium is that of a cloverleaf skull. Maximum intensity projection
from an MR venogram (D) shows nearly complete absence of the transverse and sigmoid sinuses, suggesting that the temporal
lobe hemorrhage was the result of venous hypertension or infarction.

Barkovich_Chap05.indd 536 5/6/2011 9:01:26 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 537

enlargement of the frontal horns. This combination of forces results


TABLE 5-11 Key Imaging Findings in in an appearance of turricephaly (tower head, Figs. 5-212 to 5-214).
In addition, because of the premature sutural fusion, the skull base
Craniofacial Anomalies and posterior fossa are abnormally small. This characteristic skull
shape can be detected prenatally with fetal MRI (Fig. 5-215) (1150)
Common and should allow prenatal diagnosis of the multiple suture synostosis
Multiple sutural synostosis syndrome, although the details to diagnose the precise syndrome may
not be visible.
Enlarged anterior fontanelle Anomalies of the underlying brain are present in a significant
Small skull base number of affected patients (1114,1151,1152) and their identification
is the primary reason for obtaining MR studies. Although PMG is not
Ventriculomegaly/hydrocephalus
described in these disorders, we have seen several cases (Fig. 5-216).
Anomalies of venous drainage This finding is not altogether unexpected in view of the known inter-
Dysplastic calvarial bone action of the cerebral cortex and overlying meninges (often abnormal
in craniosynostosis syndromes) during development (414,1153). Ven-
Anomalies of the external and middle ear
triculomegaly (Figs. 5-213 and 5-214) is very common, probably as a
Less common result of the small skull base (resulting in mechanical resistance to CSF
Tonsillar herniation/Chiari I malformation outflow) and small jugular foramina (resulting in increased resistance
to venous outflow); this situation results in increased venous pressure,
Agenesis/hypogenesis of the corpus callosum venous engorgement, cephalocranial disproportion, and increased
Agenesis/hypogenesis of the septum pellucidum CSF pressure (973,1151,1154–1157) (Figs. 5-213 and 5-214). Frank
Dysmorphic hippocampi hydrocephalus is present in approximately 12% of affected patients
(Fig. 5-213) (1158) and is most common in Crouzon and Pfeiffer syn-
Dysmorphic cerebral cortex dromes (as high as 60% in some series [1156]). If nearly all sutures are
closed, the ventricles may remain small in spite of increased intrac-
ranial pressure, enlarging only after craniofacial surgery has opened
the calvarium (1158) (see discussion in Chapter 8). The differentia-
Imaging of Craniosynostosis Syndromes tion of benign ventriculomegaly from hydrocephalus can be difficult,
As stated above, considerable overlap exists among the craniofacial as discussed in Chapter 8, and is dependent upon assessment of subtle
manifestations of the different syndromes; therefore, a general descrip- features such as the degree of enlargement of the anterior recesses of
tion of the imaging findings will be given (for a summary, see Table the third ventricle. Of note, enlargement of the temporal horns is not
5-11). The shape of the head, as viewed on sagittal MR images, is a good sign of hydrocephalus in this group of patients, as the temporal
quite characteristic in most patients with multiple sutural synostosis horn enlargement is merely a consequence of the middle cranial fossa
syndromes. Because the coronal, sagittal, and lambdoid sutures close being disproportionately enlarged (Fig. 5-217).
early, the brain grows upward and anteriorly through a very enlarged Venous anomalies are common and may be the result of diver-
anterior fontanelle (Figs. 5-212 to 5-214), resulting in disproportionate sion of venous outflow through enlarged emissary veins (1140,1158).

FIG. 5-214. Apert syndrome with calvarial anomaly, venous anomalies, and cephalocele. A. Sagittal T1-weighted image
shows turricephaly with enlarged ventricles. The cerebrum towers anteriorly via an enlarged anterior fontanelle. The
skull base is small and the posterior fossa structures are crowded. B. Axial T1-weighted image shows disproportionate
enlargement of the frontal horns secondary to upward extension of the cerebrum through the anterior fontanelle.

Barkovich_Chap05.indd 537 5/6/2011 9:01:27 PM


538 Pediatric Neuroimaging

FIG. 5-214. (Continued) C and D. Axial T2-weighted images show the dysplastic bone of the calvarium, with irregular spicules
extending inward; the meninges are always dysplastic, as well. Note the hypertelorism and the extension of the left occipital lobe
(encephalocele, white arrows) through the calvarium in (D). E and F. Axial CT images in a different patient show the herniation of
brain tissue (white arrows) through a defect in the occipital bone and the spicules of bone extending inward from the calvarium.

Barkovich_Chap05.indd 538 5/6/2011 9:01:28 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 539

FIG. 5-214. (Continued) G. MR venogram shows hypoplasia of


the left transverse sinus and abrupt termination (black arrow) the
right transverse sinus. H. Lateral view of 3D reformation of CT
scan shows the turricephaly and small skull base characteristic
of Apert syndrome. I. Anterosuperior view of 3D reformation of
CT scan shows bilateral coronal suture closure and the marked
enlargement of the anterior fontanelle (black arrows) that results
from the head growth in the setting of bilateral coronal suture
synostosis.

Increased intracranial pressure typically appears during the second through the foramen magnum, usually called Chiari I malformations
year of life, as the cranial sutures begin to close (1157), but may nor- (Figs. 5-212 and 5-217), in these syndromes (973,1158,1162). This phe-
malize as the emissary veins enlarge (emissary veins carry the venous nomenon seems to be particularly common in Crouzon syndrome, in
blood from intracranial to extracranial veins through enlarged emis- which lambdoid sutures close particularly early and tentorial attach-
sary foramina in the calvarium) (1159). Small jugular foramina are ment to the calvarium is particularly low (973), in Pfeiffer syndrome
present in the majority of patients with venous anomalies and are the (Fig. 5-217), and in the Kleeblattschädel deformity (Fig. 5-213) (974).
probable cause of venous hypertension (1141,1160). As described in Other common brain anomalies in craniosynostosis syndromes include
the previous section on nonsyndromic craniosynostosis, preoperative cortical dysplasias, callosal agenesis or hypogenesis, agenesis or hypo-
identification of venous anomalies is important for surgical planning; genesis of the septum pellucidum, distortions or malformations of the
therefore, venography should be a part of the preoperative evalua- hippocampus, and cephaloceles (Fig. 5-217) (1114,1140,1151,1152,
tion of these patients (Figs. 5-212 and 5-214) (1139,1141,1161). MRV, 1154,1163); all these seem to be most common in Apert syndrome.
catheter venography, or CTV will work, but CTV gives the most detail CT with bone windows or the first echo T2 sequence on MRI often
regarding the small venous channels and their transcalvarial course, shows a dysmorphic-appearing calvarium with thin, irregular bone
even if low dose techniques are used (see Chapter 1). (Figs. 5-214 and 5-217) that is difficult to separate from the underlying
The small skull base, in conjunction with high intracranial pres- dura at the time of surgery. Cephaloceles may result from brain, CSF,
sure, is the probable cause of the high incidence of cerebellar herniation or dura herniating through the abnormal skull (Figs. 5-214 and 5-217)

Barkovich_Chap05.indd 539 5/6/2011 9:01:29 PM


540 Pediatric Neuroimaging

FIG. 5-215. Fetal MR images of craniosynostosis syndromes. A and B. Apert syndrome at 23-week gestational age. Sagittal image
(A) shows no evidence of turricephaly; however, coronal image (B) shows some expansion of the middle cranial fossae (white
arrows) already at this age. C and D. Apert syndrome at 31-week gestational age. Sagittal image (C) shows superior expansion of the
frontal calvarium (white arrows), compatible with turricephaly. Coronal image (D) shows more marked expansion of the middle
cranial fossae (arrows) and some ventriculomegaly.

(1140). Patients with multiple synostoses have hypertelorism and shal- neuroradiologic findings (1164). An amazing website, called OMIM,
low orbits with marked proptosis (Figs. 5-213, 5-214, and 5-217). In has been created and has an enormous amount of information on
addition, anomalies of the external and middle ear, again resulting most diseases that have been described (http://www.ncbi.nlm.nih.gov/
from the abnormal formation of the skull base, will often be found if omim/). Readers are highly recommended to visit this site when ques-
1 mm sections are acquired with bone algorithm through the petrous tions arise concerning specific disorders. I have tried to include the
pyramids (1114,1163). chromosomal mutations associated with malformations in the appro-
priate sections of this and other chapters (especially Chapters 3 and 6).
The field is rapidly evolving as the genetic bases of many anomalies are
CHROMOSOMAL ANOMALIES elucidated. It is interesting to note that many sets of siblings have iden-
Chromosomal anomalies have only recently been systematically tical dysmorphic features and brain anomalies that still fit no known
catalogued and characterized with respect to their pathologic and genetic syndrome. As genetic evaluations become more common and

Barkovich_Chap05.indd 540 5/6/2011 9:01:30 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 541

FIG. 5-216. Apert syndrome with polymicrogyria.


Sagittal T1-weighted image (A) shows marked turri-
cephaly (white arrows) and an enlarged proximal aque-
duct (white arrowheads). Posterior fossa structures are
crowded. Axial T2-weighted images (B and C) show
areas of dysplastic bone/meninges (white arrows) and
several areas of abnormal-appearing cortex (black
arrows), consistent with polymicrogyria.

more precise, the chromosomal abnormalities that cause these disor- live births (1165). The disorder is a leading cause of mental retarda-
ders will most likely be discovered. tion with 80% of affected patients being severely retarded (IQ < 50).
A few well-known genetic syndromes that affect the brain and Although examination of the brain at autopsy shows many abnormali-
their associated brain anomalies are discussed in this section. More ties, including senile plaques and neurofibrillary tangles at an early age,
complete lists can be found in genetics and pediatric textbooks and reduced brain weight, a narrow superior temporal gyrus, and hypoplas-
on OMIM. Ideally, as essentially all of the major types of brain anoma- tic inferior frontal gyrus, imaging studies are not dramatic. Patients are
lies that are seen in genetic disorders are described in this chapter, this brachycephalic with a short, round brain, reduced AP diameter of the
imaging information, combined with the lists of chromosomal disor- frontal lobes, a narrow superior temporal gyrus, small cerebellum and
ders and their intracranial anomalies, will allow diagnoses to be more pons, and forward kinking of the brainstem (1166). Premature cerebral
easily established. In this section, I will concentrate on the well-defined atrophy and senile calcifications of the globus pallidus may be seen.
syndromes of trisomy 21 (Down syndrome), trisomy 18 (Edwards Cervical spine abnormalities are common in Down syndrome.
syndrome), trisomy 13 (Patau syndrome), the Fragile X syndrome, The most common anomaly is atlantoaxial subluxation, presum-
inversion duplication of the short arm of chromosome 8, and the Wolf- ably as a result of ligamentous laxity, which has a prevalence of 10%
Hirschhorn syndrome. to 22% (1167,1168). Ligamentous laxity may also contribute to
the increased incidence of atlanto-occipital subluxation in affected
patients (1169,1170). Superimposed upon the ligamentous laxity,
Trisomy 21
a high incidence of bony anomalies of the upper cervical spine puts
Down syndrome, caused by trisomy of chromosome 21, is the most the affected child in even greater jeopardy of spinal cord injury with
common chromosomal disorder with an incidence of about 1 in 1,000 relatively minor trauma. Odontoid anomalies, such as hypoplasia and

Barkovich_Chap05.indd 541 5/6/2011 9:01:31 PM


542 Pediatric Neuroimaging

FIG. 5-217. Infant with severe craniofacial anomaly from Pfeiffer syndrome. A. Sagittal T1-weighted image shows marked turricephaly with a very small
posterior fossa. The cerebellum is herniated (black arrows) through the foramen magnum. An occipital cephalocele (white arrows) herniates through a
calvarial defect. Anomalies of venous drainage were present. B. Coronal T1-weighted image shows enlarged middle cranial fossae and resultant temporal
horn enlargement. C. Axial T2-weighted image shows bilateral proptosis, enlarged temporal horns, and markedly compressed posterior fossa structures. The
cerebellum (black arrowheads) wraps around the brainstem. D. Axial T1-weighted image shows marked brachycephaly, expanded middle cranial fossae, and
cerebellar tissue herniated though the midline occipital calvarial defect (black arrows) to form an encephalocele. E and F. Anterior (E) and posterior (F) views
of 3D reformation of the skull show the enormous anterior fontanelle (black arrows in E), the posterior calvarial defect of the cephalocele (black arrowheads
in F), and the “Swiss cheese” appearance of the posterior calvarium resulting from the many areas of thinning of dysplastic calvarial bone.

Barkovich_Chap05.indd 542 5/6/2011 9:01:32 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 543

FIG. 5-218. Trisomy 18. Sagittal T1-weighted image (A) shows callosal agenesis, a simplified cerebral cortical gyral pattern, pontine hyp-
oplasia and cerebellar hypoplasia with a large posterior fossa (Dandy-Walker complex). Axial T1-weighted image (B) shows shallow sulci
and a simplified cortical gyral pattern.

dysplasia of the odontoid and os odontoideum, have been described Trisomy 13


(1171,1172). Patients may also have hypoplasia of the posterior arch
of C-1 (1167) and they can develop progressive basilar invagination Trisomy 13, also known as Patau syndrome, occurs in about 1 in 6,000
(1171). The craniocervical junction should always be closely scruti- live births. The clinical features include microcephaly, severe mental
nized in patients with Down syndrome, and, as stated in Chapter 4, retardation, absent eyebrow, shallow supraorbital ridges, anophthalmia,
these patients should be observed regularly for neurologic dysfunction microphthalmia with coloboma, micrognathia, cleft palate, malformed
and neck pain and should be carefully examined after even minor neck ears, and cardiac anomalies. Holoprosencephaly occurs in 80% of affected
trauma. Plain film radiographs of the cervical spine with flexion and infants. In those patients without holoprosencephaly, reported anoma-
extension views are adequate for patients without neurological deficits. lies include callosal agenesis, olfactory hypoplasia, cerebellar cortical dys-
However, patients with neurologic signs or symptoms, or with a history plasias, and inferior vermian hypoplasia (Fig. 5-219) (1165,1173).
of signs or symptoms referable to craniocervical junction anomalies,
should undergo MRI with sagittal flexion and extension T1-weighted
Fragile X Syndrome
images. T2-weighted images should also be obtained to look for areas
of high signal within the cord, indicative of prior injury. The fragile X syndrome (also known as the Martin-Bell syndrome) is
the most common genetic cause of mental retardation, being present
in 1/1,500 live-born boys (1174). The disorder is caused by an unstable
Trisomy 18 sequence of DNA in the FMR1 gene located at chromosome Xq27.3;
the sequence codes for a trinucleotide repeat that is greatly amplified
Trisomy 18, also known as Edwards syndrome, is the second most
in affected individuals (1175,1176). Affected males have a prominent
common trisomy encountered in newborns, occurring in about 1 in
forehead and mandible, narrow midface, large ears, and macroorchid-
5,000 live births. Patients are dolichocephalic with malformed low set
ism; they may show autistic behavior, perseverative speech, and tac-
ears, micrognathia, short upper lip, short palpebral fissures, epicanthal
tile defensiveness (1177). Pathologic findings include subependymal
folds, midline facial clefts, ptosis, corneal opacities, and microphthal-
and subpial heterotopia, siderosis of the globus pallidus with myelin
mos. Characteristic hand and foot anomalies, where the fingers flex
loss, and hypoplasia of the cerebellar vermis (4). The most commonly
and override (most commonly, the index finger overlaps the long fin-
described neuropathologic abnormality is abnormal dendritic spine
ger and the small finger overlaps the ring finger) and the great toe is
morphology, leading to abnormal synapses. Most patients with Fragile
dorsiflexed, are common. Anomalies of the cardiac, gastrointestinal,
X syndrome have normal imaging studies, although early atrophy has
and urogenital systems are common (717,1165,1173).
been reported. Preliminary studies indicate that fractional anisotropy
The most consistently identified neuropathologic findings are
is reduced in the frontal lobes of affected patients (1178).
anomalies of the cerebral gyral pattern, hypoplasia of the cerebel-
lum and basis pontis, hypogenesis or agenesis of the corpus callo-
sum, and dysplasia of the hippocampus, lateral geniculate body, and Inversion Duplication of Short Arm of
inferior olivary nucleus (717). CT may show dolichocephaly and cer-
Chromosome 8
ebellar hypoplasia but is not as useful as MRI in demonstrating the
gyral anomalies, callosal hypogenesis, and hippocampal dysplasia Inversion duplication of the short arm of chromosome 8 (ID8p) is a
(Fig. 5-218) (1165,1166). rare unbalanced chromosome aberration with an estimated incidence

Barkovich_Chap05.indd 543 5/6/2011 9:01:33 PM


544 Pediatric Neuroimaging

FIG. 5-219. Trisomy 13. Sagittal T1-weighted image (A) shows mild callosal hypogenesis with mild vermian hypoplasia.
Oblique coronal T1-weighted image (B) shows bilateral hippocampal hypoplasia.

of 1 in 10,000 to 30,000 newborns (1179). The clinical features vary, but 5. Jiang Y, Langley B, Lubin F, et al. Epigenetics in the nervous system.
affected children are generally characterized by minor facial dysmor- J Neurosci 2008;28:11753–11759.
phisms, severe developmental delay, infantile hypotonia (which pro- 6. Leventer RJ, Guerrini R, Dobyns WB. Malformations of cortical
gresses to spasticity), feeding difficulties, and scoliosis (1180). Epilepsy development and epilepsy. Dialogues Clin Neurosci 2008;10:47–62.
7. Barkovich AJ, Kuzniecky RI, Jackson GD, Guerrini R, Dobyns WB. A devel-
develops in about 30% (1180). The diagnosis can be made by routine
opmental and genetic classification for malformations of cortical develop-
chromosomal analysis. Imaging typically shows callosal agenesis or ment. Neurology 2005;65:1873–1887.
severe hypoplasia, hypoplasia of the inferior cerebellar vermis (often 8. Patel S, Barkovich A. Analysis and classification of cerebellar malforma-
with enlarged retrocerebellar CSF spaces), delayed white-matter myeli- tions. Am J Neuroradiol 2002;23:1074–1087.
nation, and foci of T2 and FLAIR hyperintensity in the periventricular 9. Barkovich AJ, Millen KJ, Dobyns WB. A developmental classification of
white matter. The white matter abnormalities vary from a few small malformations of the brainstem. Ann Neurol 2007;62:625–639.
spots to multifocal, confluent areas involving most of the brain (1180). 10. Barkovich AJ, Millen KJ, Dobyns WB. A developmental and genetic clas-
sification for midbrain-hindbrain malformations. Brain 2009;132:3199–
3230.
Wolf-Hirschhorn Syndrome 11. Robinson BH. Lactic acidemia. In: Scriver CR, Beaudet AL, Sly WS, Valle D,
eds. The Metabolic Basis of Inherited Disease, 6th ed. New York: McGraw-
The Wolf-Hirschhorn syndrome, caused by partial deletion of the
Hill, 1989:869–888.
short arm of chromosome 4, is characterized by severe growth retar- 12. Barkovich AJ, Peck WW. MR of Zellweger syndrome. Am J Neuroradiol
dation and mental defect, microcephaly, “Greek helmet” facies, and 1997;18:1063–1070.
closure defects (cleft lip or palate, coloboma of the eye, and cardiac 13. Volpe JJ, Adams RD. Cerebro-hepato-renal syndrome of Zellweger. An
septal defects). Most patients are never able to walk without support inherited disorder of neuronal migration. Acta Neuropathol 1972;20:
(1181,1182). Diagnosis is made by characteristic facies and the associ- 175–198.
ated anomalies. Little neuropathology information is available. MRI of 14. Lincke CR, van den Bogert C, Nijtmans LGJ, Wanders RJA, Tamminga P,
the brain is characterized by marked reduction of white matter, callosal Barth PG. Cerebellar hypoplasia in respiratory chain dysfunction. Neuro-
hypoplasia, and, in some cases, characteristic cysts in the periventricu- pediatrics 1996;27:216–218.
lar white matter adjacent to the tips of the frontal horns. 15. Barth PG. Pontocerebellar hypoplasias: an overview of a group of inherited
neurodegenerative disorders with fetal onset. Brain Dev 1993;15:411–422.
16. Colas JF, Schoenwolf GC. Towards a cellular and molecular understanding
REFERENCES of neurulation. Dev Dyn 2001;221:117–145.
17. Müller F, O’Rahilly R. The first appearance of the human nervous system at
1. Kuzniecky R. MRI in cerebral developmental malformations and epilepsy. stage 8. Anat Embryol 1981;163:1–13.
Magn Reson Imag 1995;13:1137–1145. 18. Müller F, O’Rahilly R. The first appearance of the major divisions of the
2. Shevell MI, Majnemer A, Rosenbaum P, Abrahamowicz M. Etiologic yield human brain at stage 9. Anat Embryol 1983;168:419–432.
of subspecialists’ evaluation of young children with global developmental 19. Müller F, O’Rahilly R. The first appearance of the neural tube and optic pri-
delay. J Pediatr 2000;136:593–598. mordium in the human embryo at stage 10. Anat Embryol 1985;172:157–
3. Soto-Ares G, YJoyes B, Lemaitre M, Vallee L, Pruvo J. MRI in children with 169.
mental retardation. Pediatr Radiol 2003;33:334–345. 20. Kibar Z, Capra V, Gros P. Toward understanding the genetic basis of neural
4. Norman MG, McGillivray BC, Kalousek DK, Hill A, Poskitt KJ. Congenital tube defects. Clin Genet 2007;71:295–310.
Malformations Of The Brain: Pathologic, Embryologic, Clinical, Radiologic 21. ten Donkelaar HJ, Lammens M, Cruysberg JRM, Hori A, Shiota K, Verbist
And Genetic Aspects. Oxford: Oxford University Press, 1995. B. Development and developmental disorders of the forebrain. In: ten

Barkovich_Chap05.indd 544 5/6/2011 9:01:34 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 545

Donkelaar HJ, Lammens M, Hori A, eds. Clinical Neuroembryology. Berlin: 46. Hankin MH, Schneider BF, Silver J. Death of the subcallosal glial sling is
Springer, 2006:345–428. correlated with formation of the cavum septi pellucidi. J Comp Neurol
22. Müller F, O’Rahilly R. The development of the human brain and the clo- 1988;272:191–202.
sure of the rostral neuropore at stage 11. Anat Embryol 1986;175:205–222. 47. Richards LJ. Axonal pathfinding mechanisms at the cortical midline and
23. Müller F, O’Rahilly R. The development of the human brain, the closure of in the development of the corpus callosum. Braz J Med Biol Res 2002;35:
the caudal neuropore, and the beginning of secondary neurulation at stage 1431–1439.
12. Anat Embryol 1987;176:413–430. 48. Shu T, Richards L. Cortical axon guidance by the glial wedge during the
24. Müller F, O’Rahilly R. The first appearance of the future cerebral hemi- development of the corpus callosum. J Neurosci 2001;21:2749–2758.
spheres in the human embryo at stage 14. Anat Embryol 1988;177: 49. Shu T, Li Y, Keller A, Richards LJ. The glial sling is a migratory population
495–511. of developing neurons. Development 2003;130:2929–2937.
25. Sidman RL, Rakic P. Development of the human central nervous system. 50. Tessier-Lavigne M, Goodman CS. The molecular biology of axon guidance.
In: Haymaker W, Adams RD, eds. Histology and Histopathology of the Ner- Science 1996;274:1123–1133.
vous System. Springfield: Thomas, 1982:73. 51. Wang L-C, Rachel RA, Marcus RC, Mason CA. Chemosuppression of reti-
26. Rakic P. Cell migration and neuronal ectopias in the brain. Birth Defects nal axon growth by the mouse optic chiasm. Neuron 1996;17:849–862.
1975;11:95–129. 52. Zaki W. Le processus dégénératif au cours du développement du corps cal-
27. Bystron I, Blakemore C, Rakic P. Development of the human cerebral cor- leux. Archiv Anat micr Morphol expér 1985;74:133–149.
tex: Boulder Committee revisited. Nat Rev Neurosci 2008;9:110–122. 53. Ren T, Anderson A, Shen W-B, et al. Imaging, anatomical, and molecular
28. Petanjek Z, Dujmovié A, Kostovié I, Esclapez M. Distinct origin of GABA- analysis of callosal formation in the developing human fetal brain. Anat
ergic neurons in forebrain of man, nonhuman primates and lower mam- Rec A 2006;288A:191–204.
mals. Coll Antropol 2008;32(Suppl 1):9–17. 54. Koester SE, O’Leary DD. Axons of early generated neurons in cingulate cor-
29. Noctor SC, Martinez-Cerdeno V, Ivic L, Kriegstein AR. Cortical neurons tex pioneer the corpus callosum. J Neurosci 1994;14:6608–6620.
arise in symmetric and asymmetric division zones and migrate through 55. Rash BG, Richards LJ. A role for cingulate pioneering axons in the develop-
specific phases. Nat Neurosci 2004;7(2):136–144. ment of the corpus callosum. J Comp Neurol 2001;434:147–157.
30. Glass HC, Shaw GM, Ma C, Sherr EH. Agenesis of the corpus callosum 56. Barkovich AJ, Lyon G, Evrard P. Formation, maturation, and disorders of
in California 1983–2003: a population-based study. Am J Med Genet A white matter. Am J Neuroradiol 1992;13:447–461.
2008;146A:2495–2500. 57. Barkovich AJ, Norman D. Anomalies of the corpus callosum: correlation
31. Bedeschi MF, Bonaglia MC, Grasso R, et al. Agenesis of the corpus cal- with further anomalies of the brain. Am J Neuroradiol 1988;9:493–501.
losum: clinical and genetic study in 63 young patients. Pediatr Neurol 58. Kier EL, Truwit CL. The lamina rostralis: modification of concepts con-
2006;34:186–193. cerning the anatomy, embryology and MR appearance of the rostrum of
32. Raybaud C, Levrier O, Brunel H, Girard N, Farnarier P. MR imaging of fetal the corpus callosum. Am J Neuroradiol 1997;18:715–722.
brain malformations. Childs Nerv Syst 2003;19:455–470. 59. Kier EL, Truwit CL. The normal and abnormal genu of the corpus cal-
33. Moutard M-L, Kieffer V, Feingold J, et al. Agenesis of corpus callosum: losum: an evolutionary, embryologic, anatomic, and MR analysis. Am J
prenatal diagnosis and prognosis. Childs Nerv Syst 2003;19:471–476. Neuroradiol 1996;17:1631–1641.
34. Aboitiz F, Montiel J. One hundred million years of interhemispheric 60. Déjerine J. Anatomie des Centres Nerveux, Vol 1. Paris: Rueff, 1895.
communication: the history of the corpus callosum. Braz J Med Biol Res 61. Bayer SA, Altman J. Volume 3: The Human Brain During The Second Trimes-
2003;36:409–420. ter. Boca Raton, London, New York: CRC Press, 2005.
35. Rakic P, Yakovlev PI. Development of the corpus callosum and cavum sep- 62. Larroche JC, Baudey J. Cavum septi pellucidi, cavum Vergae, cavum veli
tae in man. J Comp Neurol 1968;132:45–72. interpositi: cavités de la ligne médiane. Etude anatomique et pneumoen-
36. Lamantia AS, Rakic P. Cytological and quantitative characteristics of four céphalographique dans la période néonatale. Biol Neonate 1961;3:193–236.
cerebral commissures in the rhesus monkey. J Comp Neurol 1990;291: 63. Barkovich AJ. Apparent atypical callosal dysgenesis: analysis of MR find-
520–537. ings in six cases and their relationship to holoprosencephaly. Am J Neuro-
37. Liss L, Mervis L. The ependymal lining of the cavum septi pellucidi: a histo- radiol 1990;11:333–340.
logical and histochemical study. J Neuropath Exp Neurol 1964;23:355–367. 64. Raybaud CA, Girard N. Étude anatomique par IRM des agénésies et dys-
38. Raybaud C. Corpus callosum, other great cerebral commissures, septum plasies commissurales télencéphaliques. Neurochirurgie 1998;44(Suppl
pellucidum: anatomy, development and malformation. Neuroradiology 1):38–60.
2010;52:447–477. 65. Oba H, Barkovich AJ. A statistical analysis of callosal formation in holo-
39. Lent R, Uziel D, Baudrimont M, Fallet C. Cellular and molecular tunnels prosencephaly. Am J Neuroradiol 1995;16:453–460.
surrounding the forebrain commissures in human fetuses. J Comp Neurol 66. Barkovich AJ, Quint D. Middle interhemispheric fusion: an unusual variant
2005;483:375–382. of holoprosencephaly. Am J Neuroradiol 1993;14:431–440.
40. Livy DJ, Wahlsten D. Retarded formation of the hippocampal commissure 67. Cheng X, Hsu C-M, Currle DS, Hu JS, Barkovich AJ, Monuki ES. Central
in embryos from mouse strain lacking a corpus callosum. Hippocampus roles of the roof plate in telencephalic development and holoprosenceph-
1997;7:2–14. aly. J Neurosci 2006;26:7640–7649.
41. Hankin MH, Silver J. Development of intersecting CNS fiber tracts: 68. Palmini A, Andermann F, Olivier A, Tampieri D, Robitaille Y. Focal neu-
the corpus callosum and its perforating fiber pathway. J Comp Neurol ronal migration disorders and intractable partial epilepsy: results of surgi-
1988;272:177–190. cal treatment. Ann Neurol 1991;30:750–757.
42. Richards LJ, Plachez C, Ren T. Mechanisms regulating the development 69. Petrucci RJ, Buchheit WA, Woodruff GC, Karian JM, DeFilipp GJ. Transcal-
of the corpus callosum and its agenesis in mouse and human. Clin Genet losal parafornicial approach for third ventricle tumors: neuropsychological
2004;66:276–289. consequences. Neurosurgery 1987;20:457–464.
43. Shu T, Puche AC, Richards LJ. Development of midline glial populations at 70. Raybaud C, Girard N. The developmental disorders of the commissural
the corticoseptal boundary. J Neurobiol 2003;57:81–94. plate of the telencephalon: MR imaging study and morphologic classifica-
44. Shu T, Shen WB, Richards LJ. Development of the perforating pathway: tion. Nerv Syst Child 1999;24:348–357.
an ipsilaterally projecting pathway between the medial septum/diagonal 71. Hetts SW, Sherr EH, Chao S, Gobuty S, Barkovich AJ. Anomalies of the
band of Broca and the cingulate cortex that intersects the corpus callosum. corpus callosum: an MR Analysis of the phenotypic spectrum of associated
J Comp Neurol 2001;436:411–422. malformations. Am J Roentgenol 2006;187:1343–1348.
45. Silver J, Lorenz SE, Wahlsten D, Coughlin J. Axonal guidance during devel- 72. Probst FP. Agenesis of the corpus callosum. Acta Radiologica 1973;(Suppl
opment of the great cerebral commissures: descriptive and experimental 331):1–150.
studies, in vivo, on the role of preformed glial pathways. J Comp Neurol 73. Barkovich AJ, Kjos BO. Normal postnatal development of the corpus callo-
1982;210:10–29. sum as demonstrated by MR imaging. Am J Neuroradiol 1988;9:487–491.

Barkovich_Chap05.indd 545 5/6/2011 9:01:35 PM


546 Pediatric Neuroimaging

74. Parrish ML, Roessmann U, Levinsohn MW. Agenesis of the corpus 100. Nakata Y, Barkovich AJ, Wahl M, et al. Diffusion abnormalities and
callosum: a study of the frequency of associated malformations. Ann Neurol reduced volume of the ventral cingulum bundle in agenesis of the corpus
1979;6:349–354. callosum: a 3T imaging study. Am J Neuroradiol 2009;30:1142–1148.
75. Kendall BE. Dysgenesis of the corpus callosum. Neuroradiology 1983;25: 101. Baker LL, Barkovich AJ. The large temporal horn: MR analysis in devel-
239–256. opmental brain anomalies versus hydrocephalus. Am J Neuroradiol
76. Byrd S, Radkowski M, Falnnery A, McLone D. The clinical and radiologic 1992;13:115–122.
evaluation of absence of the corpus callosum. Eur J Radiol 1990;10:65–73. 102. Atlas SW, Zimmerman RA, Bilaniuk LT, et al. Corpus callosum and limbic
77. Ettlinger G. Agenesis of the corpus callosum. In: Vinicken PJ, Bruyn GW, system: neuroanatomic MR evaluation of developmental anomalies. Radi-
eds. Handbook of Clinical Neurology, Vol 30. Amsterdam: North Holland, ology 1986;160:355–362.
1977:285–297. 103. Barkovich AJ, Simon EM, Walsh CA. Callosal agenesis with cyst: a better
78. Parraga HC, Parrage MI, Jensen AR. Cognitive, behavioral, and psychiat- understanding and new classification. Neurology 2001;56:220–227.
ric symptoms in two children with agenesis of the corpus callosum: case 104. Pavone P, Barone R, Baieli S, Parano E, Incorpora G, Ruggieri M. Callosal
report. Int J Psychiatry 2003;33:107–113. anomalies with interhemispheric cyst: expanding the phenotype. Acta
79. Nielsen T, Montplaisir J, Lassonde M. Sleep architecture in agenesis of Paediatrica 2005;94:1066.
the corpus callosum: laboratory assessment of four cases. J Sleep Res 105. Pierre-Kahn A, Hanlo P, Sonigo P, Parisot D, McConnell RS. The contribu-
1992;1:197–200. tion of prenatal diagnosis to the understanding of malformative intracra-
80. Paul LK, Van Lancker-Sidtis D, Schieffer B, Dietrich R, Brown WS. Com- nial cysts: state of the art. Childs Nerv Syst 2000;16(10–11):619–626.
municative deficits in agenesis of the corpus callosum: nonliteral language 106. Smith AS, Levine D. Appearance of an interhemispheric cyst associ-
and affective prosody. Brain Lang 2003;85:313–324. ated with agenesis of the corpus callosum. Am J Neuroradiol 2004;25:
81. Hines RJ, Paul LK, Brown WS. Spatial attention in agenesis of the cor- 1037–1040.
pus callosum: shifting attention between visual fields. Neuropsychologia 107. Lena G, van Calenberg F, Genitori L, Choux M. Supratentorial interhemi-
2002;40:1804–1814. spheric cysts associated with callosal agenesis: surgical treatment and out-
82. Aicardi J, Lefebre J, Lerrique-Koechlin A. A new syndrome: spasm in flex- come in 16 children. Childs Nerv Syst 1995;11:568–573.
ion, callosal agenesis, ocular abnormalities. Electroencephalogr Clin Neuro- 108. Glenn OA, Goldstein RB, Li KC, et al. Fetal magnetic resonance imaging
physiol 1965;19:609–610. in the evaluation of fetuses referred for sonographically suspected abnor-
83. Pappas CTE, Rekate HL. Cervicomedullary junction decompression in a malities of the corpus callosum. J Ultrasound Med 2005;24:791–804.
case of Marshall-Smith syndrome. J Neurosurg 1991;75:317–319. 109. de Ercole C, Girard N, Cravello L, et al. Prenatal diagnosis of fetal corpus
84. Sztriha L, Espinosa-Parilla Y, Gururaj A, et al. Frameshift mutation of the callosum agenesis by ultrasonography and magnetic resonance imaging.
zinc finger homeobox 1B gene in syndromic corpus callosum agenesis Prenatal Diag 1998;18(3):247–253.
(Mowat-Wilson syndrome). Neuropediatrics 2003;34:322–325. 110. Tang PH, Bartha AI, Norton ME, Barkovich AJ, Sherr EH, Glenn OA.
85. Aicardi J. Aicardi syndrome. Brain Dev 2005;27:164–171. Agenesis of the corpus callosum: an MR imaging analysis of associated
86. Palmér L, Zetterlund B, Hård A-L, Steneryd K, Kyllerman M. Aicardi Syn- abnormalities in the fetus. Am J Neuroradiol 2009;30:257–263.
drome: presenatation at onset in Swedish children born in 1975–2002. 111. Volpe P, Paladini D, Resta M, et al. Characteristics, associations and out-
Neuropediatrics 2006;37:154–158. come of partial agenesis of the corpus callosum in the fetus. Ultrasound
87. Rosser T, Acosta M, Racker R. Aicardi syndrome: spectrum of disease and Obstet Gynecol 2006;27(5):509–516.
long-term prognosis in 77 females. Pediatr Neurol 2002;27:343–346. 112. Kuzniecky R, Murro A, King D, et al. Magnetic resonance imaging in
88. Baierl P, Markl A, Thelen M, Laub MC. MR imaging of Aicardi syndrome. childhood intractable partial epilepsy: pathologic correlations. Neurology
Am J Neuroradiol 1988;9:805–806. 1993;43:681–687.
89. Hall-Craggs MA, Harbord MG, Finn JP, Brett E, Kendall B. Aicardi syn- 113. Wyllie E, Comair Y, Kotagal P, Bulacio J, Bingaman W, Ruggieri P. Seizure
drome: MR assessment of brain structure and myelination. Am J Neurora- outcome after epilepsy surgery in children and adolescents. Ann Neurol
diol 1990;11:532–536. 1998;44:740–748.
90. Igidbashian V, Mahboubi S, Zimmerman RA. CT and MR findings in 114. Farrell MA, DeRosa M, Curran J, et al. Neuropathologic findings in corti-
Aicardi syndrome. J Comput Assist Tomogr 1987;11:357–358. cal resections (including hemispherectomies) performed for the treatment
91. Swayze II VW, Johnson VP, Hanson JW, et al. Magnetic resonance imaging of intractable childhood epilepsy. Acta Neuropathol 1992;83:246–259.
of brain anomalies in fetal alcohol syndrome. Pediatrics 1997;99:232–240. 115. Frater J, Prayson R, Morris H, III, Bingaman W. Surgical pathologic find-
92. Bodensteiner JB. The saga of the septum pellucidum: a tale of unfunded ings of extratemporal-based intractable epilepsy: a study of 133 consecu-
clinical investigations. J Child Neurol 1995;10:227–231. tive resections. Arch Pathol Lab Med 2000;124:545–549.
93. Guion-Almeida ML, Richieri-Costa A, Saavedra D, Cohen Jr MM. Fronto- 116. Pasquier S, Peoc’h M, Fabre-Bocquentin B, et al. Surgical pathology of
nasal dysplasia: analysis of 21 cases and literature review. Int J Oral Maxil- drug-resistant partial epilepsy. A 10-year-experience with a series of 327
lofac Surg 1996;25:91–97. consecutive resections. Epileptic Disord 2002;4:99–119.
94. Mowat DR, Wilson MJ, Goossens M. Mowat-Wilson syndrome. J Med 117. Tassi L, Colombo N, Francione S, et al. Focal cortical dysplasia: neuro-
Genet 2003;40:305–310. pathological subtypes, EEG, neuroimaging and surgical outcome. Brain
95. Ostrovaskaya TI, Lazjuk GI. Cerebral abnormalities in the Neu-Laxova syn- 2002;125(Pt 8):1719–1732.
drome. Am J Med Genet 1988;30:747–756. 118. Fauser S, Schulze-Bonhage A, Honegger J, et al. Focal cortical dysplasias:
96. Guion-Almeida ML, Richieri-Costa A. Callosal agenesis, iris coloboma, and surgical outcome in 67 patients in relation to histological subtypes and
megacolon in a Brazilian boy with Rubinstein-Taybi syndrome. Am J Med dual pathology. Brain 2004;127:2406–2418.
Genet 1992;43:929–931. 119. Alonso-Nanclares L, Garbelli R, Sola RG, et al. Microanatomy of the dys-
97. Segeren CM, Polderman KH, Lips P. Agenesis of the corpus callosum asso- plastic neocortex from epileptic patients. Brain 2005;128(Pt 1):158–173.
ciated with paroxysmal hypothermia: Shapiro’s syndrome. Neth J Med 120. Fauser S, Huppertz H-J, Bast T, et al. Clinical characteristics in focal corti-
1997;50(1):29–35. cal dysplasia: a retrospective evaluation in a series of 120 patients. Brain
98. Tovar-Moll F, Moll J, de Oliveira-Souza R, Bramati I, Andreiuolo PA, Lent R. 2006:129:1907–1916.
Neuroplasticity in human callosal dysgenesis: a diffusion tensor imaging 121. Blümcke I, Vinters HV, Armstrong D, Aronica E, Thom M, Spreafico R.
study. Cereb Cortex 2007;17:531–541. Malformations of cortical development and epilepsies: neuropathologi-
99. Wahl M, Strominger Z, Jeremy RJ, et al. Variability of homotopic and hetero- cal findings with emphasis on focal cortical dysplasia. Epileptic Disord
topic callosal connectivity in partial agenesis of the corpus callosum: a 3T 2009;11:181–193.
diffusion tensor imaging and Q-ball tractography study. Am J Neuroradiol 122. Dobyns WB, Andermann E, Andermann F, et al. X-linked malformations
2009;30:282–289. of neuronal migration. Neurology 1996;47:331–339.

Barkovich_Chap05.indd 546 5/6/2011 9:01:35 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 547

123. Brunelli S, Faiella A, Capra V, et al. Germline mutations in the homeobox 148. Hevner RF. From radial glia to pyramidal-projection neuron: transcription
gene EMX2 in patients with severe schizencephaly. Nat Genet 1996;12: factor cascades in cerebral cortex development. Mol Neurobiol 2006;33:
94–96. 33–50.
124. Dobyns WB, Truwit CL. Lissencephaly and other malformations of corti- 149. Heng JI, Nguyen L, Castro D, et al. Neurogenin 2 controls cortical neuron
cal development. Neuropediatrics 1995;26:132–147. migration through regulation of Rnd2. Nature 2008;455:114–118.
125. Manzini MC, Gleason D, Chang B, et al. Ethnically diverse causes of 150. Cholfin JA, Rubenstein JL. Patternin of frontal cortex subdivisions by
Walker-Warburg syndrome (WWS): FCMD mutations are a more com- Fgf17. Proc Natl Acad Sci USA 2007;104:7652–7657.
mon cause of WWS outside of the Middle East. Hum Mutat 2008;29: 151. Cholfin JA, Rubenstein JL. Frontal cortex subdivision patterning is coor-
E231–E241. dinately regulated by Fgf8, Fgf17, and Emx2. J Comp Neurol 2008;509:
126. Gleeson JG, Minnerath S, Kuzniecky RI, et al. Somatic and germline 144–155.
mosaic mutations in the doublecortin gene are associated with variable 152. Rakic P. Evolution of the neocortex: a perspective from developmental
phenotypes. Am J Hum Genet 2000;67:574–581. biology. Nat Rev Neurosci 2009;10:724–745.
127. Sheen V, Wheless J, Bodell A, et al. Periventricular heterotopia associated 153. Gaballo A, Palma M, Dicuonzo F, Carella A. Lhermitte-Duclos disease: MR
with chromosome 5p anomalies. Neurology 2003;60:1033–1036. diffusion and spectroscopy. Radiol Med (Torino) 2005;110(4):378–384.
128. Piao X, Chang BS, Bodell A, et al. Genotype-phenotype analysis of human 154. Flames N, Pla R, Gelman DM, Rubenstein JL, Puelles L, Marin O. Delin-
frontoparietal polymicrogyria syndromes. Ann Neurol 2005;58(5):680– eation of multiople subpallial progenitor domains by the combinatorial
687. expression of transcriptional codes. J Neurosci 2007;27:9682–9695.
129. Parrini E, Ramazzotti A, Dobyns WB, et al. Periventricular heterotopia: 155. Liodis P, Denaxa M, Grigoriou M, Akufo-Addo C, Yanagawa Y, Pachnis V.
phenotypic heterogeneity and correlation with Filamin A mutations. Lhx6 activity is required for the normal migration and specification of
Brain 2006;129:1892–1906. cortical interneuron subtypes. J Neurosci 2007;27:3078–3089.
130. Chang BS, Apse KA, Caraballo R, et al. A famillial syndrome of unilat- 156. Fogarty M, Grist M, Gelman D, Marin O, Pachnis V, Kessaris N. Spatial
eral polymicrogyria affecting the right hemisphere. Neurology 2006;66: genetic patterning of the embryonic neuroepithelium generates
133–135. GABAergic interneuron diversity in the adult cortex. J Neurosci 2007;27:
131. Poirier K, Keays DA, Francis F, et al. Large spectrum of lissencephaly and 10935–10946.
pachygyria phenotypes resulting from de novo missense mutations in 157. Cobos I, Long JE, Thwin MT, Rubenstein JL. Cellular patterns of tran-
tubulin alpha 1A (TUBA1A). Hum Mutat 2007;28(11):1055–1064. scription factor expression in developing cortical interneurons. Cereb Cor-
132. Cardoso C, Boys A, Parrini E, et al. Periventricular heterotopia, mental tex 2006;16(Suppl_1):i82–i88.
retardation, and epilepsy associated with 5q14.3-q15 deletion. Neurology 158. Markram H, Toledo-Rodriguez M, Wang Y, Gupta A, Silberberg G, Wu C.
2009;72:784–792. Interneurons of the neocortical inhibitory system. Nat Rev Neurosci
133. Brazel CY, Romanko MJ, Rothstein RP, Levison SW. Roles of the mamma- 2004;5:793–807.
lian subventricular zone in brain development. Prog Neurobiol 2003;603: 159. Wonders CP, Anderson SA. The origin and specification of cortical
1–21. interneurons. Nat Rev Neurosci 2006;7:687–696.
134. Petanjek Z, Berger B, Esclapez M. Origins of cortical GABAergic neurons 160. Tucker ES, Segall S, Gopalakrishna D, et al. Molecular specification and
in the cynomolgus monkey. Cereb Cortex 2009;19:249–262. patterning of progenitor cells in the lateral and medial ganglionic emi-
135. Rakic P. Neuronal migration and contact guidance in the primate tel- nences. J Neurosci 2008;28:9504–9518.
encephalon. Postgrad Med J 1978;54:25–40. 161. Miyoshi G, Hjerling-Leffler J, Karayannis T, et al. Genetic fate mapping
136. Caviness VS, Jr. Normal development of the cerebral neocortex. In: Evrard P, reveals that the caudal ganglionic eminence produces a large and diverse
Minkowski A, eds. Developmental Neurobiology. New York: Raven, 1989: population of superficial cortical interneurons. J Neurosci 2010;30:1582–
1–10. 1594.
137. Pontious A, Kowalczyk T, Englund C, Hevner RF. Role of intermediate 162. Nobrega-Pereira S, Gelman D, Bartolini G, Pla R, Pierani A, Marin O.
progenitor cells in cerebral crotex development. Dev Neurosci 2008;30: Origin and molecular specification of globus pallidus neurons. J Neurosci
24–32. 2010;30:2824–2834.
138. Noctor SC, Flint AC, Weissman TA, Dammerman RS, Kriegstein AR. 163. Flandin P, Kimura S, Rubenstein JLR. The progenitor zone of the ven-
Neurons derived from radial glial cells establish radial units in neocortex. tral medial ganglionic eminence requires Nkx2–1 to generate most of
Nature 2001;409:714–720. the globus pallidus but few neocortical interneurons. J Neurosci 2010;30:
139. Miyata T, Kawaguchi A, Okano H, Ogawa M. Asymmetric inheritance of 2812–2823.
radial glial fibers by cortical neurons. Neuron 2001;31:727–741. 164. Xu Q, Cobos I, De La Cruz E, Rubenstein JLR, Anderson SA. Origins of
140. Malatesta P, Hack MA, Hartfuss E, et al. Neuronal or glial progeny: regional cortical interneuron subtypes. J Neurosci 2004;24:2612–2622.
differences in radial glia fate. Neuron 2003;37:751–764. 165. Tanaka DH, Maekawa K, Yanagawa Y, Obata K, Murakami F. Multidirec-
141. Malatesta P, Hartfuss E, Gotz M. Isolation of radial glial cells by fluo- tional and multizonal tangential migration of GABAergic interneurons in
rescent-activated cell sorting reveals a neuronal lineage. Development the developing cerebral cortex. Development 2006;133:2167–2176.
2000;127:5253–5263. 166. Stanco A, Szekeres C, Patel N, et al. Netrin-1-alpha-3-beta-1 integrin
142. Heins N, Malatesta P, Cecconi F, et al. Glial cells generate neurons: the role interactions regulate the migration of interneurons through the cortical
of the transcription factor Pax6. Nat Neurosci 2002;5:308–315. marginal zone. Proc Natl Acad Sci USA 2009;106:7595–7600.
143. Gressens P, Evrard P. The glial fascicle: an ontogenic and phylogenic unit 167. O’Rourke NA, Dailey ME, Smith SJ, McConnell SK. Diverse migratory
guiding, supplying and distributing mammalian cortical neurons. Brain pathways in the developing cerebral cortex. Science 1992;258:299–304.
Res Dev Brain Res 1993;76:272–277. 168. O’Rourke NA, Sullivan DP, Kaznowski CE, Jacobs AA, McConnell SK.
144. Marret S, Gressens P, Evrard P. Arrest of neuronal migration by excit- Tangential migration of neurons in the developing cerebral cortex. Devel-
atory amino acids in hamster developing brain. Proc Natl Acad Sci USA opment 1995;121:2165–2176.
1996;93:15463–15468. 169. O’Rourke NA, Chenn A, McConnell SK. Postmitotic neurons migrate
145. Komuro H, Rakic P. Intracellular Ca++ fluctuations modulate the rate of tangentially in the cortical ventricular zone. Development 1997;124:
neuronal migration. Neuron 1996;17:275–285. 997–1005.
146. Rakic P. The radial edifice of cortical architecture: from neuronal silhou- 170. Letinic K, Zoncu R, Rakic P. Origin of GABAergic neurons in the human
ettes to genetic engineering. Brain Res Rev 2007;55:204–219. neocortex. Nature 2002;417:645–649.
147. Rakic P, Hashimoto-Torii K, Sarkisian MR. Genetic determinants of neu- 171. Anderson SA, Eisenstat DD, Shi L, Rubenstein JLR. Interneuron migra-
ronal migration in the cerebral cortex. Novartis Found Symp 2007;288: tion from basal forebrain to neocortex: dependence on Dlx genes. Science
45–53. 1997;278:474–476.

Barkovich_Chap05.indd 547 5/6/2011 9:01:35 PM


548 Pediatric Neuroimaging

172. Marín O, Anderson SA, Ruberstein JLR. Origin and molecular specification the subventricular zone in the development of cortical dysplasia. J Child
of striatal interneurons. J Neurosci 2000;20:6063–6076. Neurol 2009;24:275–286.
173. Kriegstein AR, Noctor SC. Patterns of neuronal migration in the embry- 198. Pombero A, Valdes L, Vieira C, Martinez S. Developmental mechanisms
onic cortex. Trends Neurosci 2004;27:392–399. and experimental models to understand forebrain malformative diseases.
174. Noctor SC, Flint AC, Weissman TA, Wong WS, Clinton BK, Kriegstein Genes Brain Behav 2007;6(Suppl 1):45–52.
AR. Dividing precursor cells of the embryonic cortical ventricular zone 199. Hennekam RCM, Barth PG. Syndromic cortical dysplasias: a review. In:
have morphological and molecular characteristics of radial glia. J Neurosci Barth PG, ed. Disorders of Neuronal Migration. London: MacKeith Press,
2002;22:3161–3173. 2003:135–169.
175. Marsh E, Fulp C, Gomez E, et al. Targeted loss of Arx results in a develop- 200. Dobyns WB, Mirzaa G, Christian SL, et al. Consistent chromosome
mental epilepsy mouse model and recapitulates the human phenotype in abnormalities identify novel polymicrogyria loci in 1p36.3, 2p16.1 –
heterozygous females. Brain 2009;132:1563–1576. p23.1, 4q21.21 – q22.1, 6q26 – q27, and 21q2. Am J Med Genet Part A
176. McConnell S, Kaznowski C. Cell cycle dependence of laminar determina- 2008;146A:1637–1654.
tion in the developing cerebral cortex. Science 1991;254:282–285. 201. Ramer J, Lin A, Dobyns W, et al. Previously apparently undescribed
177. Bohner AP, Akers RM, McConnell SK. Induction of deep layer cortical syndrome: shallow orbits, ptosis, coloboma, trigoncephaly, gyral mal-
neurons in vitro. Development 1997;124:915–923. formations, and mental and growth retardation. Am J Med Genet
178. McConnell SK. Constructing the cerebral cortex: neurogenesis and fate 1995;57:403–409.
determination. Neuron 1995;15:761–768. 202. Tranebjaerg L, Baekmark U, Dyhr-nielsen M, Kreiborg S. Partial tri-
179. Meyer G, Goffinet A. Prenatal development of reelin-immunoreactive somy 3q syndrome inherited from familial t(3;9)9q26.1;23). Clin Genet
neurons in the human neocortex. J Comp Neurol 1998;397:29–40. 1987;32:137–143.
180. Ogawa M, Miyata T, Nakajima K, et al. The reeler gene associated antigen 203. Tupler R, Maraschio P, Gerardo A, Mainieri R, Lanzi G, Tiepolo L.
on Cajal-Retzius neurons is a crucial molecule for laminal organization of A complex chromosome rearrangement with 10 breakpoints: tentative
cortical neurons. Neuron 1996;14:899–912. assignment of the locus for Williams syndrome to 4q33-q35. J Med Genet
181. D’Arcangelo G, Miao GG, Chen SC, Soares HD, Morgan JI, Curran T. 1992;29:253–255.
A protein related to extracellular matrix proteins deleted in the mouse 204. Federico A, Tomasetti P, Zollino M, et al. Association of trisomy 9p and
mutant reeler. Nature 1995;374:719–723. band heterotopia. Neurology 1999;53:430–432.
182. Hashimoto-Torii K, Torii M, Sarkisian MR, et al. Interaction between ree- 205. Golden J, Schoene W. Central nervous system malformations in Trisomy
lin and notch signaling regulates neuronal migration in the cerebral cor- 9. J Neuropathol Exp Neurol 1993;52:71–77.
tex. Neuron 2009;60:273–284. 206. Taylor A. Autosomal trisomy syndromes: a detailed study of 27 cases
183. Shatz CJ, Chun LLM, Luskin MB. The role of the subplate in the devel- of Edwards syndrome and 27 cases of Patau syndrome. J Med Genet
opment of the mammalian telencephalon. In: Peters A, Jones EG, eds. 1968;5:227–252.
Cerebral Cortex: Development and Maturation of Cerebral Cortex, Vol 7. 207. Allanson J, Ledbetter D, Donyns W. Classical lissencephaly syndromes:
New York: Plenum, 1988:35–58. does the face reflect the brain? J Med Genet 1998;35:920–923.
184. Ghosh A, Shatz CJ. A role for subplate neurons in the patterning of con- 208. Teysier M, Charrin C, Corgiola Theuil G. Ring chromosome 17. Case
nections from thalamus to neocortex. Development 1993;117:1031–1047. report and review of the literature. Ann Genet 1992;35:75–78.
185. Ghosh A, Antonini A, McConnell SK, Shatz CJ. Requirement for sub- 209. Bingham PM, Lynch D, McDonald-McGinn D, Zackai E. Polymicrogyria
plate neurons in the formation of thalamocortical connections. Nature in chromosome 22 deletion syndrome. Neurology 1998;51:1500–1502.
1990;347:179–181. 210. Ghariani S, Dahan K, Saint-Martin C, et al. Polymicrogyria in chromo-
186. McConnell SK, Ghosh A, Shatz CJ. Subplate neurons pioneer the first axon some 22q11 deletion syndrome. Eur J Pediatr Neurol 2002;6:73–77.
pathway from the cerebral cortex. Science 1989;145:278–281. 211. Robin N, Taylor C, McDonald-McGinn D, et al. Polymicrogyria and dele-
187. McConnell SK, Ghosh A, Shatz CJ. Subplate pioneers and the forma- tion 22q11.2 syndrome: window to the etiology of a common cortical
tion of descending connections from cerebral cortex. J Neurosci 1994;14: malformation. Am J Med Genet A 2006;140:2416–2425.
107–123. 212. Worthington S, Turner A, Elber J, Andrews PI. 22q11 deletion and polymi-
188. Marin-Padilla M. Structural organization of the human cerebral cortex crogyria – cause or coincidence? Clin Dysmorphol 2000;9:193–197.
prior to the appearance of the cortical plate. Anat Embryol 1983;168: 213. Kiehl TR, Chow EW, Mikulis DJ, George SR, Bassett AS. Neuropatho-
21–40. logic features in adults with 22q11.2 deletion syndrome. Cereb Cortex
189. Marin-Padilla M. Early ontogenesis of the human cerebral cortex. In: 2009;19:153–164.
Peter A, Jones EG, eds. Cerebral Cortex: Development and Maturation of 214. France N, Crome L, Abraham J. Pathological features in the De Lange syn-
the Cerebral Cortex, Vol 7. New York: Plenum, 1988:1–34. drome. Acta Paediatr Scand 1969;58:470–480.
190. Barkovich AJ, Gressens P, Evrard P. Formation, maturation, and disorders 215. Schaefer G, Bodensteiner J, Beuehler B, Lin A, Cole T. The neuroimaging
of brain neocortex. Am J Neuroradiol 1992;13:423–446. findings in Sotos syndrome. Am J Med Genet 1997;68:462–465.
191. Suzuki M, Choi BH. Repair and reconstruction of the cortical plate fol- 216. Aubourg P, Scotto J, Rocchiccioli F, Feldmann-Pautrat D, Robain O. Neo-
lowing closed cryogenic injury to the neonatal rat cerebrum. Acta Neuro- natal adrenoleukodystrophy. J Neurol Neurosurg Psychiatry 1986;49:77–86.
pathol 1991;82:93–101. 217. Kelley RI, Datta NS, Dobyns WB, et al. Neonatal adrenoleukodystro-
192. O’Leary DD, Schlaggar BL, Tuttle R. Specification of neocortical areas and phy: new cases, biochemical studies, and differentiation from Zellweger
thalamocortical connections. Annu Rev Neurosci 1994;17:419–439. and related peroxisomal polydystrophy syndromes. Am J Med Genet
193. Ayoub AE, Kostovi I. New horizons for the subplate zone and its pioneer- 1986;23:869–901.
ing neurons. Cereb Cortex 2009;19:1705–1707. 218. Kaufmann W, Theda C, Naidu S, Watkins P, Moser A, Moser H. Neuronal
194. McQuillen PS, Ferriero DM. Perinatal subplate neuron injury: impli- migration abnormality in peroxisomal bifunctional enzyme defect. Ann
cations for cortical development and plasticity. Brain Pathol 2005;15: Neurol 1996;39:268–271.
250–260. 219. Watkins P, McGuinnes M, Raymond G, et al. Distinction between peroxi-
195. Guerrini R, Marini C. Genetic malformations of cortical development. somal bifunctional enzyme and acyl-CoA oxidase deficiencies. Ann Neurol
Exp Brain Res 2006;173:322–333. 1995;38:472–477.
196. Marret S, Mukendi R, Gadisseux JF, Gressens P, Evrard P. Effect of ibo- 220. Samsom J, Barth P, de Vries J, et al. Familial mitochondrial encephalopathy
tenate on brain development: an excitotoxic mouse model of micro- with fetal ultrasonographic ventriculomegaly and intracerebral calcifica-
gyria and posthypoxic-like lesions. J Neuropathol Exp Neurol 1995;54: tions. Eur J Pediatr 1994;153:510–516.
358–370. 221. Huizing M, Ruitenbeek W, Thinnes F, et al. Deficiency of the voltage-
197. Sawai C, Takano T, Takeuchi Y. Experimental neuronal migration disor- dependent anion channel: a novel cause of mitochondriopathy. Pediatr
ders following the administration of ibotenate in hamsters: the role of Res 1996;39:760–765.

Barkovich_Chap05.indd 548 5/6/2011 9:01:35 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 549

222. Böhm N, Uy J, Kiebling M, Lehnert W. Multiple acyl CoA dehydrogena- 246. Pascual-Castroviejo I, Pascual-Pascual SI, Velazquez-Fragua R, Lapunzina P.
tion deficiency (glutaric aciduria type II), congenital polycystic kidneys, Oculocerebrocutaneous (Delleman) syndrome: report of two cases.
and symmetric warty degeneration of the cerebral cortex in two newborn Neuropediatrics 2005;36:50–54.
brothers. II. Morphology and pathogenesis. Eur J Pediatr 1982;139:60–65. 247. Moog U, Jones MC, Bird LM, Dobyns WB. Oculocerebrocutaneous syn-
223. Gellera C, Uziel G, Rimoldi M, et al. Fumarase deficiency is an autosomal drome: the brain malformation defines a core phenotype. J Med Genet
recessive encephalopathy affecting both the mitochondrial and the cyto- 2005;42:913–921.
solic enzyme. Neurology 1990;40:495–499. 248. Landy S, Donnai D. Incontinentia pigmenti (Bloch-Sulzberger syndrome).
224. Kerrigan J, Aleck K, Tarby T, Bird C, Heidenreich R. Fumaric aciduria: J Med Genet 1993;30:53–59.
clinical and imaging features. Ann Neurol 2000;47:583–588. 249. Pascual-Castroviejo I, Roche M, Martinez-Fernanez V. Incontinen-
225. Chitayat D, Meagher-Villemure K, Mamer O, et al. Brain dysgenesis and tia pigmenti: MR demonstration of brain changes. Am J Neuroradiol
congenital intracerebral calcification associated with 3-hydroxy-isobutyric 1994;15:1521–1527.
aciduria. J Pediatr 1992;121:86–89. 250. Nellhaus G, Haberland C, Hill BJ. Sturge-Weber disease with bilateral
226. Van der Knaap M, Jakocs C, Hoffman G, et al. D-2-hydroxyglutaric aciduria: intracranial calcifications at birth and unusual pathologic findings. Acta
biochemical marker or clinical disease entity? Ann Neurol 1999;45:111–119. Neurol Scand 1967;43:314–347.
227. Geerdink N, Rotteveel J, Lammens M, et al. MECP2 mutation in a boy 251. Dhamecha RD, Edwards-Brown MK. Klippel-Trenaunay-Weber syndrome
with severe encephalopoathy: clinical, neuropathological and molecular with hemimegalencephaly. J Craniofac Surg 2001;12:194–196.
findings. Neuropediatrics 2002;33:33–36. 252. Hall BD, Cadle RG, Morrill-Cornelius SM, Bay CA. Phakomatosis pigmen-
228. Burn J, Wickramasinghe B, Harding B, Baraitser M. A syndrome with tovascularis: implications for severity with special reference to mongolian
intracranial calcification and microcephaly in two sibs, resembling intra- spots associated with Sturge-Weber and Klippel-Trenaunay syndromes.
uterine infection. Clin Genet 1986;30:112–116. Am J Med Genet A 2007;143A:3047–3053.
229. Barkovich AJ, Vandermarck P, Edwards MB, Cogen PH. Congenital 253. Lyon G, Raymond G, Mogami K, Gadisseux J-F, Guistina E. Disorder of
nasal masses: CT and MR imaging features in 16 cases. Am J Neuroradiol cerebellar foliation in Walker’s lissencephaly and Neu-Laxoca syndrome.
1991;12:105–116. J Neuropathol Exp Neurol 1993;52:633–639.
230. Takanashi J, Barkovich AJ, Clegg NJ, Delgado MA. Middle interhemi- 254. Shapiro I, Borochowitz Z, Degani S, Dar H, Ibschitz I, Sharf M. Neu-Laxova
spheric variant of holoprosencephaly associated with diffuse polymicro- syndrome: Prenatal ultrasonographic diagnosis, clinical and pathological
gyria. Am J Neuroradiol 2003;24:394–397. studies, and new manifestations. Am J Med Genet 1992;43:602–605.
231. Mizuguchi M, Maekawa S, Kamoshita S. Distribution of leptomeningeal 255. Andermann F. Bilateral focal polymicrogyria in Ehlers-Danlos syndrome.
glioneuronal heterotopia in alobar holoprosencephaly. Archiv Neurol Arch Neurol 2000;57:123–127.
1994;51:951–954. 256. Cupo L, Pyeritz R, Olson J, McPhee S, Hutchins G, McKusic V. Ehlers-
232. Hughes H, Harwood-Nash D, Becker L. Craniotelencephalic dysplasia Danlos syndrome with abnormal collagen fibrils, sinus of Valsalva
in sister: further delineation of a possible syndrome. Am J Med Genet aneurysms, myocardial infarction, panacinar emphysema and cerebral
1983;14:557–565. heterotopias. Am J Med Genet 1981;71:1051–1058.
233. Boltshauser E, Isler W. Joubert syndrome: episodic hyperpnea, abnormal 257. Van der Knaap MS, Smit LME, Barth PG, et al. Magnetic resonance imag-
eye movements, retardation and ataxia associated with dysplasia of the ing in classification of congenital muscular dystrophies with brain abnor-
cerebellar vermis. Neuropadiatrie 1977;8(1):57–66. malities. Ann Neurol 1997;42:50–59.
234. Dixon-Salazar T, Silhavy JL, Marsh SE, et al. Mutations in the AHI1 gene, 258. Taratuto AL, Lubieniecki F, Di’az D, et al. Merosin-deficient congenital
encoding jouberin, cause Joubert syndrome with cortical polymicrogyria. muscular dystrophy associated with abnormal cerebral cortical gyration:
Am J Hum Genet 2004;75(6):979–987. an autopsy study. Neuromuscul Disord 1999;9(2):86–94.
235. Zaki MS, Abdel-Aleem A, Abdel-Salam GMH, et al. The molar tooth sign: 259. Philpot J, Cowan F, Pennock J, et al. Merosin-deficient congenital muscu-
a new Joubert syndrome and related cerebellar disorders classification sys- lar dystrophy: the spectrum of brain involvement on magnetic resonance
tem tested in Egyptian families. Neurology 2008;70:556–565. imaging. Neuromuscul Disord 1999;9:81–85.
236. Rosman N, Pearce J. The brain in multiple neurofibromatosis (von Reck- 260. Barkovich AJ. Neuroimaging manifestations and classification of congeni-
linghausen’s disease): a suggested neuropathological bases for the associ- tal muscular dystrophies. Am J Neuroradiol 1998;19:1389–1396.
ated mental defect. Brain 1967;90:829–837. 261. Rosman N, Rebeiz J. The cerebral defect and myopathy in myotonic dys-
237. Mukonoweshuro W, Griffiths P, Blaser S. Neurofibromatosis type 1: the trophy. A comparative clinicopathological study. Neurology 1967;17:1106–
role of neuroradiology. Neuropediatrics 1999;30:111–119. 1111.
238. Baron Y, Barkovich AJ. MR imaging of tuberous sclerosis in neonates and 262. Clement E, Mercuri E, Godfrey C, et al. Brain involvement in muscu-
young infants. Am J Neuroradiol 1999;20:907–916. lar dystrophies with defective dystroglycan glycosylation. Ann Neurol
239. Braffman BH, Bilaniuk LT, Naidich TP, et al. MR imaging of tuberous 2008;64:573–582.
sclerosis: pathogenesis of this phakomatosis, use of gadopentate dimeglu- 263. Fontaine G, Farriaux J. Un nouveau syndrome polymalformatif complexe.
mine, and literature review. Radiology 1992;183:227–238. J Genet Hum 1977;25:109–119.
240. Christophe C, Bartholome J, Blum D, et al. Neonatal tuberous sclerosis. 264. Priolo M, De Toni T, Baffico M, et al. Fontaine-Farriaux craniosynostosis:
US, CT, and MR diagnosis of brain and cardiac lesions. Pediatr Radiol second report in the literature. Am J Med Genet 2001;100:214–218.
1989;19:446–448. 265. Emery S, Karpinsky N, Hansen L, Masliah E. Abnormalities in central ner-
241. Griffiths PD, Gardner S-A, Smith M, Rittey C, Powell T. Hemimegalen- vous system development in osteogenesis imperfecta type II. Pediatr Dev
cephaly and focal megalencephaly in tuberous sclerosis complex. Am J Pathol 1999;2:124–130.
Neuroradiol 1998;19:1935–1938. 266. Trifiletti R, Incorpori G, Polizzi A, Cocuzza M, Bolan E, Parano E. Aicardi
242. Fryburg J, Lin K, Matsumoto J. Abnormal head MRI in a neurologi- syndrome with multiple tumors: a case report with literature review. Brain
cally normal boy with hypomelanosis of Ito. Am J Med Genet 1996;66: Dev 1995;17:283–285.
200–203. 267. Donnenfeld A, Packer R, Zackai E, Chee C, Sellinger B, Emanuel B. Clini-
243. Ross D, Liwnicz B, Chun R, Gilbert E. Hypomelanosis of Ito (incontinen- cal, cytogenetic, and pedigree findings in 18 cases of Aicardi syndrome.
tia pigmenti achromians) - a clinicopathologic study: macrocephaly and Am J Med Genet 1989;32:461–467.
gray matter hetertopias. Neurology 1982;32:1013–1016. 268. Nassogne M, Henrot B, Saint-Martin C, Kadhim H, Dobyns W, Sebire G.
244. Haberland C, Perou M. Encephalocraniocutaneous lipomatosis: a new Polymicrogyria and motor neuropathy in Micro syndrome. Neuropediat-
example of ectomesodermal dysgenesis. Arch Nerv 1970;22:144–155. rics 2000;31:218–221.
245. Moog U, De Die-Smulders C, Systermans J, Cobben J. Oculocerebrocuta- 269. Graham JM Jr, Hennekam R, Dobyns WB, Roeder E, Busch D. MICRO
neous syndrome: report of three additional cases and aetiological consid- syndrome: an entity distinct from COFS syndrome. Am J Med Genet A
erations. Clin Genet 1997;52:219–225. 2004;128:235–245.

Barkovich_Chap05.indd 549 5/6/2011 9:01:35 PM


550 Pediatric Neuroimaging

270. Warburg M, Sjo O, Fledelius H, Pederson S. Autosomal recessive 293. Salamon N, Kung J, Shaw SJ, et al. FDG-PET/MRI coregistration improves
microcephaly, microcornea, congenital cataract, mental retardation, detection of cortical dysplasia in patients with epilepsy. Neurology
optic atrophy, and hypogenitalism. Micro syndrome. Am J Dis Child 2008;71:1594–1601.
1993;12:1309–1312. 294. Widjaja E, Mahmoodabadi SZ, Otsubo H, et al. Subcortical alterations
271. Kadhim HJ, Lammens M, Gosseye S, Gadisseux JF, Evrard P. Brain defects in tissue microstructure adjacent to focal cortical dysplasia: detection at
in infants with Potter syndrome (oligohydramnios sequence). Pediatr diffusion-tensor MR Imaging by using magnetoencephalographic dipole
Pathol 1993;13:519–536. cluster localization. Radiology 2009;251:206–215.
272. Towfighi J, Berlin C, Ladda R, Frauenhoffer E, Lehman R. Neuropathology 295. Kim J, Bai S, Choi K, et al. Comparison of various imaging modalities
of oral-facial-digital syndromes. Arch Pathol Lab Med 1985;109:642–646. in localization of epileptogenic lesion using epilepsy surgery outcome in
273. Toriello H. Orial-facial digital syndromes, 1992. Clin Dysmorphol pediatric patients. Seizure 2009;18:504–510.
1993;2:95–105. 296. Richardson MP, Koepp MJ, Brooks DJ, Duncan JS. 11C-flumazenil PET in
274. Doss BJ, Jolly S, Qureshi F, et al. Neuropathologic findings in a case of neocortical epilepsy. Neurology 1998;51:485–492.
OFDS type VI (Varadi syndrome). Am J Med Genet 1998;77:38–42. 297. Ryvlin P, Bouvard S, Le Bars D, et al. Clinical utility of flumazenil-PET ver-
275. Takanashi J, Tada H, Ozaki H, Barkovich AJ. Malformations of cerebral sus [18F]fluorodeoxyglucose-PET and MRI in refractory partial epilepsy.
cortical development on oral-facial-digital syndrome type VI. Am J Neu- A prospective study in 100 patients. Brain 1998;121 (Pt 11):2067–2081.
roradiol 2009;30:E22–E23. 298. Sasaki M, Kuwabara Y, Yoshida T, et al. Carbon-11-methionine PET in
276. Baraitser M, Winter R. Iris coloboma, ptosis, hypertelorism and mental focal cortical dysplasia: a comparison with fluorine-18-FDG PET and
retardation: a new syndrome. J Med Genet 1988;25:41–43. technetium-99m-ECD SPECT. J Nucl Med 1998;39:974–977.
277. Ramer J, Mascari M, Manders E, Ladda R. Trigonocephaly, pachygyria, 299. Madakasira PV, Simkins R, Narayanan T, Dunigan K, Poelstra RJ, Man-
retinal coloboma, and cardiac defect: a distinct syndrome. Dysmorph Clin til J. Cortical dysplasia localized by [11C]methionine positron emission
Genet 1992;6:15–20. tomography: case report. Am J Neuroradiol 2002;23:844–846.
278. Graf W, Born D, Sarnat H. The pachygyria-polymicrogyria spectrum of 300. Fois A, Farnetani MA, Balestri P, et al. EEG, PET, SPET, and MRI in intrac-
cortical dysplasia in X-linked hydrocephalus. Eur J Pediatr Surg 1990;8 table childhood epilepsies: possible surgical correlations. Childs Nerv Syst
(Suppl 1):10–14. 1995;11:672–678.
279. Raymond AA, Fish DR, Sisodiya SM, Alsanjari N, Stevens JM, Shorvon SD. 301. Newton MR, Berkovic SF, Austin MC, Rowe CC, McKay WJ, Bladin PF.
Abnormalities of gyration, heterotopias, tuberous sclerosis, focal cortical SPECT in the localization of extra-temporal and temporal seizure foci.
dysplasia, microdysgenesis, dysembryoplastic neuroepithelial tumor and J Neurol Neurosurg Psychiatry 1995;59:26–30.
dysgenesis of the archicortex in epilepsy: clinical, EEG and neuroimaging 302. Kaiboriboon K, Lowe VJ, Chantarujikapong SI, Hogan RE. The useful-
features in 100 adult patients. Brain 1995;118:629–660. ness of subtraction ictal SPECT coregistered to MRI in single- and dual-
280. Krsek P, Maton B, Korman B, et al. Different features of histopathologi- headed SPECT cameras in partial epilepsy. Epilepsia 2002;43(4):408–414.
cal subtypes of pediatric focal cortical dysplasia. Ann Neurol 2008;63: 303. Cascino GD. Surgical treatment for extratemporal epilepsy. Curr Treat
758–769. Options Neurol 2004;6:257–262.
281. Widjaja E, Otsubo H, Raybaud CA, et al. Characteristics of MEG and MRI 304. la Fougère C, Rominger A, Förster S, Geisler J, Bartenstein P. PET and
between Taylor’s focal cortical dysplasia (type II) and other cortical dys- SPECT in epilepsy: a critical review. Epilepsy Behav 2009;15:50–55.
plasia: Surgical outcome after complete resection of MEG spike source and 305. Kuzniecky R, Hetherington H, Pan J, et al. Proton spectroscopic imaging
MR lesion in pediatric cortical dysplasia. Epilepsy Res 2008;82:147–155. at 4.1 tesla in patients with malformations of cortical development and
282. Kuzniecky R, Jackson G. Magnetic Resonance in Epilepsy. New York: Raven epilepsy. Neurology 1997;48:1018–1024.
Press, 1995. 306. Garcia PA, Laxer KD, van der Grond J, Hugg JW, Weiner MW. Proton
283. Kuzniecky R, Morawetz R, Faught E, Black L. Frontal and central lobe magnetic resonance spectroscopic imaging in patients with frontal lobe
focal dysplasia: clinical, EEG, and imaging features. Dev Med Child Neurol epilepsy. Ann Neurol 1995;37:279–281.
1995;37:159–166. 307. Woermann FG, McLean MA, Bartlett PA, Barker GJ, Duncan JS. Quantita-
284. Swartz BE, Khonsari A, Brown C, Mandelkern M, Simkins F, Krisdakum- tive short echo time proton magnetic resonance spectroscopic imaging
torn T. Improved sensitivity of 18FDG-positron emission tomography study of malformations of cortical development causing epilepsy. Brain
scans in frontal and “frontal plus” epilepsy. Epilepsia 1995;36:388–395. 2001;124:427–436.
285. Richardson MP, Koepp MJ, Brooks DJ, Fish DR, Duncan JS. Benzodi- 308. Stanley JA, Cendes F, Dubeau F, Andermann F, Arnold DL. Proton mag-
azepine receptors in focal epilepsy with cortical dysgenesis: an 11C- netic resonance spectroscopic imaging in patients with extratemporal epi-
Flumazenil PET study. Ann Neurol 1996;40:188–198. lepsy. Epilepsia 1998;39(3):267–273.
286. Newton MR, Austin MC, Chan JG, et al. Ictal SPECT using 99m-Tc- 309. Li LM, Cendes F, Andermann F, Dubeau F, Arnold DL. Spatial extent
HMPAO: methods for rapid preparation and optimal deployment of of neuronal metabolic dysfunction measured by proton MR spectro-
tracer during spontaneous seizures. J Nucl Med 1993;34:666–670. scopic imaging in patients with localization-related epilepsy. Epilepsia
287. Lee N, Radtke R, Gray L, et al. Neuronal migration disorders: positron 2000;41(6):666–674.
emission tomography correlations. Ann Neurol 1994;35:290–297. 310. Li LM, Cendes F, Bastos AC, Andermann F, Dubeau F, Arnold DL. Neu-
288. Chugani HT, Shields WD, Shewmon DA, Olson DM, Phelps ME, Peacock ronal metabolic dysfunction in patients with cortical developmental mal-
WJ. Infantile spasms: I. PET identifies focal cortical dysgenesis in crypto- formations. A proton magnetic resonance spectroscopic imaging study.
genic csases for surgical treatment. Ann Neurol 1990;27:406–413. Neurology 1998;50:755–759.
289. Chiron C, Dulac O, Nuttin C, Depas G. Functional Imaging in Cortical 311. Eriksson SH, Rugg-Gunn FJ, Symms MR, Barker GJ, Duncan JS. Diffusion
Dysplasia: SPECT. Philadelphia: Lippencott-Raven, 1996. tensor imaging in patients with epilepsy and malformations of coritcal
290. Moore K, Funke M, Constantino T, Katzman G, Lewine J. Magnetoen- development. Brain 2001;124:617–626.
cephalographically directed review of high-spatial-resolution surface-coil 312. Rugg-Gunn FJ, Eriksson SH, Symms MR, Barker GJ, Duncan JS. Diffu-
MR images improves lesion detection in patients with extratemporal epi- sion tensor imaging of cryptogenic and acquired partial epilepsies. Brain
lepsy. Radiology 2002;225:880–887. 2001;124:627–636.
291. Eliashiv D, Elsas S, Squires K, Fried I, Engel J, Jr. Ictal magnetic source 313. Cascino GD. Neuroimaging in epilepsy: diagnostic strategies in partial
imaging as a localizing tool in partial epilepsy. Neurology 2002;59:1600– epilepsy. Semin Neurol 2008;28:523–532.
1610. 314. Krsek P, Maton B, Jayakar P, et al. Incomplete resection of focal cortical
292. Schwartz ES, Dlugos D, Storm P, et al. Magnetoencephalography for pedi- dysplasia is the main predictor of poor postsurgical outcome. Neurology
atric epilepsy: how we do it. Am J Neuroradiol 2008;29:832–837. 2009;72:217–223.

Barkovich_Chap05.indd 550 5/6/2011 9:01:36 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 551

315. Lerner JT, Salamon N, Hauptman JS, et al. Assessment and surgical 338. Taylor DC, Falconer MA, Bruton CJ, Corsellis JAN. Focal dysplasia of the
outcomes for mild type I and severe type II cortical dysplasia: a critical cerebral cortex in epilepsy. J Neurol Neurosurg Psychiatr 1971;34:369–387.
review and the UCLA experience. Epilepsia 2009;50:1310–1335. 339. Cepeda C, André VM, Flores-Hernández J, et al. Pediatric cortical dyspla-
316. Haydar T, Kuan C, Flavell R, Rakic P. The role of cell death in regulating sia: correlations between neuroimaging, electrophysiology and location
the size and shape of the mammalian forebrain. Cerebral Cortex 1999;9: of cytomegalic neurons and balloon cells and glutamate/GABA synaptic
621–626. circuits. Dev Neurosci 2005;27:59–76.
317. Sheen VL, Ganesh VS, Topcu M, et al. Mutations in ARFGEF2 implicate 340. Colombo N, Tassi L, Galli C, et al. Focal cortical dysplasias: MR imaging,
vesicle trafficking in neural progenitor proliferation and migration in the histopathologic and clinical correlations in surgically treated patients with
human cerebral cortex. Nat Genet 2004;36(1):69–76. epilepsy. Am J Neuroradiol 2003;24:724–733.
318. Arnold S, Huang G, Cheung A, et al. The T-box transcription factor 341. Hildebrandt M, Pieper T, Winkler P, Kolodziejczyk D, Holthausen H,
Eomes/Tbr2 regulates neurogenesis in the cortical subventricular zone. Blümcke I. Neuropathological spectrum of cortical dysplasia in children
Genes Dev 2008;22:2479–2484. with severe focal epilepsies. Acta Neuropathologica 2005;110(1):1.
319. Sessa A, Mao CA, Hadjantonakis AK, Klein WH, Broccoli V. Tbr2 directs 342. Palmini A, Najm I, Avanzini G, et al. Terminology and classification of the
conversion of radial glia into basal precursors and guides neuronal ampli- cortical dysplasias. Neurology 2004;62(6 Suppl 3):S2–S8.
fication by indirect neurogenesis in the developing neocortex. Neuron 343. Chamberlain WA, Cohen ML, Gyure KA, et al. Interobserver and intraob-
2008;60:56–69. server reproducibility in focal cortical dysplasia (malformations of corti-
320. Tang BL. Molecular genetic determinants of human brain size. Biochem cal development). Epilepsia 2009;50(12):2593–2598.
Biophys Res Commun 2006;345:911–916. 344. Blümcke I, Thom M, Aronica E, et al. The histopathological spectrum of
321. Chae TH, Walsh CA. Genes that control the size of the cerebral cortex. focal cortical dysplasias: a revised classification system of the International
Novartis Found Symp 2007;288:79–90. League against epilepsy. Epilepsia 2010 E-published Nov 10, 2010.
322. Passemard S, Titomanlio L, Elmaleh M, et al. Expanding the clinical and 345. Barkovich AJ, Kuzniecky RI, Bollen AW, Grant PE. Focal transmantle
neuroradiologic phenotype of primary microcephaly due to ASPM muta- dysplasia: a specific malformation of cortical development. Neurology
tions. Neurology 2009;73:962–969. 1997;49:1148–1152.
323. Shen J, Gilmore EC, Marshall CA, et al. Mutations in PNKP cause micro- 346. Mackay M, Becker L, Chuang S, et al. Malformations of cortical devel-
cephaly, seizures and defects in DNA repair. Nat Genet 2010;42(3): opment with balloon cells: clinical and radiologic correlates. Neurology
245–249. 2003;60:580–587.
324. Barkovich AJ, Ferriero DM, Barr RM, et al. Microlissencephaly: a het- 347. Becker AJ, Urbach H, Scheffler B, et al. Focal cortical dysplasia of Taylor’s
erogeneous malformation of cortical development. Neuropediatrics balloon cell type: mutational analysis of the TSC1 gene indicates a patho-
1998;29:113–119. genic relationship to tuberous sclerosis. Ann Neurol 2002;52:29–37.
325. Dobyns W, Barkovich A. Microcephaly with simplified gyral pattern 348. Majores M, Blumcke I, Urbach H, et al. Distinct allelic variants of TSC1
(oligogyric microcephaly) and microlissencephaly. Neuropediatrics and TSC2 in epilepsy associated cortical malformations without balloon
1999;30:105–106. cells. J Neuropathol Exp Neurol 2005;64:629–637.
326. Lammens M, Heil J, Gabree FJ, van Engelen B, van den Heuvel L, Weemaes C. 349. Schönberger A, Niehusmann P, Urbach H, et al. Increased frequency of
Nijmegen breakage syndrome: a neuropathological study. Neuropediatrics distinct TSC2 allelic variants in focal cortical dysplasias with balloon cells
2003;34:189–193. and mineralization. Neuropathology 2009;29:559–565.
327. Bond J, Scott S, Hampshire DJ, et al. Protein-truncating mutations in ASPM 350. Lamparello P, Baybis M, Pollard J, et al. Developmental lineage of cell
cause variable reduction in brain size. Am J Hum Genet 2003;73:1170–1177. types in cortical dysplasia with balloon cells. Brain 2007;130:2267–2276.
328. Jackson AP, Eastwood H, Bell SM, et al. Identification of microcephalin, 351. Palmini A, Gambardella A, Andermann F, et al. The human dysplastic cortex
a protein implicated in determining the size of the human brain. Am is intrinsically epileptogenic. In: Guerrini R, Andermann F, Canapicchi R,
J Hum Genet 2002;71:136–142. Roger J, Zifkin B, Pfanner P, eds. Dysplasias of Cerebral Cortex and Epilepsy.
329. Stromme P, Dahl E, Flage T, Stene-Johansen H. Apple peel intestinal Philadelphia: Lippencott-Raven, 1996:43–52.
atresia in siblings with ocular anomalies and microcephaly. Clin Genet 352. Usui N, Matsuda K, Mihara T, et al. MRI of cortical dysplasia - correlation
1993;44:208–210. with pathological findings. Neuroradiology 2001;43:830–837.
330. Bellini C, Mazzella M, Arioni C, Fondelli MP, Serra G. ‘Apple-peel’ intesti- 353. Mischel PS, Nguyen LP, Vinters HV. Cerebral cortical dysplasia associated
nal atresia, ocular anomalies, and microcephaly syndrome: brain magnetic with pediatric epilepsy. Review of neuropathologic features and proposal
resonance imaging study. Am J Med Genet 2002;110:176–178. for a grading system. J Neuropath Exp Neurol 1995;54:137–153.
331. van Bever Y, van Hest L, Wolfs R, Tibboel D, van den Hoonaard TL, 354. Widdess-Walsh P, Kellinghaus C, Jeha L, et al. Electro-clinical and imag-
Gischler SJ. Exclusion of a PAX6, FOXC1, PITX2, and MYCN mutation ing characteristics of focal cortical dysplasia: correlation with pathological
in another patient with apple peel intestinal atresia, ocular anomalies and subtypes. Epilepsy Res 2005;67(1–2):25.
microcephaly and review of the literature. Am J Med Genet 2008;146A: 355. Spreafico R, Battaglia G, Arcelli P, et al. Cortical dysplasia: an immunocy-
500–504. tochemical study of three patients. Neurology 1998;50:27–36.
332. Kelley RI, Robinson D, Puffenberger EG, Strauss KA, Morton DH. Amish 356. Palmini A, Gambardella A, Andermann F, et al. Intrinsic epileptogenic-
lethal microcephaly: a new metabolic disorder with severe congenital ity of human dysplastic cortex as suggested by corticography and surgical
microcephaly and 2-ketoglutaric aciduria. Am J Med Genet 2002;112: results. Ann Neurol 1995;37:476–487.
318–326. 357. Crino P, Duhaime A, Baltuch G, White R. Differential expression of gluta-
333. Chandler KE, Del Rio A, Rakshi K, et al. Leucodysplasia, microcephaly, mate and GABA-A receptor subunit mRNA in cortical dysplasia. Neurol-
cerebral malformation (LMC): a novel recessive disorder linked to 2p16. ogy 2001;56:906–913.
Brain 2006;129(1):272–277. 358. Calcagnotto ME, Paredes MF, Tihan T, Barbaro NM, Baraban SC. Dys-
334. Berg BO. Developmental disorders of the nervous system. In: Berg BO, ed. function of synaptic inhibition in epilepsy associated with focal cortical
Principles of Child Neurology. New York: McGraw-Hill, 1996:665–690. dysplasia. J Neurosci 2005;25:9649–9657.
335. Warkany J, Lemire RJ, Cohen MM, Jr. Mental Retardation and Congenital 359. Colombo N, Salamon N, Raybaud C, Ozkara C, Barkovich AJ. Imaging of
Malformations of the Central Nervous System. Chicago: Yearbook, 1981. malformations of cortical development. Epileptic Disord 2009;11:194–205.
336. Evrard P, de Saint-Georges P, Kadhim HJ, Gadisseux J-F. Pathology of pre- 360. Demerens C, Stankoff B, Logak M, et al. Induction of myelination in
natal encephalopathies. In: French J, ed. Child Neurology and Developmen- the central nervous system by electrical activity. Proc Natl Acad Sci USA
tal Disabilities. Baltimore: Paul H. Brookes, 1989:153–176. 1996;93:9887–9892.
337. Barkovich AJ. Congenital malformations of the brain and skull. In: 361. Girard N, Zimmerman RA, Schnur R, Haselgrove J, Christensen K.
Barkovich AJ, ed. Pediatric Neuroimaging, 2nd ed. New York: Raven Press, Magnetization transfer in the investigation of patients with tuberous
1995:177–275. sclerosis. Neuroradiology 1997;39:523–528.

Barkovich_Chap05.indd 551 5/6/2011 9:01:36 PM


552 Pediatric Neuroimaging

362. Urbach H, Scheffler B, Heinrichsmeier T, et al. Focal cortical dysplasia of 386. Griffiths PD, Welch R, Gardner-Medwin D, Gholkar A, McAllister V. The
Taylor’s balloon cell type: a clinicopathological entity with characteristic radiological features of hemimegalencephaly including three cases associ-
neuroimaging and histopathological features, and favorable postsurgical ated with Proteus syndrome. Neuropediatrics 1994;25:140–144.
outcome. Epilepsia 2002;43:33–40. 387. Peserico A, Battistella PA, Bertoli P, Drigo P. Unilateral hypomelanosis of
363. Marusic P, Najm I, Ying Z, et al. Focal cortical dysplasias in eloquent cor- Ito with hemimegalencephaly. Acta Paediatr Scand 1988;77:446–447.
tex: functional characteristics and correlation with MRI and histopatho- 388. Cusmai R, Curatolo P, Mangano S, Cheminal R, Echenne B. Hemimegal-
logic changes. Epilepsia 2002;43:27–32. encephaly and neurofibromatosis. Neuropediatrics 1990;21:179–182.
364. Grant PE, Barkovich AJ, Wald LL, Dillon WP, Vigneron DB. High- 389. Galluzzi P, Cerase A, Strambi M, Buoni S, Fois A, Venturi C. Hemimegal-
resolution surface coil MR of cortical lesions in medically refractory encephaly in tuberous sclerosis complex. J Child Neurol 2002;17:677–680.
epilepsy: a prospective study. Am J Neuroradiol 1997;18:291–301. 390. Kato M, Mizuguchi M, Sakuta R, Takashima S. Hypertrophy of the cerebral
365. Knake S, Triantafyllou C, Wald LL, et al. 3T phased array MRI improves white matter in hemimegalencephaly. Pediatr Neurol 1996;14:335–338.
the presurgical evaluation in focal epilepsies: a prospective study. Neurol- 391. Wolpert SM, Cohen A, Libenson M. Hemimegalencephaly: a longitudinal
ogy 2005;65:1026–1031. MR study. Am J Neuroradiol 1994;15:1479–1482.
366. Ekstrom A, Suthana N, Behnke E, Salamon N, Bookheimer S, Fried I. 392. Tagawa T, Otani K, Futagi Y, Wakayama A, Morimoto K, Morita Y. Serial
High-resolution depth electrode localization and imaging in patients with IMP-SPECT and EEG studies in an infant with hemimegalencephaly.
pharmacologically intractable epilepsy. J Neurosurg 2008;108:812–815. Brain Dev 1994;16:475–479.
367. Sims JM. On hypertrophy and atrophy of the brain. Trans Med Chirurg Soc 393. Hoffmann KT, Amthauer H, Liebig T, et al. MRI and F-18 fluorodeoxyglu-
(Lond) 1835;19:346–348. cose positron emission tomography in hemimegalencephaly. Neuroradiol-
368. Fitz CR, Harwood-Nash DC, Boldt DW. The radiographic features of uni- ogy 2000;42:749–752.
lateral megalencephaly. Neuroradiology 1978;15:145–148. 394. Villemure J-G. Hemispherictomy techniques: a critical review. In:
369. Barkovich AJ, Chuang SH. Unilateral megalencephaly: correlation of Tuxhorn I, Holthausen H, Boenigk H, eds. Paedriatric Epilepsy Syndromes
MR imaging and pathologic characteristics. Am J Neuroradiol 1990;11: and their Surgical Treatment. London: John Libbey, 1997:729–738.
523–531. 395. King M, Stephenson J, Ziervogel M, Doyle D, Galbraith S. Hemimegalen-
370. Kalifa CL, Chiron C, Sellier N, et al. Hemimegalencephaly: MR imaging in cephaly–a case for hemispherectomy? Neuropediatrics 1985;16:46–55.
five children. Radiology 1987;165:29–33. 396. Fukuyama Y, Kawazura M, Haruna H. A peculiar form of congenital
371. Townsend JJ, Nielsen SL, Malamud N. Unilateral megalencephaly: Hama- muscular dystrophy. Report of fifteen cases. Paediatr Universit (Tokyo)
rtoma or neoplasm? Neurology 1975;25:448–453. 1960;4:5–8.
372. Flores-Sarnat L, Sarnat H, Davila-Gutierrez G, Alvarez A. Hemimegalen- 397. Fukuyama Y, Osawa M, Suzuki H. Congenital progressive muscular dys-
cephaly: part 2. Neuropathology suggests a disorder of cellular lineage. trophy of the Fukuyama type - clinical, genetic, and pathological consid-
J Child Neurol 2003;18:776–785. erations. Brain Dev 1981;3:1–29.
373. Renowden SA, Squier M. Unusual magnetic resonance and neuropatho- 398. Santavuori P, Leisti J, Kruus S. Muscle, eye and brain disease: a new syn-
logical findings in hemimegalencephaly: report of a case following hemi- drome. Neuropädiatrie (supp) 1977;8:553–558.
spherectomy. Dev Med Child Neurol 1994;36:357–369. 399. Santavuori P, Somer H, Sainio K, et al. Muscle-eye-brain disease. Brain
374. Manz HJ, Phillips TM, Rowden G, McCullough DC. Unilateral megalen- Dev 1989;11:147–153.
cephaly, cerebral cortical dysplasia, neuronal hypertrophy, and hetero- 400. Walker AE. Lissencephaly. Arch Neurol Psychiatry 1942;48:13–29.
topia: cytomorphometric, fluorometric cytochemical, and biochemical 401. Warburg M, Heuer HE. Chorioretinal dysplasia-microcephaly-mental
analyses. Acta Neuropath (Berl) 1979;45:97–103. retardation syndrome [letter]. Am J Med Genet 1994;52(1):117.
375. Bosman C, Boldrini R, Dimitri L, Di Rocco C, Corsi A. Hemimegalen- 402. Michele D, Barresi R, Kanagawa M, et al. Post-translational disruption
cephaly. Histological, immunohistochemical, ultrastructural and cyto- of dystroglycan-ligand interactions in congenital muscular dystrophies.
fluorimetric study of six patients. Childs Nerv Syst 1996;12:765–775. Nature 2002;418:417–422.
376. Sasaki M, Hashimoto T, Furushima W, et al. Clinical aspects of hemi- 403. Moore SA, Saito F, Chen J, et al. Deletion of brain dystroglycan recapitu-
megalencephaly by means of a nationwide survey. J Child Neurol 2005;20: lates aspects of congenital muscular dystrophy. Nature 2002;418:422–425.
337–341. 404. Kanagawa M, Toda T. The genetic and molecular basis of muscular dys-
377. Tinkle BT, Schorry EK, Franz DN, Crone KR, Saal HM. Epidemiology trophy: roles of cell-matrix linkage in the pathogenesis. J Hum Genet
of hemimegalencephaly: a case series and review. Am J Med Genet A 2006;51:915–926.
2005;139A(3):204–211. 405. Li S, Jin Z, Koirala S, et al. GPR56 Regulates Pial Basement Membrane
378. Flores-Sarnat L. Hemimegalencephaly. I. Genetic, clinical, and imaging Integrity and Cortical Lamination. J Neurosci 2008;28(22):5817–5826.
aspects. J Child Neurol 2002;17:373–384. 406. Beltran-Valero de Bernabe D, Voit T, Longman C, et al. Mutations in the
379. Salamon N, Andres M, Chute DJ, et al. Contralateral hemimicrencephaly FKRP gene can cause muscle-eye-brain disease and Walker-Warburg syn-
and clinical-pathological correlations in children with hemimegalenceph- drome. J Med Genet 2004;41:e61.
aly. Brain 2006;129:352–365. 407. Brockington M, Blake DJ, Prandini P, et al. Mutations in the fukutin-
380. Nakahashi M, Sato N, Yagishita A, et al. Clinical and imaging character- related protein gene (FKRP) cause a form of congenital muscular dystro-
istics of localized megalencephaly: a retrospective comparison of diffuse phy with secondary laminin alpha2 deficiency and abnormal glycosylation
hemimegalencephaly and multilobar cortical dysplasia. Neuroradiology of alpha-dystroglycan. Am J Hum Genet 2001;69(6):1198–1209.
2009;51:821–830. 408. van Reeuwijk J, Maugenre S, van den Elzen C, et al. The expanding phe-
381. Sarwar M, Schafer M. Brain malformations in linear nevus sebaceous syn- notype of POMT1 mutations: from Walker-Warburg syndrome to con-
drome: an MR study. J Comput Assist Tomogr 1988;12:338–340. genital muscular dystrophy, microcephaly, and mental retardation. Hum
382. Hager BC, Dyme IZ, Guertin SR, Tyler RJ, Tryciecky EW, Fratkin JD. Lin- Mutat 2006;27(5):453–459.
ear nevus sebaceous syndrome: megalencephaly and heterotopic gray 409. Saito Y, Yamamoto T, Mizuguchi M, et al. Altered glycosylation of [alpha]-
matter. Pediatr Neurol 1991;7:45–49. dystroglycan in neurons of Fukuyama congenital muscular dystrophy
383. Pavone L, Curatolo P, Rizzo R, et al. Epidermal nevus syndrome: a neuro- brains. Brain Res 2006;1075(1):223–228.
logic variant with hemimegalencephaly, gyral malformation,mental retar- 410. van Reeuwijk J, Grewal P, Salih M, et al. Intragenic deletion in the
dation, seizures, and facial hemihypertrophy. Neurology 1991;41:266–271. LARGE gene causes Walker-Warburg syndrome. Hum Genet 2007;121(6):
384. Happle R. How many epidermal nevus syndromes exist? A clinicogenetic 685–690.
classification. J Am Acad Dermatol 1991;25:550–556. 411. van Reeuwijk J, Janssen M, van den Elzen C, et al. POMT2 mutations cause
385. Dietrich RB, Glidden DE, Roth GM, Martin RA, Demo DS. The Proteus (alpha)-dystroglycan hypoglycosylation and Walker-Warburg syndrome.
syndrome: CNS manifestations. Am J Neuroradiol 1998;19:987–990. J Med Genet 2005;42(12):907–912.

Barkovich_Chap05.indd 552 5/6/2011 9:01:36 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 553

412. Van Maldergem L, Yuksel-Apak M, Kayserili H, et al. Cobblestone-like 434. Kobayashi K, Nakahori Y, Miyake M, et al. An ancient retrotransposal
brain dysgenesis and altered glycosylation in congenital cutis laxa, Debre insertion causes Fukuyama-type congenital muscular dystrophy. Nature
type. Neurology 2008;71(20):1602–1608. 1998;394:388–392.
413. Inoue T, Ogawa M, Mikoshiba K, Aruga J. Zic deficiency in the cortical 435. Toda T, Segawa M, Nomura Y, et al. Localization of a gene for Fukuyama
marginal zone and meninges results in cortical lamination defects resem- type congenital muscular dystrophy to chromosome 9q31-33. Nat Genet
bling those in type II lissencephaly. J Neurosci 2008;28:4712–4725. 1993;5:283–286.
414. Zarbalis K, Siegenthaler JA, Choe Y, May SR, Peterson AS, Pleasure SJ. Cor- 436. Beltran-Valero de Bernabe D, van Bokhoven H, van Beusekom E, et al.
tical dysplasia and skull defects in mice with a Foxc1 allele reveal the role A homozygous nonsense mutation in the fukutin gene causes a Walker-
of meningeal differentiation in regulating cortical development. Proc Natl Warburg syndrome phenotype. J Med Genet 2003;40(11):845–848.
Acad Sci USA 2007;104:14002–14007. 437. Godfrey C, Escolar D, Brockington M, et al. Fukutin gene mutations in
415. Echenne B, Arthuis M, Billard C, et al. Congenital muscular dystrophy steroid-responsive limb girdle muscular dystrophy. Ann Neurol 2006;60:
and CT scan anomalies. Report of a collaborative study of the Societé de 603–610.
Neurologie Infantile. J Neurol Sci 1986;75:7–22. 438. Aida N, Yagishita A, Takada K, Katsumata Y. Cerebellar MR in Fukuyama
416. Krijgsman JB, Barth PG, Stam FC, Sloof JF, Jaspar HH. Congenital muscu- congenital muscular dystrophy: polymicrogyria with cystic lesions. Am
lar dystrophy and cerebral dysgenesis in a Dutch family. Neuropaediatrics J Neuroradiol 1994;15:1755–1759.
1980;11:108–120. 439. Aida N, Tamagawa K, Takada K, et al. Brain MR in Fukuyama congenital
417. Topaloglu H, Yalaz K, Kale G, Ergin M. Congenital muscular dystrophy muscular dystrophy. Am J Neuroradiol 1996;17:605–614.
with cerebral involvement - report of a case of “occidental type cerebro- 440. Hino N, Kobayashi M, Shibata N, Yamamoto T, Saito K, Osawa M. Clinico-
muscular dystrophy”? Neuropediatrics 1990;21:53–54. pathological study on eyes from cases of Fukuyama type congenital mus-
418. Topaloglu H, Yalaz K, Renda Y, et al. Occidental type cerebromuscular cular dystrophy. Brain Dev 2001;23:97–107.
dystrophy: a report of eleven cases. J Neurol Neurosurg Psychiatr 1991;54: 441. Takada K, Nakamura H, Takashima S. Cortical dysplasia in Fukuyama
226–229. congenital muscular dystrophy: a Golgi and angioarchitectonic analysis.
419. Topaloglu H, Kale G, Yalnizoglu D, et al. Analysis of “pure” congenital Acta Neuropathol 1988;76:170–178.
muscular dystrophies in thirty-eight cases. How different is the classical 442. Takada K. Fukuyama congenital muscular dystrophy as a unique disor-
type 1 from the occidental type of cerebromuscular dystrophy? Neurope- der of neuronal migration: a neuropathological review and hypothesis.
diatrics 1994;25:94–100. Yonago Acta Medica 1988;31:1–16.
420. Voit T. Congenital muscular dystrophies: 1997 update. Brain Dev 443. Takada K, Nakamura H. Cerebellar micropolygyria in Fukuyama congeni-
1998;20:65–74. tal muscular dystrophy: observations in fetal and pediatric cases. Brain
421. Tomé FMS. The saga of congenital muscular dystrophy. Neuropediatrics Dev 1990;12:774–778.
1999;30:55–65. 444. Cormand B, Avela K, Pihko H, et al. Assignment of the muscle-eye-brain
422. Pini A, Merlini L, Tomé FMS, Chevallay M, Gobbi G. Merosin-negative disease gene to 1p32–34 by linkage analysis and homozygosity mapping.
congenital muscular dystrophy, occipital epilepsy with periodic spasms Am J Hum Genet 1999;64:126–135.
and focal cortical dysplasia. Report of three Italian cases in two families. 445. Vervoort VS, Holden KR, Ukadike KC, Collins JS, Saul RA, Srivastava AK.
Brain Dev 1996;18:316–322. POMGnT1 gene alterations in a family with neuological abnormalities.
423. van der Knaap MS, Smit LME, Barth PG, et al. MRI in classification of Ann Neurol 2004;56:143–148.
congenital muscular dystrophies with brain abnormalities. Ann Neurol 446. Longman C, Brockington M, Torelli S, et al. Mutations in the human
1997;42:50–59. LARGE gene cause MDC1D, a novel form of congenital muscular dystro-
424. Villanova M, Mercuri E, Bertini E, et al. Congenital muscular dystrophy phy with severe mental retardation and abnormal glycosylation of alpha-
associated with calf hypertrophy, microcephaly and severe mental retarda- dystroglycan. Hum Mol Genet 2003;12(21):2853–2861.
tion in three Italian families: evidence for a novel CMD syndrome. Neuro- 447. Valanne L, Pihko H, Katevuo K, Karttunen P, Somer H, Santavuori P.
muscul Disord 2000;10:541–547. MRI of the brain in muscle-eye-brain (MEB) disease. Neuroradiology
425. Triki C, Louhichi N, Meziou M, et al. Merosin-deficient congenital muscu- 1994;36:473–476.
lar dystrophy with mental retardation and cerebellar cysts, unlinked to the 448. Piao X, Basel-Vanagaite L, Straussberg R, et al. An autosomal recessive
LAMA2, MEB and CMD1B loci, in three Tunisian patients. Neuromuscul form of bilateral frontoparietal polymicrogyria maps to chromosome
Disord 2003;13:4–12. 16q12.2-21. Am J Hum Genet 2002;70:1028–1033.
426. Yoshioka M. Phenotypic spectrum of Fukutinopathy: most severe pheno- 449. Koirala S, Jin Z, Piao X, Corfas G. GPR56-regulated granule cell adhesion is
type of Fukutinopathy. Brain Dev 2009;31:419–422. essential for rostral cerebellar development. J Neurosci 2009;29:7439–7449.
427. Gerding H, Gullotta F, Kuchelmeister K, Busse H. Ocular findings in Walk- 450. Chang B, Piao X, Bodell A, et al. Bilateral frontoparietal polymicrogyria:
er-Warburg syndrome. Child Nerv Syst 1993;9:418–420. clinical and radiological features in 10 families with linkage to chromo-
428. Williams RS, Swisher CN, Jennings M, Ambler M, Caviness VSJ. Cerebro- some 16. Ann Neurol 2003;53:596–606.
ocular dysgenesis (Walker-Warburg syndrome): neuropathologic and 451. Yoshioka M, Kuroki S, Sasaki H, Baba K, Toda T. A variant of congenital
etiologic analysis. Neurology 1984;34:1531–1541. muscular dystrophy. Brain Dev 2002;24:24–29.
429. Dobyns WB, Kirkpatrick JB, Hittner HM, Roberts RM, Kretzer FL. Syn- 452. Topaloglu H, Brockington M, Yuva Y, et al. FKRP gene mutations cause
dromes with lissencephaly. 2: Walker-Warburg and cerebral occular mus- congenital muscular dystrophy, mental retardation, and cerebellar cysts.
cular syndromes and a new syndrome with Type 2 lissencephaly. Am J Med Neurology 2003;60:988–992.
Genet 1985;22:157–195. 453. Raybaud C, Girard N, Canto-Moreira N, Poncet M. High-definition mag-
430. Beltran-Valero de Bernabe D, Currier S, Steinbrecher A, et al. Mutations netic resonance imaging identification of cortical dysplasias: micropoly-
in the O-mannosyltransferase gene POMT1 give rise to the severe neu- gyria versus lissencephaly. In: Guerrini R, Andermann F, Canapicchi R,
ronal migration disorder Walker-Warburg syndrome. Am J Hum Genet Roger J, Zifkin B, Pfanner P, eds. Dysplasias of Cerebral Cortex and Epilepsy.
2002;71:1033–1043. Philadelphia: Lippincott-Raven, 1996:131–143.
431. Rhodes RE, Hatten HP, Jr, Ellington KS. Walker-Warburg syndrome. Am J 454. Francis F, Meyer G, Fallet-Bianco C, et al. Human disorders of cortical
Neuroradiol 1992;13:123–126. development: from passt to present. Eur J Neurosci 2006;23:877–893.
432. Haltia M, Leivo I, Somer H, et al. Muscle-eye-brain disease: a neuropatho- 455. Yingling J, Youn YH, Darling DL, et al. Neuroepithelial stem cell prolifera-
logical study. Ann Neurol 1997;41:173–180. tion requires LIS1 for precise spindle orientation and symmetric division.
433. Kostovic I, Judas M, Radios M, Hrabac P. Laminar organization of the Cell 2008;132:474–486.
human fetal cerebrum revealed by histochemical markers and magnetic 456. Hatten ME. Neurons don’t even make it to the starting gate. J Cell Biol
resonance imaging. Cereb Cortex 2002;12:536–544. 2005;170:867–871.

Barkovich_Chap05.indd 553 5/6/2011 9:01:36 PM


554 Pediatric Neuroimaging

457. Nasrallah IM, McManus MF, Pancoast MM, Wynshaw-Boris A, Golden JA. reveals a high deletion mutation rate in isolated lissencephaly sequence.
Analysis of non-radial interneuron migration dynamics and its disruption Genet Med 1998;1:29–33.
in Lis1+/− mice. J Comp Neurol 2006;496(6):847–858. 481. Cardoso C, Leventer RJ, Matsumoto N, et al. The location and type
458. Lo Nigro C, Chong CS, Smith AC, Dobyns WB, Carrozzo R, Ledbetter of mutation predict malformation severity in isolated lissencephaly
DH. Point mutations and an intragenic deletion in LIS1, the lissencephaly caused by abnormalities within the LIS1 gene. Hum Molec Genet 2000;9:
causative gene in isolated lissencephaly sequence and Miller-Dieker syn- 3019–3028.
drome. Hum Molec Genet 1997;6:157–164. 482. Leventer RJ, Cardoso C, Ledbetter DH, Dobyns WB. LIS1 missense muta-
459. Dobyns W, Elias E, Newlin A, Pagon R, Ledbetter D. Causal heterogeneity tions cause milder lissencephaly phenotypes including a child with nor-
in isolated lissencephaly. Neurology 1992;42:1375–1388. mal IQ. Neurology 2001;57:416–422.
460. Dobyns WB, Stratton RF, Greenberg F. Syndromes with lissencephaly. 483. Matsumoto N, Leventer RJ, Kuc JA, et al. Mutation analysis of the DCX
I: Miller-Dieker and Norman-Roberts syndromes and isolated lissencephaly. gene and geneotype/phenotype correlation in subcortical band heteroto-
Am J Med Genet 1984;18:509–526. pia. Eur J Hum Genet 2001;9:5–12.
461. Dobyns WB. The neurogenetics of lissencephaly. Neurologic Clinics 484. Stewart RM, Richman DP, Caviness VS, Jr. Lissencephaly and pachygyria: an
1989;7:89–105. architectonic and topographical analysis. Acta Neuropathol 1975;31:1–12.
462. Ross ME, Allen KM, Srivastava AK, et al. Linkage and physical mapping 485. Dobyns WB, Truwit CL, Ross ME, et al. Differences in the gyral pattern
of X-linked lissencephaly/SBH (XLIS): a gene causing neuronal migration distinguish chromosome 17-linked and X-linked lissencephaly. Neurology
defects in human brain. Hum Mol Genet 1997;6:555–562. 1999;53:270–277.
463. Pinard JM, Motte J, Chiron C, Brian R, Andermann E, Dulac O. Subcorti- 486. Forman MS, Squier W, Dobyns WB, Golden JA. Genotypically defined
cal laminar heterotopia and lissencephaly in 2 families: a single X-linked lissencephalies show distinct pathologies. J Neuropathol Exp Neurol
dominant gene. J Neurol Neurosurg Psychiatr 1994;57:914–920. 2005;64(10):847–857.
464. Matsumoto N, Leventer RJ, Kuc J, et al. Mutation analysis of the DCX 487. Barkovich AJ, Chuang SH, Norman D. MR of neuronal migration anoma-
(XLIS) gene and X chromosome inactivation in females with subcortical lies. Am J Neuroradiol 1987;8:1009–1017.
band heterotopia. Eur J Hum Genet 2001;9:5–12. 488. Zimmerman RA, Bilaniuk LT, Grossman RI. Computed tomography
465. Kumar RA, Pilz DT, Babatz TD, et al. TUBA1A mutations cause wide in migration disorders of human brain development. Neuroradiology
spectrum lissencephaly (smooth brain) and suggest that multiple neu- 1983;25:257–263.
ronal migration pathways converge on alpha tubulins. Hum Mol Genet 489. Bahi-Buisson N, Poirier K, Boddaert N, et al. Refinement of cortical
2010;19:2817–2827. dysgeneses spectrum associated with TUBA1A mutations. J Med Genet
466. Fallet-Bianco C, Loeuillet L, Poirier K, et al. Neuropathological phenotype 2008;45:647–653.
of a distinct form of lissencephaly associated with mutations in TUBA1A. 490. Morris-Rosendahl DJ, Najm J, Lachmeijer AM, et al. Refining the pheno-
Brain 2008;131:2304–2320. type of alpha-1a Tubulin (TUBA1A) mutation in patients with classical
467. Pilz DT, Matsumoto N, Minnerath S, et al. LIS1 and XLIS (DCX) muta- lissencephaly. Clin Genet 2008;74:425–433.
tions cause most classical lissencephaly, but different patterns of malfor- 491. Fong KW, Ghai S, Toi A, Blaser S, Winsor EJT, Chitayat D. Prenatal ultra-
mation. Hum Mol Genet 1998;7(13):2029–2037. sound findings of lissencephaly associated with Miller-Dieker syndrome
468. Uyanik G, Morris-Rosendahl DJ, Stiegler J, et al. Location and type of and comparison with pre- and postnatal magnetic resonance imaging.
mutation in the LIS1 gene do not predict phenotypic severity. Neurology Ultrasound Obstet Gynecol 2004;24:716–723.
2007;69(5):442–447. 492. Ross ME, Swanson K, Dobyns WB. Lissencephaly with cerebellar hypopla-
469. Mei D, Lewis R, Parrini E, et al. High frequency of genomic deletions and sia (LCH): a heterogeneous group of cortical malformations. Neuropedi-
duplication in the LIS1 gene in lissencephaly: implications for molecular atrics 2001;32:256–263.
diagnosis. J Med Genet 2008;45:355–361. 493. Kappeler C, Dhenain M, Phan Dinh Tuy F, et al. Magnetic resonance imag-
470. Aicardi J. The agyria-pachygyria complex: a spectrum of cortical malfor- ing and histological studies of corpus callosal and hippocampal abnormal-
mations. Brain Dev 1991;13:1–8. ities linked to doublecortin deficiency. J Comp Neurol 2007;500:239–254.
471. Saillour Y, Carion N, Quelin C, et al. LIS1-related isolated lissencephaly: 494. Hong SE, Shugart YY, Huang DT, et al. Autosomal recessive lissencephaly
spectrum of mutations and relationships with malformation severity. Arch with cerebellar hypoplasia (LCH) is associated with human reelin gene
Neurol 2009;66:1007–1015. mutations. Nat Genet 2000;26:93–96.
472. Barkovich AJ, Koch TK, Carrol CL. The spectrum of lissencephaly: 495. Barkovich AJ, Jackson DE, Jr, Boyer RS. Band heterotopia: a newly recog-
report of ten cases analyzed by magnetic resonance imaging. Ann Neurol nized neuronal migration anomaly. Radiology 1989;171:455–458.
1991;30:139–146. 496. Livingston JH, Aicardi J. Unusual MRI appearance of diffuse subcortical
473. Garcia CA, Dunn D, Trevor R. The lissencephaly syndrome in siblings. heterotopia or “double cortex” in two children. J Neurol Neurosurg Psy-
Arch Neurol 1978;35:608–611. chiatry 1990;53:617–620.
474. Gleeson JG, Allen KA, Fox JW, et al. Doublecortin, a brain-specific gene 497. Palmini A, Andermann F, Aicardi J, et al. Diffuse cortical dysplasia, or
mutated in human X-linked lissencephaly and double cortex syndrome, the “double cortex” syndrome: the clinical and epileptic spectrum in 10
encodes a putative signaling protein. Cell 1998;92:63–72. patients. Neurology 1991;41:1656–1662.
475. Guerrini R, Moro F, Andermann E, et al. Nonsyndromic mental retarda- 498. D’Agostino M, Bernasconi A, Das S, et al. Subcortical band heterotopia
tion and crytogenic epilepsy in women with doublecortin gene mutations. (SBH) in males: clinical, imaging and genetic findings in comparison with
Ann Neurol 2003;54:30–37. females. Brain 2002;125:2507–2522.
476. Aigner L, Uyanik G, Couillard-Despres S, et al. Somatic mosaicism and 499. Ono J, Mano T, Andermann E, et al. Band heterotopia or double cortex in
variable penetrance in doublecortin-associated migration disorders. Neu- a male: bridging structures suggest abnormality of the radial glial guide
rology 2003;60:329–332. system. Neurology 1997;48:1701–1703.
477. Barkovich AJ, Guerrini R, Battaglia G, et al. Band heterotopia: correlation 500. Pilz D, Stoodley N, Golden J. Neuronal migration, cerebral cortical
of outcome with MR imaging parameters. Ann Neurol 1994;36:609–617. development, and cerebral cortical anomalies. J Neuropathol Exp Neurol
478. Pilz D, Kuc J, Matsumoto N, et al. Subcortical band heterotopia in rare 2002;61:1–11.
affected males can be caused by missense mutations in DCX (XLIS) or 501. Kato M, Kanai M, Soma O, et al. Mutation of the doublecortin gene in
LIS1. Hum Molec Genet 1999;8:1757–1760. male patients with double cortex syndrome: somatic mosaicism detected
479. Sicca F, Kelemen A, Genton P, et al. Mosaic mutations of the LIS1 gene by hair root analysis. Ann Neurol 2001;50:547–551.
cause subcortical band heterotopia. Neurology 2003;61:1042–1046. 502. Viot G, Sonigo P, Simon I, et al. Neocortical neuronal arrangement
480. Pilz DT, Macha ME, Precht KS, Smith ACM, Dobyns WB, Ledbetter DH. in LIS1 and DCX lissencephaly may be different. Am J Med Genet
Fluorescence in situ hybridization analysis with LIS1 specific probes 2004;126A(2):123–128.

Barkovich_Chap05.indd 554 5/6/2011 9:01:36 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 555

503. Franzoni E, Bernardi B, Marchiani V, Crisanti AF, Marchi R, Fonda C. 525. Rossi M, Guerrini R, Dobyns WB, Andria G, Winter RM. Characterization
Band brain heterotopia. Case report and literature review. Neuropediatrics of brain malformations in the Baraitser-Winter syndrome and review of
1995;26:37–40. the literature. Neuropediatrics 2003;34:287–292.
504. Falconer J, Wada J, Martin W, Li D. PET, CT, and MRI imaging of neu- 526. Raymond AA, Fish DR, Stevens JM, Sisodiya SM, Alsanjari N, Shorvon SD.
ronal migration anomalies in epileptic patients. Can J Neurol Sci 1990;17: Subependymal heterotopia: a distinct neuronal migration disorder associ-
35–39. ated with epilepsy. J Neurol Neurosurg Psychiatry 1994;57:1195–1202.
505. Miura K, Watanabe K, Maeda N, et al. MR imaging and positron emission 527. Smith AS, Weinstein MA, Quencer RM, et al. Association of heterotopic
tomography of band heterotopia. Brain Dev 1993;15:288–290. gray matter with seizures: MR imaging. Radiology 1988;168:195–198.
506. De Volder AG, Gadisseux J-F, Michel CJ, et al. Brain glucose utilization 528. Barkovich AJ, Kjos BO. Gray matter heterotopias: MR characteristics and
in band heterotopia: synaptic activity of “double cortex.” Pediatr Neurol correlation with developmental and neurological manifestations. Radiol-
1994;11:290–294. ogy 1992;182:493–499.
507. Morell F, Whisler W, Hoeppner T, et al. Electrophysiology of hetero- 529. Mitchell L, Thomas P, Zacharin M, Scheffer I. Ectopic posterior pituitary
topic gray matter in the “Double Cortex” syndrome. Epilepsia 1992;33 lobe and periventricular heterotopia: cerebral malformations with the
(Suppl 3):76. same underlying mechanism? Am J Neuroradiol 2002;23:1475–1481.
508. Pinard J-M, Feydy A, Carlier R, Perez N, Pierot L, Burnod Y. Func- 530. Barkovich AJ, Kuzniecky RI. Gray matter heterotopia. Neurology
tional MRI in double cortex: functionality of heterotopia. Neurology 2000;55:1603–1608.
2000;54:1531–1533. 531. Wieck G, Leventer RJ, Squier WM, et al. Periventricular nodular heteroto-
509. Spreer J, Martin P, Greenlee M, et al. Functional MRI in patients with band pia with overlying polymicrogyria. Brain 2005;128(12):2811–2821.
heterotopia. Neuroimage 2001;14:357–365. 532. Guerrini R, Parrini E. Neuronal migration disorders. Neurobiol Dis
510. Dobyns W, Berry-Kravis E, Havernick N, Holden K, Viskochil D. X-linked 2009;38:154–166.
lissencephaly with absent corpus callosum and ambiguous genitalia. Am J 533. Sheen V, Dixon P, Fox J, et al. Mutations in the X-linked filamin 1 gene
Med Genet 1999;86:331–337. cause periventricular nodular heterotopia in males as well as in females.
511. Berry-Kravis E, Israel J. X-linked pachygyria and agenesis of the corpus Hum Mol Genet 2001;10:1775–1783.
callosum: evidence for an x chromosome lissencephaly locus. Ann Neurol 534. Sheen V, Topcu M, Berkovic S, et al. Autosomal recessive form of periven-
1994;36:229–233. tricular heterotopia. Neurology 2003;60:1108–1112.
512. Kitamura K, Yanazawa M, Sugiyama N, et al. Mutation of ARX causes 535. Dobyns WB, Guerrini R, Czapansky-Beilman D, et al. Bilateral periven-
abnormal development of forebrain and testes in mice and X-linked tricular nodular heterotopia with mental retardation and syndactyly in
lissencephaly with abnormal genitalia in humans. Nat Genet 2002;32: boys: a new X-linked mental retardation syndrome. Neurology 1997;49:
359–369. 1042–1047.
513. Butt SJB, Cobos I, Golden JA, Kessaris N, Pachnis V, Anderson SA. Tran- 536. Eksioglu YZ, Scheffer IE, Cardenas P, et al. Periventricular heterotopia:
scriptional regulation of cortical interneuron development. J Neurosci an X-linked dominant epilepsy locus causing aberrant cerebral cortical
2007;27(44):11847–11850. development. Neuron 1996;16:77–87.
514. Fulp CT, Cho G, Marsh ED, Nasrallah IM, Labosky PA, Golden JA. Iden- 537. Huttenlocher PR, Taravath S, Mojtahedi S. Periventricular heterotopia and
tification of Arx transcriptional targets in the developing basal forebrain. epilepsy. Neurology 1994;44:451–455.
Hum Molec Genet 2008;17:3740–3760. 538. Neal J, Apse K, Sahin M, Walsh CA, Sheen V. Deletion of chromosome
515. Kato M, Das S, Petras K, et al. Mutations of ARX are associated with strik- 1p36 is associated with periventricular nodular heterotopia. Am J Med
ing pleiotropy and consistent genotype-phenotype correlation. Hum Genet A 2006;140:1692–1695.
Mutat 2004;23(2):147–159. 539. Ferland R, Gaitanis J, Apse K, Tantravahi U, Walsh CA, Sheen V. Periven-
516. Bonneau D, Toutain A, Laquerrier A, et al. X-linked lissencephaly with tricular nodular heterotopia and Williams syndrome. Am J Med Genet A
absent corpus callosum and ambiguous genitalia (XLAG): clinical, mag- 2006;140:1305–1311.
netic resonance imaging, and neuropathological findings. Ann Neurol 540. Ferland RJ, Batiz LF, Neal J, et al. Disruption of neural progenitors along
2002;51:340–349. the ventricular and subventricular zones in periventricular heterotopia.
517. Kato M, Dobyns WB. X-linked lissencephaly with abnormal genitalia as a Hum Mol Genet 2009;18:497–516.
tangential migration disorder causing intractable epilepsy: propsal for a 541. d’Orsi G, Tinuper P, Bisulli F, et al. Clinical features and long term out-
new term, “interneuronopathy”. J Child Neurol 2005;20:392–397. come of epilepsy in periventricular nodular heterotopi. Simple compared
518. Miyata R, Hayashi M, Miyai K, Akashi T, Kato M, Kohyama J. Analysis with plus forms. J Neurol Neurosurg Psychiatry 2004;75:873–878.
of the hypothalamus in a case of X-linked lissencephaly with abnormal 542. Tassi L, Colombo N, Cossu M, et al. Electroclinical, MRI and neuropatho-
geneitalia (XLAG). Brain Dev 2009;31:456–460. logical study of 10 patients with nodular heterotopia, with surgical out-
519. Hourihane J, Bennett C, Chaudhuri R, Robb S, Martin N. A sibship with comes. Brain 2005;128:321–337.
a neuronal migration defect, cerebellar hypoplasia and congenital lym- 543. Moro F, Carrozzo R, Veggiotti P, et al. Familial periventricular heterotopia:
phedema. Neuropediatrics 1993;24:43–46. missense and distal truncating mutations of the FLN1 gene. Neurology
520. Schurig V, Orman AV, Bowen P. Nonprogressive cerebellar disorder with 2002;58:916–921.
mental retardation and autosomal recessive inheritance in Hutterites. Am 544. Martin N, Debussche C, De Broucker T, Mompoint D, Marsault C, Nahum H.
J Med Genet 1981;9:43–53. Gadolinium-DTPA enhanced MR imaging in tuberous sclerosis. Neurora-
521. D’Arcangelo G, Nakajima K, Miyata T, Ogawa M, Mikoshiba K, Curran T. diology 1990;31:492–497.
Reelin is a secreted glycoprotein recognized by the CR-50 monoclonal 545. Gomez MR. Tuberous Sclerosis, 2nd ed. New York: Raven Press, 1988.
antibody. J Neurosci 1997;17:23–31. 546. Inoue Y, Nakajima S, Fukuda P, et al. Magnetic resonance images of tuber-
522. Boycott KM, Flavelle S, Bureau A, et al. Homozygous deletion of the very ous sclerosis: further observations and clinical correlations. Neuroradiol-
low density lipoprotein receptor gene causes autosomal recessive cer- ogy 1988;30:379–384.
ebellar hypoplasia with cerebral gyral simplification. Am J Hum Genet 547. Guerrini R, Mei D, Sisodiya S, et al. Germline and mosaic mutations of
2005;77:477–483. FLN1 in men with periventricular heterotopia. Neurology 2004;63(1):
523. Sanner G. The dysequilibrium syndrome. A genetic study. Neuropadiatrie 51–56.
1973;4:403–413. 548. Poussaint TY, Fox JW, Dobyns WB, et al. Periventricular nodular hetero-
524. Dobyns WB, Leventer RJ. Lissencephaly: the clinical and molecular genetic topia in patients with filamin-1 gene mutations: neuroimaging findings.
basis of diffuse malformations of neuronal migration. In: Barth PG, ed. Pediatr Radiol 2000;30:748–755.
Disorders of Neuronal Migration. ICNA Series. London: MacKeith Press, 549. Oda T, Nagai Y, Fujimoto S, et al. Hereditary nodular heterotopia accom-
2003:24–57. panied by mega cisterna magna. Am J Med Genet 1993;47:268–271.

Barkovich_Chap05.indd 555 5/6/2011 9:01:36 PM


556 Pediatric Neuroimaging

550. Mitchell L, Simon E, Filly R, Barkovich A. The antenatal diagnosis of 575. Hayashi N, Tsutsumi Y, Barkovich AJ. Polymicrogyria without
subependymal heterotopia. Am J Neuroradiol 2000;21:296–300. porencephaly/schizencephaly. MRI analysis of the spectrum and the prev-
551. Bargalló N, Puerto B, De Juan C, Martinez-Crespo J, Lourdes Olondo alence of macroscopic findings in the clinical population. Neuroradiology
M. Hereditary subependymal heterotopia associated with mega cisterna 2002;44:647–655.
magna: antenatal diagnosis with magnetic resonance imaging. Ultrasound 576. Barkovich AJ. Morphology of subcortical heterotopia: a magnetic reso-
Obstet Gynecol 2002;20:86–89. nance study. Am J Neuroradiol 2000;21:290–295.
552. Barkovich AJ. Subcortical heterotopia: a distinct clinico-radiologic entity. 577. Takanashi J, Barkovich A. The changing MR imaging appearance of polymi-
Am J Neuroradiol 1996;17:1315–1322. crogyria: a consequence of myelination. Am J Neuroradiol 2003;24:788–793.
553. Dubeau F, Tampieri D, Andermann E, et al. Periventricular and subcor- 578. Barkovich AJ, Hevner R, Guerrini R. Syndromes of bilateral symmetrical
tical nodular heterotopia: comparison of clinical findings and results of polymicrogyria. Am J Neuroradiol 1999;20:1814–1821.
surgical treatment. In: Guerrini R, ed. Dysplasias of Cerebral Cortex and 579. Barkovich AJ. MRI Analysis of Sulcation Morphology in Polymicrogyria.
Epilepsy. Philadelphia: Lippencott-Raven, 1996:395–406. Epilepsia 2010;51(Suppl 1):17–22.
554. Marsh L, Lim KO, Sullivan EV, Lane B, Spielman D. Proton magnetic 580. Barkovich AJ. Abnormal vascular drainage in anomalies of neuronal
resonance spectroscopy of a gray matter heterotopia. Neurology 1996;47: migration. Am J Neuroradiol 1988;9:939–942.
1571–1574. 581. Kuzniecky R, Andermann F, Guerrini R. Congenital bilateral perisylvian
555. Englund C, Fink A, Lau C, et al. Pax6, Tbr2, and Tbr1 are expressed syndrome: study of 31 patients. The congenital bilateral perisylvian syn-
sequentially by radial glia, intermediate progenitor cells, and postmitotic drome milticenter collaborative study. Lancet 1993;341:608–612.
neurons in developing neocortex. J Neurosci 2005;25(1):247–251. 582. Guerreiro MM, Andermann E, Guerrini R, et al. Familial perisylvian poly-
556. Leventer RJ, Jansen A, Pilz DT, et al. Clinical and imaging heterogeneity of microgyria: a new familial syndrome of cortical maldevelopment. Ann
polymicrogyria: a study of 328 patients. Brain 2010;133:1415–1427. Neurol 2000;48:39–48.
557. Barkovich AJ, Linden CL. Congenital cytomegalovirus infection of the 583. Guerrini R, Dravet C, Raybaud C, et al. Neurological findings and seizure
brain: imaging analysis and embryologic considerations. Am J Neuroradiol outcome in children with bilateral opercular macrogyric-like changes
1994;15:703–715. detected by MRI. Dev Med Child Neurol 1992;34(8):694–705.
558. Wright R, Johnson D, Neumann M, et al. Congenital lymphocytic chorio- 584. Gropman AL, Barkovich AJ, Vezina LG, Conry JA, Dubovsky EC, Packer
meningitis virus syndrome: a disease that mimics congenital toxoplasmo- RJ. Pediatric congenital bilateral perisylvian syndrome: clinical and MRI
sis or Cytomegalovirus infection. Pediatrics 1997;100:E9. features in 12 patients. Neuropediatrics 1997;28:198–203.
559. Hallervorden J. Ueber eine Kohlenoxydvergiftung im Fetalleben mit Ent- 585. Becker PS, Dixon AM, Troncoso JC. Bilateral opercular polymicrogyria.
wicklunsstorung der Hirnrinde. Allg Z Psychiatr 1949;124:289–298. Ann Neurol 1989;25:90–92.
560. Barkovich AJ, Rowley HA, Bollen A. Correlation of prenatal events with 586. Rolland Y, Adamsbaum C, Sellier N, Robain O, Ponsot G, Kalifa G. Oper-
the development of polymicrogyria. Am J Neuroradiol 1995;16:822–827. cular malformations: clinical and MRI features in 11 children. Pediatr
561. Kuzniecky R. Familial diffuse cortical dysplasia. Arch Neurol 1994;51: Radiol 1995;25:S2–S8.
307–310. 587. Jansen A, Andermann E. Genetics of the polymicrogyria syndromes. J Med
562. Villard L, Nguyen K, Cardoso C, et al. A locus for bilateral perisylvian Genet 2005;42(5):369–378.
polymicrogyria maps to Xq28. Am J Hum Genet 2002;70:1003–1008. 588. Oliveira EPM, Hage SRV, Guimaraes C, et al. Characterization of language
563. Caraballo RH, Cersósimo RO, Mazza E, Fejerman N. Focal polymicrogyria and reading skills in familial polymicrogyria. Brain Dev 2008;30:254–260.
in mother and son. Brain Dev 2000;22:336–339. 589. Guerrini R, Barkovich A, Sztriha L, Dobyns W. Bilateral frontal polymi-
564. Borgatti R, Triulzi F, Zucca C, et al. Bilateral perisylvian polymicrogyria in crogyria. Neurology 2000;54:909–913.
three generations. Neurology 1999;52:1910–1913. 590. Guerrini R, Dulac O, Canapicchi R, et al. Epilepsie révélatrice d’une
565. Leventer RJ, Lese CM, Cardosi C, Rosenberry J, Weiss A, Stoodley N. dysplasie corticale occipito-pariétale parasagittale bilatérale. Epilepsies
A study of 220 patient swith polymicrogyria deliniates distinct pheno- 1994;6:131–139.
types and reveals genetic loci on chromosome 1p, 2p, 6q, 21q, 22q, and 591. Pascual-Castroviejo I, Pascual-Pascual S, Viano J, Martinez V, Palencia R.
Xq. Am J Hum Genet 2001;69:177. Unilateral polymicrogyria: a common cause of hemiplegia of prenatal ori-
566. Brooks AS, Bertoli-Avella AM, Burzynski GM, et al. Homozygous non- gin. Brain Dev 2001;23:216–222.
sense mutations in KIAA1279 are associated with malformations of the 592. Guerrini R, Dravet C, Bureau M, et al. Diffuse and localized dysplasias of
central and enteric nervous systems. Am J Hum Genet 2005;77:120–126. cerebral cortex: clinical presentation, outcome, and proposal for a mor-
567. Aligianis IA, Johnson CA, Gissen P, et al. Mutations of the catalytic phologic MRI classification based on a study of 90 patients. In: Guerrini R,
subunit of RAB3GAP cause Warburg Micro syndrome. Nat Genet Mar Andermann F, Canapicchi R, Roger J, Zifkin BG, Pfanner P, eds. Dys-
2005;37:221–223. plasias of Cerebral Cortex and Epilepsy. Philadelphia: Lippincott-Raven,
568. Roll P, Rudolf G, Pereira S, et al. SRPX2 mutations in disorders of language 1996:255–269.
cortex and cognition. Hum Mol Genet 2006;15:1195–1207. 593. Kuzniecky R, Andermann F, Guerrini R. The epileptic syndrome in the
569. Baala L, Briault S, Etchevers HC, et al. Homozygous silencing of T-box congenital bilateral perisylvian syndrome. CBPS Multicenter collaborative
transcription factor EOMES leads to microcephaly with polymicrogyria study. Neurology 1994;44:379–385.
and corpus callosum agenesis. Nat Genet 2007;39(4):454–456. 594. Jacobs K, Kharazia V, Prince D. Mechanisms underlying epileptogenesis in
570. Jaglin XH, Poirier K, Saillour Y, et al. Mutations in the beta-tubulin gene cortical malformations. Epilepsy Res 1999;36:165–188.
TUBB2B result in asymmetrical polymicrogyria. Nat Genet 2009;41: 595. Moore C, Toriello H, Abuelo D, et al. Macrocephaly-cutis marmorata
746–752. telangiectatica congenita: a distinct disorder with developmental delay
571. Guerrini R, Dravet C, Raybaud C, et al. Epilepsy and focal gyral anomalies and connective tissue abnormalities. Am J Med Genet 1997;70:67–73.
detected by MRI: electroclinico-morphological correlations and follow- 596. Giuliano F, David A, Edery P, et al. Macrocephaly-cutis marmorata telang-
up. Dev Med Child Neurol 1992;34(8):706–718. iectatica congenita: seven cases including two with unusual cerebral mani-
572. Barkovich AJ, Kjos BO. Non-lissencephalic cortical dysplasia: correlation festations. Am J Med Genet A 2004;126A:99–103.
of imaging findings with clinical deficits. Am J Neuroradiol 1992;13:95– 597. Mirzaa G, Dodge NN, Glass I, et al. Megalencephaly and perisylvian poly-
103. microgyria with postaxial polydactyly and hydrocephalus: a rare brain
573. Leventer R. Polymicrogyria and Related Disorders of Cortical Development: malformation syndrome associated with mental retardation and seizures.
A Clinical, Imaging, and Genetic Study. Melbourne: Child Health Institute, Neuropediatrics 2004;35:353–359.
University of Melbourne, 2007. 598. Garavelli L, Guareschi E, Errico S, et al. Megalencephaly and perisylvian
574. Guerrini R, Dubeau F, Dulac O, et al. Bilateral parasagittal parietooccipital polymicrogyria with postaxial polydactyly and hydrocephalus (MPPH):
polymicrogyria and epilepsy. Ann Neurol 1997;41:65–73. report of a new case. Neuropediatrics 2007;38(4):200–203.

Barkovich_Chap05.indd 556 5/6/2011 9:01:36 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 557

599. Gripp KW, Hopkins E, Vinkler C, et al. Significant overlap and possible 623. Meencke HJ, Janz D. The significance of microdysgenesis in primary
identity of macrocephaly capillary malformation and megalencephaly generalized epilepsy: an answer to the considerations of Lyon and Gastaut.
polymicrogyria-polydactyly hydrocephalus syndromes. Am J Med Genet Epilepsia 1985;26:368–371.
A 2009;149A(5):868–876. 624. Kim S, Na D, Byun H, et al. Focal cortical dysplasia: comparison of MRI
600. Martínez-Lage JF, Guillén-Navarro E, Almagro MJ, Felipe-Murcia M, and FDG-PET. J Comput Assist Tomogr 2000;24:296–302.
López-Guerrero A, Galarza M. Hydrocephalus and Chiari type 1 mal- 625. Chan S, Chin SS, Nordli DR, Goodman RR, DeLaPaz RL, Pedley TA.
formation in macrocephaly-cutis marmorata telangiectatica congenita: Prospectice magneitc resonance imaging identification of focal cor-
a case-based update. Childs Nerv Syst 2009;26:13–18. tical dysplasia, including the non-balloon cell subtype. Ann Neurol
601. Göhlich-Ratmann G, Baethmann M, Lorenz P, et al. Megalencephaly, 1998;44:749–757.
mega corpus callosum, and complete lack of motor development: a previ- 626. Saint Martin C, Adamsbaum C, Robain O, Chiron C, Kalifa G. An unusual
ously undescribed syndrome. Am J Med Genet 1998;79:161–167. presentation of focal cortical dysplasia. Am J Neuroradiol 1995;16:840–842.
602. Yakovlev PI, Wadsworth RC. Schizencephalies. A study of the congenital 627. Yagishita A, Arai N, Maehara T, Shimizu H, Tokumaru AM, Oda M.
clefts in the cerebral mantle. 1. Clefts with fused lips. J Neuropathol Exp Focal cortical dysplasia: appearance on MR images. Radiology 1997;203:
Neurol 1946;5:116–130. 553–559.
603. Yakovlev PI, Wadsworth RC. Schizencephalies. A study of the congenital 628. Raymond AA, Fish DR, Stevens JM, Cook MJ, Sisodiya SM, Shorvon SD.
clefts in the cerebral mantle. 2. Clefts with hydrocephalus and lips sepa- Association of hippocampal sclerosis with cortical dysgenesis in patients
rated. J Neuropathol Exp Neurol 1946;5:169–206. with epilepsy. Neurology 1994;44:1841–1845.
604. Curry CJ, Lammer EJ, Nelson V, Shaw GM. Schizencephaly: heterogeneous 629. Kuzniecky R, Garcia JH, Faught E, Morawetz RB. Cortical dysplasia in
etiologies in a population of 4 million California births. Am J Med Genet A temporal lobe epilepsy: magnetic resonance imaging correlations. Ann
2005;137(2):181–189. Neurol 1991;29:293–298.
605. Granata T, Farina L, Faiella A, et al. Familial schizencephaly associated 630. Daumas-Duport C, Scheithauer BW, Chodkiewicz J-P, Laws ER, Jr,
with EMX2 mutation. Neurology 1997;48:1403–1406. Vedrenne C. Dysembryoplastic neuroepithelial tumor: a surgically curable
606. Haverkamp F, Zerres K, Ostertun B, Emons D, Lentze M. Familial schi- tumor of young patients with intractable partial seizures. Neurosurgery
zencephaly: further delineation of a rare disorder. J Med Genet 1995;32: 1988;23:545–556.
242–244. 631. Daumas-Duport C. Dysembryoplastic neuroepithelial tumors. Brain
607. Robinson RO. Familial schizencephaly. Dev Med Child Neurol Pathol 1993;3:283–295.
1991;33:1010–1014. 632. Prayson R, Khajavi K, Comair Y. Cortical architectural abnormalities and
608. Tietjen I, Erdogan FS, Currier S, et al. EMX2-independent familial MIB1 immunoreactivity in gangliogliomas: a study of 60 patients with
schizencephaly: Clinical and genetic analyses. Am J Med Genet Part A intracranial tumors. J Neuropathol Exp Neurol 1995;54:513–520.
2005;135A(2):166–170. 633. Prayson RA, Frater J. Cortical dysplasia in extratemporal lobe intractable
609. Tietjen I, Bodell A, Apse K, et al. Comprehensive EMX2 genotyping of a epilepsy: a study of 52 cases. Ann Diagn Pathol 2003;7:139–146.
large schizencephaly case series. Am J Med Genet Part A 2007;143A(12): 634. Marin-Padilla M, Parisi JE, Armstrong DL, Sargent SK, Kaplan JA. Shaken
1313–1316. infant syndrome: developmental neuropathology, progressive cortical
610. Granata T, Battaglia G, D’Incerti L, et al. Schizencephaly: clinical find- dysplasia, and epilepsy. Acta Neuropathol (Berl) 2002;103(4):321–332.
ings. In: Guerrini R, ed. Dysplasias of the Cerebral Cortex and Epilepsy. 635. Simon E, Hevner R, Pinter J, Kinsman S, Hahn J, Barkovich AJ. Assess-
Philadelphia: Lippencott-Raven, 1996:407–415. ment of deep gray nuclei in holoprosencephaly. Am J Neuroradiol 2000;21:
611. Barkovich AJ, Kjos BO. Schizencephaly: correlation of clinical findings 1955–1961.
with MR characteristics. Am J Neuroradiol 1992;13:85–94. 636. Roessler E, Muenke M. Holoprosencephaly: a paradigm for the complex
612. Aniskiewicz AS, Frumkin NL, Brady DE, Moore JB, Pera A. Magnetic res- genetics of brain development. J Inher Metab Dis 1998;21:481–497.
onance imaging and neurobehavioral correlates in schizencephaly. Arch 637. Olsen CL, Hughes JP, Youngblood LG, Sharpe-Stimac M. Epidemiology
Neurol 1990;47(8):911–916. of holoprosencephaly and phenotypic characteristics of affected children:
613. Miller GM, Stears JC, Guggenheim MA, Wilkening GN. Schizencephaly: a New York State, 1984–1989. Am J Med Genet 1997;73:217–226.
clinical and CT study. Neurology 1984;34:997–1001. 638. Probst FP. The Prosencephalies: Morphology, Neuroradiological Appearances
614. Packard AM, Miller VS, Delgado MR. Schizencephaly: correlations of and Differential Diagnosis. Berlin: Springer-Verlag, 1979.
clinical and radiologic features. Neurology 1997;48:1427–1434. 639. Fitz CR. Holoprosencephaly and related entities. Neuroradiology
615. Chuang SH, Fitz CR, Chilton SJ, Harwood-Nash DC, Donoghue V. Schi- 1983;25:225–238.
zencephaly: spectrum of CT findings in association with septo-optic dys- 640. DeMyer W. Holoprosencephaly. In: Vinker PI, Bruyn JW, eds. Handbook of
plasia. Paper Presented at: Annual Meeting of the Radiological Society of Clinical Neurology, Vol 30. Amsterdam, The Netherlands: North Holland,
North America, Washington, DC, 1984. 1977:431–478.
616. Barkovich AJ, Fram EK, Norman D. Septo-optic dysplasia: MR imaging. 641. DeMyer W. Holoprosencephaly (cyclopia-arhinencephaly). In: Myrian-
Radiology 1989;171:189–192. thopoulos N, ed. Malformations, Vol 50. Amsterdam, New York: Elsevier,
617. Barkovich AJ, Norman D. MR of schizencephaly. Am J Neuroradiol 1987:225–244.
1988;9:297–302. 642. Muenke M, Bone LJ, Mitchell HF, et al. Physical mapping of the holopros-
618. Denis D, Chateil J-F, Brun M, et al. Schizencephaly: clinical and imaging encephaly critical region in 21q22.3, exclusion of SIM2 as a candidate gene
features in 30 infantile cases. Brain Dev 2000;22:475–483. for holoprosencephaly, and mapping of SIM2 to a region of chromosome
619. Hayashi N, Tsutsumi Y, Barkovich AJ. Morphological features and asso- 21 important for Down syndrome. Am J Hum Genet 1995;57:1074–1079.
ciated anomalies of schizencephaly in the clinical population: detailed 643. Schell U, Wienberg J, Kohler A, et al. Molecular characterization of break-
analysis of MR images. Neuroradiology 2002;44:418–427. points in patients with holoprosencephaly and definition of the HPE2
620. Raybaud C, Girard N, Levrier O, Peretti-Viton P, Manera L, Farnarier P. critical region 2p21. Hum Mol Genet 1996;5:223–229.
Scizencephaly: correlation between the lobar topography of the cleft(s) 644. Gurrieri F, Trask BJ, van den Engh G, et al. Physical mapping of the
and absence of the septum pellucidum. Childs Nerv Syst 2001;17: holoprosencephaly critical region on chromosome 7q36. Nat Genet
217–222. 1993;3:247–251.
621. Blümcke I, Thom M, Aronica E, et al. The histopathological spectrum of 645. Brown SA, Warburton D, Brown LY, et al. Holoprosencephaly due to
focal cortical dysplasias: a revised classification system of the International mutations in ZIC2, a homologue of drosophila odd-paired. Nat Genet
League against Epilepsy. Epilepsia (in press). 1998;20:180–183.
622. Barkovich AJ, Kuzniecky RI, Dobyns WB, Jackson GD, Becker LE, Evrard P. 646. Lehman NL, Zaleski DH, Sanger WG, Adickes ED. Holoprosencephaly
A classification scheme for malformations of cortical development. associated with an apparent isolated 2q37.1–2q37.3 deletion. Am J Med
Neuropediatrics 1996;27(2):59–63. Genet 2001;100:179–181.

Barkovich_Chap05.indd 557 5/6/2011 9:01:36 PM


558 Pediatric Neuroimaging

647. Ming J, Kaupas M, Roessler E, et al. Mutations in PATCHED-1, the 672. Coleman LT, McCubbin JP, Smith LJ, Reddihough DS, Gardner RJM.
receptor for SONIC HEDGEHOG, are associated with holoprosencephaly. Syntelencephaly presenting with spastic diplegia. Neuropediatrics
Hum Genet 2002;110:297–301. 2000;31:206–210.
648. Kamnasaran D, Chen C, Devriendt K, Mehta L, Cox DW. Defining a holo- 673. Pulitzer SB, Simon EM, Crombleholme TM, Golden JA. Prenatal MR find-
prosencephaly locus on human chromosome 14q13 and characterization ings of the middle interhemispheric variant of holoprosencephaly. Am J
of potential candidate genes. Genomics 2005;85:608–621. Neuroradiol 2004;25:1034–1036.
649. Roessler E, Du Y-Z, Mullor JL, et al. Loss-of-function mutations in the 674. Azoulay R, Fallet-Bianco C, Garel C, Grabar S, Kalifa G, Adamsbaum C.
human GLI2 gene are associated with pituitary anomalies and holopros- MRI of the olfactory bulbs and sulci in human fetuses. Pediatr Radiol
encephaly-like features. Proc Natl Acad Sci 2003;100:13424–13429. 2006;36:97–107.
650. Kato M, Nanba E, Akaboshi S, et al. Sonic hedgehog signal peptide muta- 675. Schneider JF, Floemer F. Maturation of the olfactory bulbs: MR imaging
tion in a patient with holoprosencephaly. Ann Neurol 2000;47:514–516. findings. Am J Neuroradiol 2009;30:1149–1152.
651. Ming JE, Muenke M. Multiple hits during early embryonic development: 676. Nishimura Y. Embryological study of nasal cavity development in human
digenic diseases and holoprosencephaly. Am J Hum Genet 2002;109: embryos with reference to congenital nostril atresia. Acta Anat (Basel)
1017–1032. 1993;147:140–144.
652. Belloni E, Muenke M, Roessler E, et al. Identification of sonic hedge- 677. Lin AE, Siebert JR, Graham JM, Jr. Central nervous system malformations
hog as a candidate gene responsible for holoprosencephaly. Nat Genet in the CHARGE association. Am J Med Genet 1990;37:304–310.
1996;14:353–356. 678. Blustajn J, Kirsch CFE, Panigrahy A, Netchine I. Olfactory anomalies in
653. Roessler E, Belloni E, Gaudenz K, et al. Mutations in the human sonic CHARGE syndrome: imaging findings of a potential major diagnostic cri-
hedgehog gene cause holoprosencephaly. Nat Genet 1996;14:357–360. terion. Am J Neuroradiol 2008;29:1266–1269.
654. Cohen MM, Jr. The hedgehog signaling network. Am J Med Genet 679. Olsen OE, Gjelland K, Reigstad H, Rosendahl K. Congenital absence of the
2003;123:5–28. nose: a case report and literature review. Pediatr Radiol 2001;31:225–232.
655. Marcorelles P, Loget P, Fallet-Bianco C, Roume J, Encha-Razavi F, Dele- 680. Belet N, Belet U, Tekat A, Kucukoduk U. Proboscis lateralis: radiological
zoide A. Unusual variant of holoprosencephaly in monosomy 13q. Pediatr evaluation. Pediatr Radiol 2002;32:99–101.
Dev Pathol 2002;5:170–178. 681. Albernaz VS, Castillo M, Mukherji SK, Ihmeidan IH. Congenital arhinia.
656. Brown L, Odent S, David V, et al. Holoprosencephaly due to mutations Am J Neuroradiol 1996;17:1312–1314.
in ZIC2: alanine tract expansion mutations may be caused by parental 682. Krol BJ, Hulka GF, Drake A. Congenital nasal pyriform aperture stenosis
somatic recombination. Hum Mol Genet 2001;10:791–796. in the monozygotic twin of a child with holoprosencephaly. Otolaryngol
657. Fernandes M, Hébert JM. The ups and downs of holoprosencephaly: dor- Head Neck Surg 1998;118:679–681.
sal versus ventral patterning forces. Clin Genet 2008;73:413–423. 683. Tavin E, Stecker E, Marion R. Nasal pyriform aperture stenosis and the hol-
658. Huang X, Litingtung Y, Chiang C. Ectopic sonic hedgehog signaling oprosencephaly spectrum. Int J Pediatr Otorhinolaryngol 1994;28:199–204.
impairs telencephalic dorsal midline development: implication for human 684. Truwit CL, Barkovich AJ, Grumbach MM. MR imaging of Kallmann syn-
holoprosencephaly. Hum Mol Genet 2007;16:1454–1468. drome: a disorder of the olfactory and genital systems caused by faulty
659. Warr N, Powles-Glover N, Chappell A, Robson J, Norris D, Arkell RM. neuronal migration. Am J Neuroradiol 1993;14:427–438.
Zic2-associated holoprosencephaly is caused by a transient defect in 685. de Morsier G. Études sur les dysraphies cranio-encephaliques. III. Agene-
the organizer region during gastrulation. Hum Molec Genet 2008;17: sie du septum lucidum avec malformation du tractus optique. La dysplasie
2986–2996. septo-optique. Schweiz Arch Neurol Neurochir Psychiatr 1956;77:267–292.
660. Simon EM, Hevner R, Pinter JD, et al. The middle interhemispheric vari- 686. Dattani MT, Martinez-Barbera JP, Thomas PO, et al. Mutations in the
ant of holoprosencephaly. Am J Neuroradiol 2002;23:151–156. homeobox gene HESX1/Hesx1 associated with septo-optic dysplasia in
661. Filly RA, Chinn DH, Callen PW. Alobar holoprosencephaly: ultrasonic human and mouse. Nat Genet 1998;19:125–133.
prenatal diagnosis. Radiology 1984;151:455–459. 687. Brodsky MC, Glasier CM. Optic nerve hypoplasia: clinical significance of
662. Nyberg DA, Mack LA, Bronstein A, Hirsch J, Pagan RO. Holoprosenceph- associated central nervous system abnormalities on magnetic resonance
aly: prenatal sonographic diagnosis. Am J Neuroradiol 1987;8:871–878. imaging. Arch Ophthalmol 1993;111:66–74.
663. Cohen MM, Jr. Perspectives on holoprosencephaly. I. Epidemiology, genet- 688. Ouvrier R, Billson F. Optic nerve hypoplasia: a review. J Child Neurol
ics, and syndromology. Teratology 1989;40:211–235. 1986;1:181–188.
664. Volpe JJ. Neural tube formation and prosencephalic development. In: 689. Shuman RM, Leech RW. Optic nerve hypoplasia: one part of a spectrum.
Volpe JJ, ed. Neurology of the Newborn, 5th ed. Philadelphia: Saunders, J Child Neurol 1986;1:180.
2008:3–50. 690. Stevens CA, Dobyns WB. Septo-optic dysplasia and amniotic bands: further
665. Cohen MM, Jr, Sulik KK. Perspectives on holoprosencephaly: Part II. evidence for a vascular pathogenesis. Am J Med Genet A 2004;125A:12–16.
Central nervous system, craniofacial anatomy, syndrome commentary, 691. Dattani M, Robinson I. HESX1 and Septo-Optic Dysplasia. Rev Endor
diagnostic approach, and experimental studies. J Craniofac Genet Dev Biol Metab Disord 2002;3:289–300.
1992;12:196–244. 692. Dattani MT, Robinson IC. The molecular basis for developmental disor-
666. Barkovich A, Simon E, Clegg N, Kinsman S, Hahn J. Analysis of the cere- ders of the pituitary gland in man. Clin Genet 2000;57:337–346.
bral cortex in holoprosencephaly with attention to the sylvian fissures. Am 693. Tanaka T, Gleeson JG. Genetics of brain development and malformation
J Neuroradiol 2002;23:143–150. syndromes. Curr Opin Pediatr 2000;12:523–528.
667. Plawner L, Delgado M, Miller V, et al. Neuroanatomy of holoprosenceph- 694. Kelberman D, Dattani M. Genetics of septo-optic dysplasia. Pituitary
aly as a predictor of function: beyond the face predicting the brain. Neu- 2007;10:393–407.
rology 2002;59:1058–1066. 695. Stanhope R, Preece MA, Brook CGD. Hypoplastic optic nerves and pitu-
668. Lewis A, Simon E, Barkovich A, et al. Middle interhemispheric variant of itary dysfunction: a spectrum of anatomical and endocrine abnormalities.
holoprosencephaly: a distinct cliniconeuroradiologic subtype. Neurology Arch Dis Child 1984;59:111–114.
2002;59:1860–1865. 696. Skarf B, Hoyt CS. Optic nerve hypoplasia in children: association with
669. Simon E, Hevner R, Pinter J, et al. The dorsal cyst in holoprosenceph- anomalies of the endocrine and CNS. Arch Ophthalmol 1984;102:62–67.
aly and the role of the thalamus in its formation. Neuroradiology 2001;43: 697. Izenberg N, Rosenblum M, Parks JS. The endocrine spectrum of septo-
787–791. optic dysplasia. Clin Pediatr 1984;23:632–636.
670. Cohen MM, Jr, Jirasek JE, Guzman RT, Gorlin RJ, Peterson MQ. Holo- 698. Hoyt WF, Kaplan SL, Grumbach MM, Glaser TS. Septo-optic dysplasia
prosencephaly and facial dysmorphia: nosology, etiology, and pathogen- and pituitary dwarfism (letter). Lancet 1970;1:893–894.
esis. Birth Defects 1971;7:125–135. 699. Arslanian SA, Rouzfus WE, Foley TP, Jr, Becker DJ. Hormonal, metabolic,
671. Barkovich A, Simon E, Glenn O, et al. MRI shows abnormal white matter and neuroradiologic abnormalities associated with septo-optic dysplasia.
maturation in classical holoprosencephaly. Neurology 2002;59:1968–1971. Acta Endocrinol 1984;107:282–288.

Barkovich_Chap05.indd 558 5/6/2011 9:01:36 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 559

700. Wilson DM, Enzmann DR, Hintz RL, Rosenfeld G. CT findings in 726. Styne DM. Neurologic manifestations of endocrine diseases. In: Berg BO,
septo-optic dysplasia: discordance between clinical and radiological find- ed. Neurologic Aspects of Pediatrics. Boston: Butterworth-Heinemann,
ings. Neuroradiology 1984;26:279–283. 1992:439–466.
701. Barkovich AJ, Norman D. Absence of septum pellucidum: a useful sign 727. Triulzi F, Scotti G, di Natale B, et al. Evidence of a congenital midline brain
in the diagnosis of congenital brain malformations. Am J Neuroradiol anomaly in pituitary dwarfs: a magnetic resonance imaging study in 101
1988;9:1107–1114. patients. Pediatrics 1994;93:409–416.
702. Riedl S, Vosahlo J, Battelino T, et al. Refining clinical phenotypes in septo- 728. Argyroupoulou MI, Kiortsis DN. MRI of the hypothalamic-pituitary axis
optic dysplasia based on MRI findings. Eur J Pediatr 2008;167:1269–1276. in children. Pediatr Radiol 2005;35:1045–1055.
703. Birkebaek NH, Patel L, Wright NB, et al. Endocrine status in patients with 729. Melo M, Marui S, Carvalho L, et al. Hormonal, pituitary magnetic reso-
optic nerve hypoplasia: relationship to midline central nervous system nance, LHX4 and HESX1 evaluation in patients with hypopituitarism and
abnormalities and appearance of the hypothalamic-pituitary axis on mag- ectopic posterior pituitary lobe. Clin Endocrinol (Oxf) 2007;66:95–102.
netic resonance imaging. J Clin Endocrinol Metab 2003;88:5281–5286. 730. Kelly WM, Kucharczyk W, Kucharczyk J, et al. Posterior pituitary ectopia:
704. Brodsky MC. Septo-optic dysplasia: a reappraisal. Semin Ophthalmol and MR feature of pituitary dwarfism. Am J Neuroradiol 1988;9:453–460.
1991;6:227–232. 731. Kuriowa T, Okabe Y, Hasuo K, Yasumori K, Mizushima A, Masuda K. MR
705. Belhocine O, André C, Kalifa G, Adamsbaum C. Does asymptomatic septal imaging of pituitary dwarfism. Am J Neuroradiol 1991;12:161–164.
agenesis exist? A review of 34 cases. Pediatr Radiol 2005;35:410–418. 732. Abrahams JJ, Trefelner E, Boulware SD. Idiopathic growth hormone defi-
706. Trandafir T, Lipot C, Froicu P. On a possible neural ridge origin of the ciency; MR findings in 35 patients. Am J Neuroradiol 1991;12:155–160.
adenohypophysis. Endocrinologie 1990;28:67–72. 733. Chen S, Leger J, Garel C, Hassan M, Czernichow P. Growth hormone
707. Müller F, O’Rahilly R. The human brain at stage 16, including the initial deficiency with ectopic neurohypophysis: anatomical variations and rela-
evagination of the neurohypophysis. Anat Embryol 1989;179:551–569. tionship between the visibility of the pituitary stalk asserted by magnetic
708. Ikeda H, Suzuki J, Sasano N, et al. The development and morphogenesis of resonance imaging and anterior pituitary function. J Clin Endocrinol
the human pituitary gland. Anat Embryol 1988;178:327–336. Metab 1999;84(7):2408–2413.
709. Scully K, Rosenfeld M. Pituitary development: regulartory codes in mam- 734. Kornreich L, Horev G, Lazar L, Schwarz M, Sulkes J, Pertzelan A. MR find-
malian organogenesis. Science 2002;295:2231–2235. ings in growth hormone deficiency: correlation with severity of hypopitu-
710. Rizzoti K, Lovell-Badge R. Early development of the pituitary gland: itarism. Am J Neuroradiol 1998;19:1495–1499.
induction and shaping of Rathke’s pouch. Rev Endocr Metab Disord 735. Moon W, Porto L, Lanfermann H, Weis R, Zanella F. Agenesis of internal
2005;6:161–172. carotid artery associated with congenital anterior hypopituitarism. Neuro-
711. Gleiberman AS, Fedtsova NG, Rosenfeld MG. Tissue interactions in the radiology 2002;44:138–142.
induction of anterior pituitary: role of the ventral diencephalon, mesen- 736. Böttner A, Keller E, Kratzsch J, et al. PROP1 mutations cause progressive
chyme, and notochord. Dev Biol 1999;213:340–353. deterioration of anterior pituitary function including adrenal insufficiency:
712. Kelberman D, Dattani M. Hypothalamic and pituitary development: novel a longitudinal analysis. J Clin Endocrinol Metab 2004;89:5256–5265.
insights into the aetiology. Eur J Endocrinol 2007;157(Suppl 1):S3–S14. 737. Deladoey J, Fluck C, Buyukgebiz A, et al. ‘Hot Spot’ in the PROP1 gene
713. Kelberman D, Dattani M. Role of transcription factors in midline central responsible for combined pituitary hormone deficiency. J Clin Endocrinol
nervous system and pituitary defects. Endocr Dev 2009;14:67–82. Metab 1999;84:1645–1650.
714. Monuki ES. The morphogen signaling network in forebrain development 738. Vallette-Kasic S, Barlier A, Teinturier C, et al. PROP1 gene screening in
and holoprosencephaly. J Neuropathol Exp Neurol 2007;66:566–575. patients with multiple pituitary hormone deficiency reveals two sites of
715. Sarnat HB, Flores-Sarnat L. Neuropathologic research strategies in holo- hypermutability and a high incidence of corticotroph deficiency. J Clin
prosencephaly. J Child Neurol 2001;16:918–931. Endocrinol Metab 2001;86:4529–4535.
716. Schwanzel-Fukuda M, Bick D, Pfaff DW. Luteinizing hormone-releasing 739. Halász Z, Toke J, Patocs A, et al. High prevalence of PROP1 gene mutations
hormone (LHRL)-expressing cells do not migrate normally in an inher- in Hungarian patients with childhood-onset combined anterior pituitary
ited hypogonadal (Kallmann) syndrome. Mol Brain Res 1989;6:311–326. hormone deficiency. Endocrine 2006;30:255–260.
717. Friede RL. Developmental Neuropathology, 2nd ed. Berlin: Springer-Verlag, 740. Voutetakis A, Argyropoulou M, Sertedaki A, et al. Pituitary magnetic
1989. resonance imaging in 15 patients with Prop1 gene mutations: pituitary
718. Lemire RJ, Loeser JD, Leech RW, Alvord EC. Normal and Abnormal Devel- enlargement may originate from the intermediate lobe. J Clin Endocrinol
opment of the Human Nervous System. Hagarstown: Harper and Row, Metab 2004;89:2200–2206.
1975. 741. do Amaral LLF, Ferreira RM, Ferreira NPFD, et al. Combined pituitary hor-
719. di Iorgi N, Secco A, Napoli F, Calandra E, Rossi A, Maghnie M. Devel- mone deficiency and PROP-1 mutation in two siblings: a distinct MR imag-
opmental abnormalities of the posterior pituitary gland. Endocr Dev ing pattern of pituitary enlargement. Am J Neuroradiol 2007;28:1369–1370.
2009;14:83–94. 742. Kallmann FJ, Schoenfeld WA, Barrera SE. The genetic aspects pf primary
720. Manara R, Citton V, Rosetto M, Padoan A, D’Avella D. Hypophyseal trip- eunuchoidism. Am J Ment Defic 1944;48:203–236.
lication: Case report and embryologic considerations. Am J Neuroradiol 743. Schwanzel-Fukuda M, Abraham S, Crossin KL, Edelman GM, Pfaff DW.
2009;30:1328–1329. Immunocytochemical demonstration of neural cell adhesion molecule
721. Kollias SS, Ball WS, Jr, Prenger EC. Review of the embryologic develop- (NCAM) along the migration route of luteinizing hormone releasing hor-
ment of the pituitary gland and report of a case of hypophyseal duplica- mone (LHRH) neurons in mice. J Comp Neurol 1992;321:1–18.
tion detected by MRI. Neuroradiology 1995;37:3–12. 744. Valverde F, Heredia M, Santacana M. Characterization of neuronal cell
722. Harmon-Kérautret M, Ares GS, Demondion X, Rouland V, Franke J-P, varieties migrating from the olfactory epithelium during prenatal devel-
Pruvo J-P. Duplication of the pituitary gland in a newborn with median opment in the rat. Immunocytochemical study using antibodies against
cleft face syndrome and nasal teratoma. Pediatr Radiol 1998;28:290–292. olfactory marker protein and luteinizing hormone-releasing hormone.
723. Tagliavini F, Pilleri G. Mammillo-hypophyseal duplication (diplo- Dev Brain Res 1993;71:209–220.
mammillo-hypophysis). Acta Neuropathol 1986;69:38–44. 745. Franco B, Guioli S, Pragliola A, et al. A gene deleted in Kallmann’s syn-
724. Shroff M, Blaser S, Jay V, Chitayat D, Armstrong D. Basilar artery duplica- drome shares homology with neural cell adhesion and axonal pathfinding
tion associated with pituitary duplication: a new finding. Am J Neuroradiol molecules. Nature 1991;353:529–536.
2003;2003:956–961. 746. Dode C, Levilliers J, Dupont JM, et al. Loss-of-function mutations in
725. de Penna G, Pimenta M, Drummond J, et al. Duplication of the hypo- FGFR1 cause autosomal dominant Kallmann syndrome. Nat Genet
physis associated with precocious puberty: presentation of two cases and 2003;33:440–442.
review of pituitary embryogenesis. Arq Bras Endocrinol Metabol 2005;49: 747. Sato N, Katsumata N, Kagami M, et al. Clinical assessment and mutation
323–327. analysis of Kallmann Syndrome 1 (KAL1) and fibroblast growth factor

Barkovich_Chap05.indd 559 5/6/2011 9:01:37 PM


560 Pediatric Neuroimaging

receptor 1 (FGFR1, or KAL2) in five families and 18 sporadic patients. 772. Mafee MF, Goldberg MF. Persistent hyperplastic primary vitreous (PHPV):
J Clin Endocrinol Metab 2004;89(3):1079–1088. role of computed tomography and magnetic resonance. Radiol Clin North
748. Dode C, Teixeira L, Levilliers J, et al. Kallmann syndrome: mutations in the Am 1987;25:683–692.
genes encoding prokineticin-2 and prokineticin receptor-2. PLoS Genet 773. Shields JA, Shields CL, Scartozzi R. Survey of 1264 patients with orbital
2006;2:e175 (Electronic Article). tumors and simulating lesions: the 2002 Montgomery Lecture, part 1.
749. Kim H-G, Kurth I, Lan F, et al. Mutations in CHD7, encoding a Ophthalmology 2004;111(5):997–1008.
chromatin-remodeling protein, cause idiopathic hypogonadotropic 774. Haddad R, Font RL, Reeser F. Persistent hyperplastic primary vitreous:
hypogonadism and Kallmann syndrome. Am J Hum Genet 2008;83: a clinicopathologic study of 62 cases and review of the literature. Surv
511–519. Ophthalmol 1978;23(2):123–134.
750. Falardeau J, Chung WCJ, Beenken A, et al. Decreased FGF8 signaling 775. Goldberg MF, Mafee M. Computed tomography for diagnosis of persistent
causes deficiency of gonadotropin-releasing hormone in humans and hyperplastic primary vitreous (PHPV). Ophthalmology 1983;90:442–451.
mice. J Clin Invest 2008;118:2822–2831. 776. Chung EM, Specht CS, Schroeder JW. Pediatric orbit tumors and tumor-
751. Hardelin JP. Kallmann syndrome: towards molecular pathogenesis. Mol like lesions: neuroepithelial lesions of the ocular globe and optic nerve.
Cell Endocrinol 2001;179:75–81. Radiographics 2007;27:1159–1186.
752. Van Dop C, Burstein S, Conte FA, Grumbach MM. Isolated gonado- 777. Edward DP, Mafee M, Garcia-Valenzuela E, Weiss RA. Coats’ disease and
tropin deficiency in boys: clinical characteristics and growth. J Pediatr persistent hyperplastic primary vitreous. Role of MR imaging and CT.
1987;111:684–692. Radiol Clin North Am 1998;36:1119–1131.
753. Klingmüller D, Dewes W, Krahe T, Brecht G, Schweikert H-U. MR imaging 778. Mafee MF. Imaging of the globe. In: Valvassori GE, Mafee MF, Carter BL,
of the brain in patients with anosmia and hypothalamic hypogonadism eds. Imaging of Head and Neck. New York: Thieme, 1995:216–246.
(Kallmann’s syndrome). J Clin Endocrin Metab 1987;65:581–584. 779. Galluzzi P, Hadijstilianou T, Venturi C. Leukocoria: clinical and neurora-
754. Ozanics V, Jakobiec FA. Prenatal development of the eye and its adnexa. In: diological findings. Riv Neuroradiol 2004;17:61–78.
Jacobiec FA, ed. Ocular Anatomy, Embryology and Teratology. Philadelphia: 780. Gomori JM, Grossman RI, Steiner I. High-field magnetic resonance imag-
Harper and Row, 1982:11–96. ing of intracranial hematomas. Isr J Med Sci 1988;24(4–5):218–223.
755. O’Rahilly R. The early development of the eye in staged human embryos. 781. Anderson DR. The development of the trabecular meshwork and its
Contrib Embryol Carnegie Inst 1966;38:1–42. abnormality in primary infantile glaucoma. Trans Am Opthalmol 1981;79:
756. Mann IC. The Development of the Human Eye. London: Cambridge 458–485.
University Press, 1964. 782. Yanoff M, Fine BS. Ocular Pathology, 4th ed. St Louis: Mosby-Year Book,
757. Hever AM, Williamson KA, van Heyningen V. Developmental mal- 1996.
formations of the eye: the role of PAX6, SOX2 and OTX2. Clin Genet 783. Lavanya R, Teo L, Friedman DS, et al. Comparison of anterior chamber
2006;69:459–470. depth measurements using the IOLMaster, scanning peripheral anterior
758. Mihelec M, St Heaps L, Flaherty M, et al. Chromosomal rearrangements chamber depth analyser, and anterior segment optical coherence tomog-
and novel genes in disorders of eye development, cataract and glaucoma. raphy. Br J Ophthalmol 2007;91(8):1023–1026.
Twin Res Hum Genet 2008;11:412–421. 784. Plasilova M, Ferakova E, Kadasi L, et al. Linkage of autosomal recessive
759. Verma AS, Fitzpatrick D. Anophthalmia and microphthalmia. Orphanet J primary congenital glaucoma to the GLC3A locus in Roms (gypsies) from
Rare Dis 2007;2:47(Electronic Journal). Slovakia. Hum Hered 1998;48:30–33.
760. Smith CG, Gallie BL, Morin JD. Normal and abnormal development of 785. Choudhary AK, Jha B. Imaging findings of congenital glaucoma in Opitz
the eye. In: Cawford JS, Morin JD, eds. The Eye in Childhood. New York: syndrome. Am J Neuroradiol 2008;29:1003–1005.
Grune and Stratton, 1983:1–17. 786. Sherman JL, McLean IW, Brallier DR. Coats’ disease: CT-pathologic cor-
761. Morini F, Pacilli M, Spitz L. Bilateral anophthalmia and esophageal atre- relation in two cases. Radiology 1983;146:77–78.
sia: report of a new patient and review of the literature. Am J Med Genet 787. Ball WS, Jr, Kulwin DR. The eye and orbit. In: Ball WS, Jr, ed. Pediatric
2005;132A:60–62. Neuroradiology. Philadelphia: Lippincott-Raven, 1997:565–606.
762. Fantes J, Ragge NK, Lynch S-A, et al. Mutations in SOX2 cause anophthal- 788. Lumsden A, Krumlauf R. Patterning the vertebrate neuraxis. Science
mia. Nat Genet 2003;33:461–463. 1996;274:1009–1115.
763. Bridge H, Cowey A, Ragge N, Watkins K. Imaging studies in congenital 789. Lim Y, Golden JA. Patterning the developing diencephalon. Brain Res Rev
anophthalmia reveal preservation of brain architecture in “visual” cortex. 2007;53:17–26.
Brain 2009;132:3467–33480. 790. Nakamura H, Katahira T, Matsunaga E, Sato K. Isthmus organizer
764. Bilaniuk LT, Farber M. Imaging of developmental anomalies of the eye for midbrain and hindbrain development. Brain Res Brain Res Rev
and orbit. Am J Neuroradiol 1992;13:793–803. 2005;49(2):120–126.
765. Bardakjian T, Weiss A, Schneider AS. Anophthalmia/micropthalmia over- 791. Broccoli V, Boncinelli E, Wurst W. The caudal limit of Otx2 expression
view. Gene Rev 2007; http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi? positions the isthmic organizer. Nature 1999;401:164–168.
book=gene&part=anophthalmia-ov. Accessed August 21, 2009. 792. Wurst W, Bally-Cuif L. Neural plate patterning: upstream and downstream
766. Azuma N, Yamaguchi Y, Handa H, et al. Mutations of the PAX6 gene of the isthmic organizer. Nat Rev Neurosci 2001;2:99–108.
detected in patients with a variety of optic-nerve malformations. Am J 793. Sotelo C. Cellular and genetic regulation of the development of the cer-
Hum Genet 2003;72:1565–1570. ebellar system. Prog Neurobiol 2004;72:295–339.
767. Asai-Coakwell M, French CR, Berry KM, et al. GDF6, a novel locus 794. Tanabe Y, Jessell TM. Diversity and pattern in the developing spinal cord.
for a spectrum of ocular developmental anomalies. Am J Hum Genet Science 1996;274:1115–1123.
2007;80:306–315. 795. Gaufo GO, Wu S, Capecchi MR. Contribution of Hox genes to the diver-
768. Mafee MF, Jampol LM, Langer BG, Tso M. Computed tomography of sity of the hindbrain sensory system. Development 2004;131:1259–1266.
optic nerve colobomas, morning glory anomaly, and colobomatous cyst. 796. Ye W, Shimamura K, Rubenstein JLR, Hynes MA, Rosenthal A. FGF and
Radiol Clin North Am 1987;25:693–699. Shh signals control dopaminergic and serotonergic cell fate in the anterior
769. Gregory-Evans CY, Williams MJ, Halford S, Gregory-Evans K. Ocular neural plate. Cell 1998;93(5):755–766.
coloboma: a reassessment in the age of molecular neuroscience. J Med 797. Canning CA, Lee L, Irving C, Mason I, Jones CM. Sustained interactive
Genet 2004;41:881–891. Wnt and FGF signaling is required to maintain isthmic identity. Dev Biol
770. Murphy BL, Griffin JF. Optic nerve coloboma (morning glory syndrome): 2007;305:276–286.
CT findings. Radiology 1994;191:59–61. 798. Chizhikov VV, Lindgren AG, Currle D, Rose M, Monuki ES, Millen KJ.
771. Simmons JD, LaMasters D, Char D. Computed tomography of ocular The roof plate regulates cerebellar cell-type specification and prolifera-
colobomas. Am J Roentgenol 1983;141:1223–1226. tion. Development 2006;133:2793–2804.

Barkovich_Chap05.indd 560 5/6/2011 9:01:37 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 561

799. O’Rahilly R, Muller F. The Embryonic Human Brain. An Atlas of 823. von Knebel Doeberitz C, Sievers J, Sadler M, et al. Destruction of meningeal
Developmental Stages. New York: Wiley-Liss, 1994. cells over the newborn hamster cerebellum with 6-hydroxydopamine pre-
800. Lin JC, Cai L, Cepko CL. The external granule layer of the developing vents foleation and lamination in the rostral cerebellum. Neuroscience
chick cerebellum generates granule cells and cells of the isthmus and ros- 1986;17:409–426.
tral hindbrain. J Neurosci 2001;21:159–168. 824. Aldinger KA, Lehmann OJ, Hudgins L, et al. FOXC1 is required for normal
801. Goldowitz D, Hamre K. The cells and molecules that make a cerebellum. cerebellar development and is a major contributor to chromosome 6p25.3
Trends Neurosci 1998;21:375–382. Dandy-Walker malformation. Nat Genet 2009;41:1037–1042.
802. Salinas P, Fletcher C, Copeland N, Jenkins N, Nusse R. Maintenance of 825. Bonnevie K, Brodal A. Hereditary hydrocephalus in the house mouse. IV.
Wnt-3 expression in Purkinje cells of the mouse cerebellum depends on The development of cerebellar anomalies during fetal life with notes on
interactions with granule cells. Development 1994;120:1277–1286. the normal development of the mouse cerebellum. Lkr Norske Vidensk
803. Bloch-Gallego E, Causeret F, Ezan F, Backer S, Hidalgo-Sanchez M. Devel- Akad Oslo 1 Matj-nat KI 1946;4:4–60.
opment of precerebellar nuclei: instructive factors and intracellular medi- 826. Calabrò F, Arcuri T, Jinkins JR. Blake’s pouch cyst: an entity within the
ators in neuronal migration, survival and axon pathfinding. Brain Res Rev Dandy-Walker continuum. Neuroradiology 2000;42:290–295.
2005;49:253–266. 827. Tortori-Donati P, Fondelli M, Rossi A, Carini S. Cystic malformations
804. Bourrat F, Sotelo C. Early development of the rat precerebellar system: of the posterior cranial fossa originating from a deficit of the posterior
migratory routes, selective aggregation and neuritic differentiation of the membranous area. Mega cisterna magna and persisting Blake’s pouch: two
inferior olive and lateral reticular nucleus neurons. An overview. Arch Ital separate entities. Childs Nerv Syst 1996;12:303–308.
Biol 1990;128:151–170. 828. Robinson AJ, Goldstein R. The cisterna magna septa: vestigial remnants of
805. Kawauchi D, Taniguchi H, Watanabe H, Saito T, Murakami F. Direct Blake’s pouch and a potential new marker for normal development of the
visualization of nucleogenesis by precerebellar neurons: involvement rhombencephalon. J Ultrasound Med 2007;26:83–95.
of ventricle-directed, radial fibre-associated migration. Development 829. Demaerel P. Abnormalities of cerebellar foliation and fissuration: classifi-
2006;133:1113–1123. cation, neurogenetics and clinicoradiological correlations. Neuroradiology
806. Wingate RJ, Hatten ME. The role of the rhombic lip in avian cerebellum 2002;44:639–646.
development. Development. 1999;126:4395–4404. 830. Parisi MA, Dobyns WB. Human malformations of the midbrain and
807. Yamada M, Terao M, Terashima T, et al. Origin of climbing fiber hindbrain: review and proposed classification scheme. Mol Genet Metab
neurons and their developmental dependence on Ptf1a. J Neurosci 2003;80:36–53.
2007;27(41):10924–10934. 831. Lancioni A, Pizzo M, Fontanella B, et al. Lack of Mid1, the Mouse Ortholog
808. Fink AJ, Englund C, Daza RA, et al. Development of the deep cerebel- of the Opitz Syndrome Gene, Causes Abnormal Development of the Ante-
lar nuclei: transcription factors and cell migration from the rhombic lip. rior Cerebellar Vermis. J Neurosci 2010;30(8):2880–2887.
J Neurosci 2006;26(11):3066–3076. 832. Bosley TM, Alorainy IA, Salih MA, et al. The clinical spectrum of homozy-
809. Rakic P, Sidman RL. Histogenesis of cortical layers in human cerebellum, gous HOXA1 mutations. Am J Med Genet A 2008;146A:1235–1240.
particularly the lamina dessicans. J Comp Neurol 1970;139:473–500. 833. Tischfield MA, Bosley TM, Salih MA, et al. Homozygous HOXA1 muta-
810. Komuro H, Yacubova E, Yacubova E, Rakic P. Mode and tempo of tan- tions disrupt human brainstem, inner ear, cardiovascular and cognitive
gential cell migration in the cerebellar external granular layer. J Neurosci development. Nat Genet 2005;37:1035–1037.
2001;21:527–540. 834. Sarnat HB, Benjamin DR, Siebert JR, Kletter GB, Cheyette SR. Agenesis of
811. Karam S, Burrows R, Logan C, Koblar S, Pasquale E, Bothwell M. Eph the mesencephalon and metencephalon with cerebellar hypoplasia: puta-
receptors and ephrins in the developing chick cerebellum: relationship to tive mutation in the EN2 gene – report of 2 cases in early infancy. Pediatr
sagittal patterning and granule cell migration. J Neurosci 2000;20:6488– Dev Pathol 2002;5:54–68.
6500. 835. Mamourian A, Miller G. Neonatal pontomedullary disconnection with
812. Lin JC, Cepko CL. Granule cell raphes and parasagittal domains of aplasia or destruction of the lower brain stem: a case of pontoneocerebel-
Purkinje cells: complementary patterns in the developing chick cerebel- lar hypoplasia? Am J Neuroradiol 1994;15:1483–1485.
lum. J Neurosci 1998;18:9342–9353. 836. Bednarek N, Scavarda D, Mesmin F, Sabouraud P, Motte J, Morville P.
813. Moore KL, Persaud TVN. The Developing Human: Clinically Orientated Midbrain disconnection: An aetiology of severe central neonatal hypoto-
Embryology, 6th ed. Philadelphia: W B Saunders, 1998. nia. Eur J Paediatr Neurol 2005;9(6):419–422.
814. Moore CA. The Development of Mossy Fibres and Climbing Fibres: Embry- 837. Poretti A, Boltshauser E, Plecko B. Brainstem disconnection: case report
onic and Postnatal Features. New York: Alan R Liss, 1987. and review of the literature. Neuropediatrics 2007;38:210–212.
815. Sarnat HB, Menkes JH. How to construct a neural tube. J Child Neurol 838. Barth PG, de Vries L, Nikkels PG, Troost D. Congenital brainstem dis-
2000;15:110–124. connection associated with a syrinx of the brainstem. Neuropediatrics
816. Oberdick J, Baader S, Schilling K. From zebra stripes to postal zones: deci- 2008;39:1–7.
phering patterns of gene expression in the cerebellum. Trends Neurosci 839. Okumura A, Lee T, Shimojima K, et al. Brainstem disconnection associ-
1998;21:383–390. ated with nodular heterotopia and proatlantal arteries. Am J Med Genet A
817. Machold R, Fishell G. Math1 is expressed in temporally discrete pools of 2009;149A:2479–2483.
cerebellar rhombic lip neural progenitors. Neuron 2005;48:17–24. 840. Hoshino M, Nakamura S, Mori K, et al. Ptf1a, a bHLH transcriptional
818. Ilijic E, Guidotti A, Mugnaini E. Moving up or moving down? Malpo- gene, defines GABAergic neuronal fates in cerebellum. Neuron 2005;47(2):
sitioned cerebellar unipolar brush cells in reeler mouse. Neuroscience 201–213.
2005;136:633–647. 841. Sellick GS, Barker KT, Stolte-Dijkstra I, et al. Mutations in PTF1A cause
819. Trabousli EI. Congenital abnormalities of cranial nerve development: pancreatic and cerebellar agenesis. Nat Genet 2004;36(12):1301–1305.
overview, molecular mechanisms and further evidence of heterogeneity 842. Bolduc ME, Limperopoulos C. Neurodevelopmental outcomes in children
and complexity of syndromes with congenital limitation of eye move- with cerebellar malformations: a systemic review. Dev Med Child Neurol
ments. Trans Am Ophthalomol Soc 2004;102:373–390. 2009;51:256–267.
820. Pattyn A, Hirsch M, Goridis C, Brunet JF. Control of hindbrain motor 843. Boltshauser E, Steinlin M, Martin E, Deonna T. Unilateral cerebellar apla-
neuron differentiation by the homeobox gene Phox2b. Development sia. Neuropediatrics 1996;27:50–53.
2000;127(7):1349–1358. 844. Scuotto A, Accardo C, De Martino A, Guarracino A. Unilateral cer-
821. Grishkat H, Eisenman L. Development of the spinocerebellar projections ebellar aplasia. Report of an asymptomatic adult case. Riv Neuroradiol
in the prenatal mouse. J Comp Neurol 1995;363:93–108. 2001;19:659–664.
822. Voogd J, Glickstein M. The anatomy of the cerebellum. Trends Neurosci 845. Poretti A, Prayer D, Boltshauser E. Morphological spectrum of prenatal
1998;21:370–375. cerebellar disruptions. Eur J Paediatr Neurol 2009;13:397–407.

Barkovich_Chap05.indd 561 5/6/2011 9:01:37 PM


562 Pediatric Neuroimaging

846. Sener RN, Jinkins JR. Subtotal agenesis of the cerebellum in an adult. 871. Truwit CL, Barkovich AJ, Shanahan R, Maroldo TV. MR imaging of
Neuroradiology 1993;35:286–287. rhombencephalosyanpsis: report of three cases and review of the litera-
847. Gardner RJM, Colemen LT, Mitchell LA, et al. Near total absence of the ture. Am J Neuroradiol 1991;12:957–965.
cerebellum. Neuropediatrics 2001;32:62–68. 872. Widjaja E, Blaser S, Raybaud C. Diffusion tensor imaging of midline pos-
848. Arts WFM, Hofstee Y, Drejer GF, Beverstock GC, Oosterwijk JC. Cerebel- terior fossa malformations. Pediatr Radiol 2006;36(6):510–517.
lar and brainstem hypoplasia in a child with a partial monosom for the 873. Barkovich AJ, Kjos BO, Norman D, Edwards MSB. Revised classification
short arm of chromosome 5 and partial trisomy for the short arm of chro- of posterior fossa cysts and cyst-like malformations based on results of
mosome 10. Neuropediatrics 1995;26:41–44. multiplanar MR imaging. Am J Neuroradiol 1989;10:977–988.
849. Illarioshkin SN, Tanaka H, Markova ED, Nikolskaya NN, Ivanova- 874. Raimondi AJ, Sato K, Shimoji T. The Dandy-Walker Syndrome. Basel:
Smolenskaya IA, Tsuji S. X-linked nonprogressive congenital cerebellar Karger press, 1984.
hypoplasia: clinical description and mapping to chromosome Xq. Ann 875. Raybaud C. Cystic malformations of the posterior fossa – abnormalities
Neurol 1996;40:75–83. associated with development of the roof of the fourth ventricle and adja-
850. Limperopoulos C, du Plessis AJ. Disorders of cerebellar growth and devel- cent meningeal structures. J Neuroradiol 1982;9:103–133.
opment. Curr Opin Pediatr 2006;18:621–627. 876. Hart MN, Malamud N, Ellis WG. The Dandy-Walker syndrome. A clinico-
851. Garel C. MRI of the Fetal Brain. Normal Development and Cerebral Pathol- pathological study based on 28 cases. Neurology 1972;22:771–780.
ogies. Berlin: Springer, 2004. 877. Grinberg I, Northrup H, Ardinger H, Prasad C, Dobyns WB, Millen KJ.
852. Al-Baradie R, Yamada K, St Hilaire C, et al. Duane radial ray syndrome Heterozygous deletion of the linked genes ZIC1 and ZIC4 is involved in
(Okihiro syndrome) maps to 20q13 and results from mutations in SALL4, Dandy-Walker malformation. Nat Genet 2004;36:1053–1055.
a new member of the SAL family. Am J Hum Genet 2002;71:1195–1199. 878. Bindal AK, Storrs BB, McLone DG. Management of the Dandy-Walker
853. Michielse CB, Bhat M, Brady A, et al. Refinement of the locus for heredi- syndrome. Pediatr Neurosci 1990–91;16:163–169.
tary congenital facial palsy on chromosome 3q21 in two unrelated families 879. Osenbach RK, Menezes AH. Diagnosis and management of the Dandy-
and screening of positional candidate genes. Eur J Hum Genet 2006;14: Walker malformation: 30 years of experience. Pediatr Neurosurg
1306–1312. 1992;18:179–189.
854. Möbius PJ. Über angeborene doppelseitige Abducens-Facialis-Lahmung. 880. Boddaert N, Klein O, Ferguson N, et al. Intellectual prognosis of the
Müchener medizinische Wochenschrift 1888;35:91–94. Dandy-Walker malformation in children: the importance of vermian
855. Towfighi J, Marks K, Palmer E, et al. Mobius syndrome. Neuropathologic lobulation. Neuroradiology 2003;45:320–324.
observations. Acta Neuropathol (Berl) 1979;48:11–17. 881. Maria BL, Zinreich SJ, Carson BC, Rosenbaum AE, Freeman JM. Dandy-
856. Huang HT, Hwang CW, Lai PH, Chen CC. Möbius syndrome as a syn- Walker syndrome revisited. Pediatr Neurosci 1987;13:45–51.
drome of rhombencephalic maldevelopment: a case report. Pediatr Neo- 882. Golden JA, Rorke LB, Bruce DA. Dandy-Walker syndrome and associated
natol 2009;50:36–38. anomalies. Pediatr Neurosci 1987;13:38–44.
857. Verzijl HT, Valk J, de Vries R, Padberg GW. Radiologic evidence for absence 883. Klein O, Pierre-Kahn A, Boddaert N, Parisot D, Brunelle F. Dandy-
of the facial nerve in Möbius syndrome. Neurology 2005;64:849–855. Walker malformation: prenatal diagnosis and prognosis. Childs Nerv Syst
858. Pedraza S, Gámez J, Rovira A, et al. MRI findings in Möbius syndrome: 2003;19:484–489.
correlation with clinical features. Neurology 2000;55:1058–1060. 884. Masdeu JC, Dobben GD, Azar-Kia B. Dandy-Walker syndrome stud-
859. Obersteiner H. Ein Kleinhirn ohne Wurm. Arb Neurol Inst (Wien) ied by computed tomography and pneumoencephalography. Radiology
1914;21:124–136. 1983;147:109–114.
860. Romanengo M, Tortori-Donati P, Di Rocco M. Rhombencephalosynapsis 885. Cinalli G, Vinikoff L, Zerah M, Renier D, Pierre-Kahn A. Dandy-Walker
with facial anomalies and probably autosomal recessive inheritance: a case malformation associated with syringomyelia. J Neurosurg 1997;86:571.
report. Clin Genet 1997;52:184–186. 886. Lhermitte J, Duclos P. Sur un ganglioneurome diffus du cortex du cervelet.
861. Gomy I, Heck B, Santos AC, et al. Two new Brazilian patients with Gomez- Bull Assoc Fr Étude Cancer 1920;9:99–107.
Lopez-Hernandez syndrome. Am J Med Genet 2008;146A:649–657. 887. Milbouw G, Born JD, Martin D, et al. Clinical and radiological aspects of
862. Brocks D, Irons M, Sadeghi-Najad A, McCauley R, Wheeler P. Gomez- dysplastic gangliocytoma (Lhermitte-Duclos disease): report of two cases
Lopez-Hernandez syndrome: expansion of the phenotype. Am J Med and review of the literature. Neurosurgery 1988;22:124–128.
Genet 2000;94:405–408. 888. Roski RA, Roessmann U, Spetzler RF, Kaufman B, Nulsen FE. Clinical
863. Gomez MR. Cerebellotrigeminal and focal dermal dysplasia: a newly rec- and pathological study of dysplastic gangliocytoma. J Neurosurg 1981;55:
ognized neurocutaneous syndrome. Brain Dev 1979;1:253–256. 318–321.
864. Lopez-Hernandez A. Craniosynostosis, ataxia, trigeminal anesthesia and 889. Reeder RF, Saunders RL, Roberts DW, Fratkin JD, Cromwell LD. MRI in
parietal alopecia with pons-vermis fusion anomaly (atresia of the fourth the diagnosis and treatment of Lhermitte-Duclos disease (dysplastic gan-
ventricle). Neuropediatrics 1982;13:99–102. gliocytoma of the cerebellum). Neurosurgery 1988;23:240–245.
865. Pasquier L, Marcorelles P, Loget P, et al. Rhombencephalosynapsis and 890. Kulkantrakorn K, Awwad EE, Levy B, et al. MRI in Lhermitte-Duclos dis-
related anomalies: a neuropathological study of 40 fetal cases. Acta Neuro- ease. Neurology 1997;48:725–731.
pathol 2009;117:185–200. 891. Ambler M, Pogacar S, Sidman R. Lhermitte-Duclos disease (granule cell
866. Elliott R, Harter DH. Rhombencephalosynapsis associated with auto- hypertrophy of the cerebellum): pathological analysis of the first familial
somal dominant polycystic kidney disease Type 1. J Neurosurg Pediatr case. J Neuropathol Exp Neurol 1969;28:622–647.
2008;2:435–437. 892. Nelen MR, Padberg GW, Peeters EA, et al. Localization of the gene for
867. Napolitano M, Righini A, Zirpoli S, Rustico M, Nicolini U, Triulzi F. Cowden disease to chromosome 10q22-23. Nat Genet 1996;13:114–116.
Prenatal magnetic resonance imaging of rhombencephalosynapsis and 893. Wells G, Lasner T, Yousem D, Zager E. Lhermitte-Duclose disease and
associated brain anomalies: report of 3 cases. J Comput Assist Tomogr Cowden’s syndrome in an adolescent patient. Case Report. J Neurosurg
2004;28:762–765. 1994;81:133–136.
868. Tan TY, McGillivray G, Goergen SK, White SM. Prenatal magnetic reso- 894. Eng C, Murday V, Seal S, et al. Cowden syndrome and Lhermitte-Duclos
nance imaging in Gomez-Lopez-Hernandez syndrome and review of the disease in a family: a single genetic syndrome with pleiotropy? J Med Genet
literature. Am J Med Genet 2005;138A:369–373. 1994;31:458–461.
869. Toelle S, Yalcinkaya C, Kocer N, et al. Rhombencephalosynapsis: clini- 895. Nelen M, Padberg G, Peeters E, et al. Germline mutations in the PTEN/
cal findings and neuroimaging in 9 children. Neuropediatrics 2002;33: MMAC1 gene in patients with Cowden disease. Nat Genet 1996;13:
209–214. 114–116.
870. Poretti A, Alber FD, Bürki S, Toelle S, Boltshauser E. Cognitive outcome 896. Padberg GW, Schot JDL, Vielvoye GJ, Bots GT, de Beer FC. Lhermitte-
in children with rhombencephalosynapsis. Eur J Paediatr Neurol 2009;13: Duclos disease and Cowden disease: a single phakomatosis. Ann Neurol
28–33. 1991;29:517–523.

Barkovich_Chap05.indd 562 5/6/2011 9:01:37 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 563

897. Smith RR, Grossman RI, Goldberg H, et al. MR imaging of Lhermitte-Duclos 921. Valente EM, Marsh SE, Castori M, et al. Distinguishing the four genetic causes
disease: a case report. Am J Neuroradiol 1989;10:187–189. of Jouberts syndrome-related disorders. Ann Neurol 2005;57:513–519.
898. Meltzer CC, Smirniotopoulos JG, Jones RV. The striated cerebellum: an 922. Sun T, Collura RV, Ruvolo M, Walsh CA. Genomic and evolutionary
MR imaging sign in Lhermitte-Duclos disease (dysplastic gangliocytoma). analyses of asymmetrically expressed genes in human fetal left and right
Radiology 1995;194:699–703. cerebral cortex. Cereb Cortex 2006;16(Suppl 1):i18–i25.
899. Awwad EE, Levy E, Martin DS, Merenda GO. Atypical MR appearance of 923. Yachnis AT, Rorke LB. Neuropathology of Joubert syndrome. J Child Neu-
Lhermitte-Duclos disease with contrast enhancement. Am J Neuroradiol rol 1999;14:655–659.
1995;16:1719–1720. 924. Saito Y, Ito M, Ozawa Y, et al. Changes of neurotransmitters in the brain-
900. Thomas B, Krishnamoorthy T, Radhakrishnan VV, Kesavadas C. Advanced stem of patients with respiratory pattern disorders during childhood.
MR imaging in Lhermitte-Duclos disease: moving closer to pathology and Neuropediatrics 1999;30:133–140.
pathophysiology. Neuroradiology 2007;49:733–738. 925. Saleem SN, Zaki MS. Role of MR imaging in prenatal diagnosis of preg-
901. Moonis G, Ibrahim M, Melhem ER. Diffusion weighted MRI in Lhermitte- nancies at risk for Joubert Syndrome and related cerebellar disorders. Am
Duclos disease: report of two cases. Neuroradiology 2004;46:351–354. J Neuroradiol 2010;31(3):424–429.
902. Klisch J, Juengling F, Spreer J, et al. Lhermitte-Duclos disease: assessment 926. Dretakis EK. Familial idiopathic scoliosis associated with congenital
with MR imaging, positron emission tomography, single photon emission encephalopathy in three children of the same family. Acta Orthop Trauma-
CT, and MR spectroscopy. Am J Neuroradiol 2001;22:824–830. tol Hellenica 1970;22:51–55.
903. Nagaraja S, Powell T, Griffiths PD, Wilkinson ID. MR imaging adn 927. Dretakis EK, Kondoyannis PN. Congenital scoliosis associated with
spectroscopy in Lhermitte-Duclos disease Neuroradiology 2004;46(5): encephalopathy in five children of two families. J Bone Joint Surg
355–358. 1974;56A:1747–1750.
904. Najm J, Horn D, Wimplinger I, et al. Mutations of CASK cause an X-linked 928. Crisfield RJ. Scoliosis with progressive external ophthalmoplegia in four
brain malformation phenotype with microcephaly and hypoplasia of the siblings. J Bone Joint Surg 1974;56B:484–489.
brainstem and cerebellum. Nat Genet 2008;40:1065–1067. 929. Jen J, Coulin CJ, Bosley TM, et al. Familial horizontal gaze palsy with progres-
905. Hevner RF, Hodge RD, Daza RAM, Englund C. Transcription factors in sive scoliosis maps to chromosome 11q23–25. Neurology 2002;59(3):432–435.
glutamatergic neurogenesis: Conserved programs in neocortex, cerebel- 930. Jen JC, Chan W-M, Bosley TM, et al. Mutations in a human ROBO gene
lum, and adult hippocampus. Neurosci Res 2006;55(3):223–233. disrupt hindbrain axon pathway crossing and morphogenesis. Science
906. Joubert M, Eisenring JJ, Robb JP, Andermann F. Familial agenesis of the 2004;304(5676):1509–1513.
cerebellar vermis. A syndrome of episodic hyperpnea, abnormal eye 931. Bosley TM, Salih MAM, Jen JC, et al. Neurologic features of horizontal
movements, ataxia, and retardation. Neurology 1969;19:813–825. gaze palsy and progressive scoliosis with mutations in ROBO3. Neurology
907. King MD, Dudgeon J, Stephenson JBP. Joubert’s syndrome with retinal 2005;64(7):1196–1203.
dysplasia: neonatal tachypnea as the clue to a genetic brain-eye malforma- 932. Rossi A, Catala M, Biancheri R, Di Comite R, Tortori-Donati P. MR imag-
tion. Arch Dis Child 1984;59:709–718. ing of brain-stem hypoplasia in horizontal gaze palsy with progressive
908. Kendall B, Kingsley D, Lambert SR, Taylor D, Finn P. Joubert syndrome: scoliosis. Am J Neuroradiol 2004;25(6):1046–1048.
A clinical-radiological study. Neuroradiology 1990;31:502–506. 933. Haller S, Wetzel SG, Lütschg J. Functional MRI, DTI and neurophysiol-
909. Boltshauser E, Herdon M, Dumermuth G, Isler W. Joubert syndrome: ogy in horizontal gaze palsy with progressive scoliosis. Neuroradiology
clinical and polygraphic observations in a further case. Neuropediatrics 2008;50:453–459.
1981;12:181–191. 934. Sicotte NL, Salamon G, Shattuck DW, et al. Diffusion tensor MRI shows
910. Gleeson J, Keeler L, Parisi M, et al. Molar tooth sign of the midbrain abnormal brainstem crossing fibers associated with ROBO3 mutations.
junction: occurence in multiple distinct syndromes. Am J Med Genet Neurology 2006;67(3):519–521.
2004;125A:125–134. 935. Amoiridis G, Tzagournissakis M, Christodoulou P, et al. Patients with hor-
911. Satran D, Pierpont MEM, Dobyns WB. Cerebello-Oculo-Renal syndromes izontal gaze palsy and progressive scoliosis due to ROBO3 E319K muta-
including Arima, Senior-Löken and COACH syndromes: more than just tion have both uncrossed and crossed central nervous system pathways
variants of Joubert syndrome. Am J Med Genet 1999;86:459–469. and perform normally on neuropsychological testing. J Neurol Neurosurg
912. Badano JL, Mitsuma N, Beales PL, Katsanis N. The ciliopathies: an emerg- Psychiatry 2006;77:1047–1053.
ing class of human genetic disorders. Ann Rev Genomics Hum Genet 936. Gutowski NJ, Bosley TM, Engle EC. Workshop Report 110th ENMC
2006;7:125–148. International Workshop: The congenital cranial dysinnervation disorders
913. Louie CM, Gleeson JG. Genetic basis of Joubert syndrome and related (CCDDs). Neuromusc Disord 2003;13:573–578.
disorders of cerebellar development. Hum Mol Genet 2005;14(Suppl 937. Engle E. Applications of molecular genetics to the understanding of con-
2):R235–R242. genital ocular motility disorders. Ann NY Acad Sci 2002;956:55–63.
914. Valente EM, Brancati F, Silhavy JL, et al. AHI1 gene mutations cause spe- 938. Wang S, Zwaan J, Mullaney P, et al. Congenital fibrosis of the extraocular
cific forms of Joubert syndrome-related disorders. Ann Neurol 2006;59(3): muscles type 2 (CFOEM2), an inherited exotropic strabismus fixus, maps
527–534. to distal 11q13. Am J Hum Genet 1998;63:517–525.
915. Valente EM, Silhavy JL, Brancati F, et al. Mutations in CEP290, which 939. Lim KH, Engle EC, Demer JL. Abnormalities of the oculomotor nerve in
encodes a centrosomal protein, cause pleiotropic forms of Joubert syn- congenital fibrosis of the extraocular muscles and congenital oculomotor
drome. Nat Genet 2006;38:623–625. palsy. Invest Ophthalmol Vis Sci 2007;48(4):1601–1606.
916. Lagier-Tourenne C, Boltshauser E, Breivik N, et al. Homozygosity map- 940. Delleman JW, Oorthuys JWE, Bleeker-Wagemakers EM, ter Haar BGA,
ping of a third Joubert syndrome locus to 6q23. J Med Genet 2004;41: Ferguson JW. Orbital cyst in addition to congenital cerebral and focal der-
273–277. mal malformations: a new entity. Clin Genet 1984;25:470–472.
917. Cantagrel V, Silhavy J, Bielas S, et al. Mutations in the cilia gene ARL13B 941. Burgett R, Kawasaki A. Mesencephalic clefts and eye movement disorders.
lead to the classical form of Joubert syndrome. Am J Hum Genet Arch Ophthalmol 1997;115:824.
2008;83:170–179. 942. Lagreze W, Warner J, Zamani A, Gouras G, Koralnik I, Bienfang D. Mes-
918. Mykytyn K. Clinical variability in ciliary disorders. Nat Genet 2007;39: encephalic clefts with associated eye movement disorders. Arch Opthalmol
818–819. 1996;114:429–432.
919. Giordano L, Vignoli A, Pinelli L, et al. Joubert syndrome with bilateral 943. de la Monte S, Horowitz S, Larocque A, Richardson Jr E. Keyhole aqueduct
polymicrogyria: clinical and neuropathological findings in two brothers. syndrome. Arch Neurol 1986;43:926–929.
Am J Med Genet 2009;149A:1511–1515. 944. Savoiardo M, Minati L, Farina L, et al. Spontaneous intracranial hypoten-
920. Saar K, Al-Gazali L, Sztriha L, et al. Homozygosity mapping in families sion with deep brain swelling. Brain 2007;130:1884–1893.
with Joubert syndrome identifies a locus on chromosome 9q34.3 and evi- 945. Fredericks E, Van Nuis C. Diverticulum of the rostral cerebral aqueduct
dence for genetic heterogeneity. Am J Hum Genet 1999;65:1666–1671. with ocular dysfunctions. Arch Neurol 1967;16:32–36.

Barkovich_Chap05.indd 563 5/6/2011 9:01:37 PM


564 Pediatric Neuroimaging

946. Tavano A, Grasso R, Gagliardi C, et al. Disorders of cognitive and affec- 973. Cinalli G, Renier D, Sebag G, Sainte-Rose C, Arnaud E, Pierre-Kahn A.
tive development in cerebellar malformations. Brain 2007;130(10):2646– Chronic tonsillar herniation in Crouzon’s and Apert’s syndromes: the role of
2660. premature synostosis of the lambdoid suture. J Neurosurg 1995;83:575–582.
947. Ventura P, Presicci A, Perniola T, Campa MG, Margari L. Mental retarda- 974. Cinalli G, Spennato P, Sainte-Rose C, et al. Chiari malformation in cranio-
tion and epilepsy in patients with isolated cerebellar hypoplasia. J Child synostosis. Childs Nerv Syst 2005;21:889–901.
Neurol 2006;21(9):776–781. 975. Francis PM, Beals S, Rekate HL, Pittman HW, Manwaring K, Reiff J.
948. Doya K. Complementary roles of basal ganglia and cerebellum in learning Chronic tonsillar herniation and Crouzon’s syndrome. Pediatr Neurosurg
and motor control. Curr Opin Neurobiol 2000;10:732–739. 1992;18:202–206.
949. Poretti A, Leventer R, Cowan F, et al. Cerebellar cleft: a form of prenatal 976. Boltshauser E, Schmitt B, Wichmann W, Valavanis A, Sailer H, Yonekawa
cerebellar disruption. Neuropediatrics 2008;39:106–112. Y. Cerebellomedullary compression in recessive craniometaphyseal dys-
950. Soto-Ares G, Deries B, Delmaire C, Devisme L, Ruchoux MM, Pruvo J. plasia. Neuroradiology 1996;38:S193–S195.
Dysplasie du cortex cérébelleux: aspects en IRM et signification. J Radiol 977. Marden FA, Wippold FJ, II. MR imaging features of craniodiaphyseal dys-
2004;85:729–740. plasia. Pediatr Radiol 2004;34:167–170.
951. Takanashi J, Sugita K, Barkovich AJ, Takano H, Kohno Y. Partial midline 978. Nishimura G, Aoki K, Haga N, Hasegawa T. Syringohydromyelia in Hajdu-
fusion of the cerebellar hemispheres with vertical folia: a new cerebellar Cheney syndrome. Pediatr Radiol 1996;26:59–61.
malformation? Am J Neuroradiol 1999;20:1151–1153. 979. Emery JL, MacKenzie N. Medullo-cervical dislocation deformity (Chiari
952. Jackson JM, Sadove AM, Weaver DD, Edwards MK, Bull M. Unilateral II deformity) related to neurospinal dysraphism (myelomeningocele).
duplication of the cerebellar hemisphere and internal, middle, and exter- Brain 1973;96:155–162.
nal ear: a clinical case study. Plast Reconstr Surg 1990;86:550–553. 980. Banik R, Lin D, Miller NR. Prevalence of Chiari I malformation and
953. Barth PG, Majoie CB, Caan MWA, et al. Pontine tegmental cap dyspla- cerebellar ectopia in patients with pseudotumor cerebri. J Neurol Sci
sia: a novel brain malformation with a defect in axonal guidance. Brain 2006;247:71–75.
2007;130(9):2258–2266. 981. Atkinson JLD, Weinshenker BG, Miller GM, Piepgras DG, Mokri B.
954. Jissendi-Tchofo P, Doherty D, McGillivray G, et al. Pontine tegmental cap Acquired Chiari I malformation secondary to spontaneous spinal cere-
dysplasia: mr imaging and diffusion tensor imaging features of impaired brospinal fluid leakage and chronic intracranial hypotension syndrome in
axonal navigation. Am J Neuroradiol 2009;30:113–119. seven cases. J Neurosurg 1998;88:237–242.
955. Chiari H. Über Veränderungen des Kleinhirns infolge von Hydrocephalie 982. Fishman RA, Dillon WP. Dural enhancement and cerebral displacement
des Grosshirns. Deutsch Medizinische Wocherschrift 1891;17:1172–1175. secondary to intracranial hypotension. Neurology 1993;43:609–611.
956. Wu Y, Chin C, Chan K, Barkovich A, Ferreiro D. Pediatric Chiari I mal- 983. Couch JR. Spontaneous intracranial hypotension: the syndrome and its
formations: do clinical and radiologic features correlate? Neurology complications. Curr Treat Options Neurol 2008;10:3–11.
1999;53:1271–1276. 984. Schievink WI, Maya MM, Louy C, Moser FG, Tourje J. DIagnostic crite-
957. Steinbok P. Clinical features of Chiari I malformations. Childs Nerv Syst ria for spontaneous spinal CSF leakes and intracranial hypotension. Am J
2004:20:329–331. Neuroradiol 2008;29:853–856.
958. Aitken LA, Lindan CE, Sidney S, et al. Chiari type I malformation in a 985. Chumas PD, Armstrong DC, Drake JM, et al. Tonsillar herniation: the rule
pediatric population. Pediatr Neurol 2009;40:449–454. rather than the exception after lumboperitoneal shunting in the pediatric
959. Greenlee J, Donovan K, Hasan D, Menezes A. Chiari I malformation in population. J Neurosurg 1993;78:568–573.
the very young child: the spectrum of presenations and experience in 31 986. Hoffman HJ, Tucker WS. Cephalocranial disproportion. A complica-
children under age 6 years. Pediatrics 2002;110:1212–1219. tion of the treatment of hydrocephalus in children. Child’s Brain 1976;2:
960. Yoshimi A, Nomura K, Furune S. Sleep apnea syndrome associated with a 167–176.
type I Chiari malformation. Brain Dev 2002;24:49–51. 987. Barkovich AJ, Wippold FJ, Sherman JL, Citrin CM. Significance of cerebel-
961. Levitt P, Cohn M. Sleep apnea and the Chiari I malformation: case report. lar tonsillar ectopia on MR. Am J Neuroradiol 1986;7:795–799.
Neurosurgery 1988;23:508–510. 988. Mikulis DJ, Diaz O, Egglin TK, Sanchez R. Variance of the position of
962. Ruff M, Oakes W, Fisher S, Spock A. Sleep apnea and vocal cord paralysis the cerebellar tonsils with age: preliminary report. Radiology 1992;183:
secondary to type I Chiari malformation. Pediatrics 1987;80:231–234. 725–728.
963. Listernick R, Tomita T. Persistent crying in infancy as a presentation of 989. Elster AD, Chen MYM. Chiari I malformations: clinical and radiologic
Chiari type 1 malformation. J Pediatr 1991;118:567–569. reappraisal. Radiology 1992;183:347–353.
964. Albers FW, Ingels KJ. Otoneurological manifestations in Chiari I malfor- 990. Vu BT, Mikulis DJ, Armstrong D, Pron G. Normal position of the cerebel-
mation. J Laryngol Otol 1993;107:441–443. lar tonsils in relation to the foramen magnum in children. Paper Presented
965. Schady W, Metcalfe R, Butler P. The incidence of craniocervical bony at: American Society of Neuroradiology, 31st Annual Meeting, Vancouver,
anomalies in the adult Chiari malformation. J Neurol Sci 1987;82: BC, Canada, 1993.
193–203. 991. Tubbs R, McGirt M, Oakes W. Surgical experience in 130 pediatric patients
966. Milhorat TH, Chou MW, Trinidad EM, et al. Chiari I malformation rede- with Chiari I malformations. J Neurosurg 2003;99:291–296.
fined: clinical and radiographic findings for 364 symptomatic patients. 992. Wolpert S, Bhadelia R, Bogdan A, Cohen A. Chiari I malformations:
Neurosurgery 1999;44:1005–1017. assessment with phase-contrast velocity MR. Am J Neuroradiol 1994;15:
967. Stovner LJ, Bergan U, Nilsen G, Sjaastad O. Posterior cranial fossa dimen- 1299–1308.
sions in the Chiari I malformation: relation to pathogenesis and clinical 993. Bhadelia RA, Bogdan AR, Wolpert SM. Analysis of cerebrospinal fluid flow
presentation. Neuroradiology 1993;35:113–118. waveforms with gated phase-contrast MR velocity measurements. Am J
968. Vega A, Quintana F, Berciano J. Basichondrocranium anomalies in Neuroradiol 1995;16:389–400.
adult Chiari type I malformation: a morphometric study. J Neurol Sci 994. Heiss J, Patronas N, DeVroom H, et al. Elucidating the pathophysiology of
1990;99:137–145. syringomyelia. J Neurosurg 1999;91:553–562.
969. Menezes AH. Primary craniovertebral anomalies and the hindbrain her- 995. Ventureyra E, Aziz H, Vassilyadi M. The role of cine flow MRI in children
niation syndrome: data base analysis. Pediatr Neurosurg 1995;23:260–269. with Chiari I malformation. Childs Nerv Syst 2003;19:109–113.
970. Grabb PA, Mapstone TB, Oakes WJ. Ventral brain stem compression in 996. McGirt MJ, Nimjee SM, Floyd J, Bulsara KR, George TM. Correlation of
pediatric and young adult patients with Chiari I malformations. Neuro- cerebrospinal fluid flow dynamics and headache in Chiari I malformation.
surgery 1999;44:520–528. Neurosurgery 2005;56(4):716–721.
971. Caldarelli M, Novegno F, Di Rocco C. A late complication of CSF shunt- 997. Appleby A, Foster JB, Hankinson J, Hudgson P. The diagnosis and
ing: acquired Chiari I malformation. Childs Nerv Syst 2009;25:443–452. management of the Chiari anomalies in adult life. Brain 1969;91:
972. Mazzanti L, Ambrosetto P, Libri R, Pascarella R, Balsamo A, Tani G. 131–140.
Involvement of the skull base anf vault in chronic idiopathic hyperphos- 998. Banerji NR, Millar JHD. Chiari malformations presenting in adult life.
phatasia. Pediatr Radiol 1999;29:16–18. Brain 1974;97:157–168.

Barkovich_Chap05.indd 564 5/6/2011 9:01:37 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 565

999. Naidich TP, McLone DG, Fulling KH. The Chiari II malformation: Part 1023. Fleming A, Copp AJ. A genetic risk factor for mouse neural tube defects:
IV. The hindbrain deformity. Neuroradiology 1983;25:179–197. defining the embryonic basis. Hum Molec Genet 2000;9:575–581.
1000. Cleland J. Contribution to the study of spinal bifida, encephalocele, and 1024. O’Rahilly R, Müller F. The two sites of fusion of the neural folds and the
anencephalus. J Anat Physiol 1883;17:257–292. two neuropores in the human embryo. Teratology 2002;65:162–170.
1001. Rintoul N, Sutton L, Hubbard A, et al. A new look at myelomeningoceles: 1025. Gluckman TJ, George TM, McLone DG. Postneurulation rapid brain
functional level, vertebral level, shunting, and the implications for fetal growth represents a critical time for encephalocele formation: a chick
intervention. Pediatrics 2002;109:409–413. model. Pediatr Neurosurg 1996;25:130–136.
1002. Vandertop WP, Asai A, Hoffman HJ, et al. Surgical decompression for 1026. Marin-Padilla M. Morphogenesis of experimental encephalocele (cranio-
symptomatic Chiari II malformation in neonates with myelomeningo- schisis occulta). J Neurol Sci 1980;46:83–99.
cele. J Neurosurg 1992;77:541–544. 1027. Diebler C, Dulac O. Cephaloceles: clinical and neuroradiological appear-
1003. Worley G, Schuster JM, Oakes WJ. Survival at 5 years of a cohort of new- ance. Neuroradiology 1983;25:199–216.
born infants with myelomeningocele. Dev Med Child Neurol 1996;38: 1028. Yokota A, Matsukado Y, Fuwa I, Moroki K, Nagahiro S. Anterior basal
816–822. encephalocele of the neonatal and infantile period. Neurosurgery
1004. Talwar D, Baldwin M, Horbatt C. Epilepsy in children with meningomy- 1986;19:468–478.
elocele. Pediatr Neurol 1995;13:29–32. 1029. Sessions RB. Nasal dermal sinuses - new concepts and explanations.
1005. Copp AJ, Greene ND. Genetics and development of neural tube defects. Laryngoscope 1982;92 (Suppl 29):1–28.
J Pathol 2010;220:217–230. 1030. Couly GF, Coltey PM, Le Douarin NM. The developmental fate of the
1006. McLone DG, Knepper PA. The cause of Chiari II malformation: A unified cephalic mesoderm in quail-chick chimaras. Development 1992;114:
theory. Pediatr Neurosci 1989;15:1–12. 1–15.
1007. Bouchard S, Davey M, Rintoul N, Walsh D, Rorke L, Adzick N. Correction 1031. Noden DM, Trainor PA. Relations and interactions between cranial
of hindbrain herniation and anatomy of the vermis after in utero repair mesoderm and neural crest populations. J Anat 2005;207:575–601.
of myelomeningocele in sheep. J Pediatr Surg 2003;38(3):451–458. 1032. Yokota A, Kajiwara H, Kohchi M, Fuwa I, Wada H. Parietal cephalocele:
1008. Tulipan N, Sutton L, Bruner J, Cohen B, Johnson M, Adzick N. The effect clinical importance of its atretic form and associated malformations.
of intrauterine myelomeningocele repair of the incidence of shunt-de- J Neurosurg 1988;69:545–551.
pendent hydrocephalus. Pediatr Neurosurg 2003;38:27–33. 1033. Chapman PH, Swearingen B, Caviness VS. Subtorcular occipital enceph-
1009. Naidich TP, Pudlowski RM, Naidich JB. Computed tomographic signs of aloceles: anatomical considerations relevant to operative management.
the Chiari II malformation: Part II. Midbrain and cerebellum. Radiology J Neurosurg 1989;71:375–381.
1980;134:391–398. 1034. Simpson DA, David DJ, White J. Cephaloceles: treatment, outcome and
1010. Naidich TP, Pudlowski RM, Naidich JB, Gornish M, Rodriguez FJ. Com- antenatal diagnosis. Neurosurgery 1984;15:14–21.
puted tomographic signs of the Chiari II malformation: Part I. Skull and 1035. Groen RJM, van Ouwerkerk WJR. Cerebellar dermoid tumor and occipi-
dural partitions. Radiology 1980;134:64–71. tal meningocele in a monozygotic twin: clues to the embryogenesis of
1011. Naidich TP, Pudlowski RM, Naidich JB. Computed tomographic signs craniospinal dysraphism. Childs Nerv Syst 1995;11:414–417.
of the Chiari II malformation: Part III. Ventricles and cisterns. Radiology 1036. Lo BW, Kulkarni AV, Rutka JT, et al. Clinical predictors of developmen-
1980;134:657–663. tal outcome in patients with cephaloceles. J Neurosurg Pediatr 2008;2:
1012. Wong SK, Barkovich AJ, Callen AL, Filly RA. Supratentorial abnormali- 254–257.
ties in the Chiari II malformation: III. The interhemispheric cyst. J Ultra- 1037. Kotil K, Kilinc B, Bilge T. Diagnosis and management of large occipito-
sound Med 2009;28:999–1006. cervical cephaloceles: a 10-year experience. Pediatr Neurosurg 2008;44:
1013. Filly MR, Filly RA, Barkovich AJ, Goldstein RB. Supratentorial abnor- 193–198.
malities in the Chiari II malformation, IV: the too-far-back ventricle. 1038. Bartels R, Merx J, van Overbeeke J. Falcine sinus and occipital encepha-
J Ultrasound Med 2010;29:243–248. locele: a magnetic resonance venography study. J Neurosurg 1998;89:
1014. Wolpert SM, Anderson M, Scott RM, Kwan ES, Runge VM. The Chiari 738–741.
II malformation: MR imaging evaluation. Am J Neuroradiol 1987;8:783– 1039. Martinez-Lage JF, Poza M, Sola J, et al. The child with a cephalocele: etiol-
791. ogy, neuroimaging, and outcome. Childs Nerv Syst 1996;12:540–550.
1015. Vachha B, Adams RC, Rollins NK. Limbic tract anomalies in pedi- 1040. Maas KP, Barkovich AJ, Dong L, Edwards MSB, Piecuch RE, Charlton V.
atric myelomeningocele and Chiari II malformation: anatomic cor- Selected indications for and applications of magnetic resonance angiog-
relations with memory and learning – initial investigation. Radiology raphy in children. Pediatr Neurosurg 1994;20:113–125.
2006;240:194–202. 1041. Agthong S, Wiwanitkit V. Encephalomeningocele cases over 10 years in
1016. Cama A, Tortori-Donati P, Piatelli GL, Fondelli MP, Andreussi L. Chiari Thailand: a case series. BMC Neurol 2002;2:3.
complex in children – neuroradiological diagnosis, neurosurgical treat- 1042. Suwanwela C, Sukabote C, Suwanwela N. Frontoethmoidal encepha-
ment and proposal of a new classi-fication (312 cases). Eur J Pediatr Surg lomeningocele. Surgery 1971;69:617–625.
1995;5(Suppl 1):35–38. 1043. Suwanwela C, Suwanwela N. A morphological classification of sincipital
1017. Isik N, Elmaci I, Silav G, Celik M, Kalelioğlu M. Chiari malformation encephalomeningoceles. J Neurosurg 1972;36:201–211.
type III and results of surgery: a clinical study: report of eight surgically 1044. Mahapatra A, Suri A. Anterior encephaloceles: a study of 92 cases. Pediatr
treated cases and review of the literature. Pediatr Neurosurg 2009;45: Neurosurg 2002;36:113–118.
19–28. 1045. Belden CJ, Mancuso AA, Kotzur IM. The developing anterior skull
1018. Naidich TP, Altman NR, Braffman BH, McLone DG, Zimmerman RA. base: CT appearance from birth to 2 years of age. Am J Neuroradiol
Cephaloceles and related malformations. Am J Neuroradiol 1992;13: 1997;18:811–818.
655–690. 1046. Aryan H, Jandial R, Farin A, Chen J, Granville R, Levy M. Intradural cra-
1019. David DJ. Cephaloceles: classification, pathology, and management – a nial congenital dermal sinuses: diagnosis and management. Childs Nerv
review. J Craniofac Surg 1993;4:192–202. Syst 2006;22:243–247.
1020. Nahas Z, Tatlipinar A, Limb CJ, Francis HW. Spontaneous meningoen- 1047. Choudhury AR, Bandey SA, Haleem A, Sharif H. Glial heterotopias of the
cephalocele of the temporal bone: clinical spectrum and presentation. nose. Childs Nerv Syst 1996;12:43–47.
Arch Otolaryngol Head Neck Surg 2008;134:509–518. 1048. Naidich TP, McLone DG, Bauer BS, Kernahan DA, Zaparackas ZG. Mid-
1021. Van Allen M, Kalousek D, Chernoff G, et al. Evidence for multi-site line craniofacial dysraphism: midline cleft upper lip, basal encephalocele,
closure of the neural tube in humans. Am J Med Genet 1993;47:723–743. callosal agenesis, and optic nerve dysplasia. Concepts Pediat Neurosurg
1022. Nakatsu T, Uwabe C, Shiota K. Neural tube closure in humans initiates 1983;4:186–207.
at multiple sites: evidence from human embryos and implications for 1049. Naidich TP, Osborn RE, Bauer B, Naidich MJ. Median cleft face syn-
the pathogenesis of neural tube defects. Anat Embryol (Berl) 2000;201: drome: MR and CT data from 11 children. J Comput Asst Tomogr 1988;12:
455–466. 57–64.

Barkovich_Chap05.indd 565 5/6/2011 9:01:37 PM


566 Pediatric Neuroimaging

1050. Curnes JT, Oakes WJ. Parietal cephaloceles: radiographic and magnetic determined by and ALX4 homeobox gene mutation. Am J Neuroradiol
resonance imaging evaluation. Pediatr Neurosci 1988;14:71–76. 2004;25:1836–1839.
1051. Drapkin A. Rudimentary cephalocele or neural crest remnant? Neurosur- 1077. Bartsch O, Wuyts W, Van Hul W, et al. Delineation of a contiguous gene
gery 1990;26:667–673. syndrome with multiple exostoses, enlarged parietal foramina, craniofa-
1052. Yamazaki T, Enomoto T, Iguchi M, Nose T. Atretic cephalocele – report cial dysostosis, and mental retardation, caused by deletions on the short
of two cases with special reference to embryology. Childs Nerv Syst arm of chromosome 11. Am J Hum Genet 1966;58:734–741.
2001;17:674–678. 1078. Mendoza-Londono R, Lammer EJ, Watson R, et al. Characterization of a
1053. Morioka T, Hashiguchi K, Samura K, et al. Detailed anatomy of intrac- new syndrome that associates craniosynostosis, delayed fontanel closure,
ranial venous anomalies associated with atretic parietal cephaloceles parietal foramina, imperforate anus, and skin eruption: CDAGS. Am J
revealed by high-resolution 3D-CISS and high-field T2-weighted reversed Hum Genet 2005;77:161–168.
MR images. Childs Nerv Syst 2009;25:309–315. 1079. Fink AM, Maixner W. Enlarged parietal foramina: MR imaging features
1054. Brunelle F, Baraton J, Renier D, et al. Intracranial venous anomalies asso- in the fetus and neonate. Am J Neuroradiol 2006;27:1379–1381.
ciated with atretic cephalocoeles. Pediatr Radiol 2000;30:743–747. 1080. Frieden I. Aplasia cutis congenita: a clinical review and proposal for clas-
1055. Patterson R, Egelhoff J, Crone K, Ball WJ. Atretic parietal cephaloceles sification. J Am Acad Dermatol 1986;14:646–660.
revisited: an enlarging clinical and imaging spectrum? Am J Neuroradiol 1081. McMurray B, Martin L, St John Dignan P, Fogelson M. Hereditary aplasia
1998;19:791–795. cutis congenita and associated defects. Three instances in one family and
1056. Abe T, Lüdecke D, Wada A, Matsumoto K. Transsphenoidal cephaloceles a survey of reported cases. Clin Pediatr (Phila) 1977;16:610–614.
in adults. A report of two cases and review of the literature. Acta Neuro- 1082. Piatt J, Lagrange A. Congenital defects of the scalp and skull. In: Albright
chir (Wien) 2000;142:397–400. l, Pollack I, Adelson D, eds. Principles and Practice of Pediatric Neurosur-
1057. Morioka M, Marubayashi T, Masumitsu T, Miura M, Ushio Y. Basal gery. New York: Thieme Medical, 1999:209–218.
encephaloceles with morning glory syndrome, and progressive hormonal 1083. Aisenson HR. Closing of the anterior fontanelle. Pediatrics 1950;6:
and visual disturbances: case report and review of the literature. Brain 223–225.
Dev 1995;17:196–201. 1084. Duc G, Largo RH. Anterior fontanel: size and closure in term and pre-
1058. Martinez-Lage JF, Capel A, Costa TR, Perez-Espejo MA, Poza M. The term infants. Pediatrics 1986;78:904–908.
child with a mass on its head: diagnostic and surgical strategies. Childs 1085. Uzura M, Furuya Y, Akashi K, Sekino H. The persistence of an open ante-
Nerv Syst 1992;8:247–252. rior fontanel in a 4-year-old girl. Childs Nerv Syst 2005;21:83–85.
1059. Pannell BW, Hendrick EB, Hoffman HJ, Humphreys RP. Dermoid cysts of 1086. Kakita A, Inenaga C, Kameyama S, et al. Cerebral lipoma and the under-
the anterior fontanelle. Neurosurgery 1982;10:317–323. lying cortex of the temporal lobe: pathological features associated with
1060. Riebel T, David S, Thomale UW. Calvarial dermoids and epidermoids in the malformation. Acta Neuropathol (Berl) 2005;109:339–345.
infants and children: sonographic spectrum and follow-up. Childs Nerv 1087. Yildiz H, Hakyemez B, Koroglu M, Yesildag A, Baykal B. Intracranial lipo-
Syst 2008;24:1327–1332. mas: importance of localization. Neuroradiology 2006;48:1–7.
1061. Kaplan SB, Kemp SS, Oh KS. Radiographic manifestations of congenital 1088. Osaka K, Handa R, Matsumoto S, Yasuda M. Development of the cere-
anomalies of the skull. Radiol Clin North Am 1991;29:195–218. brospinal fluid pathway in the normal and abnormal human embryos.
1062. Yoon SH, Park S-H. A study of 77 cases of surgically excised scalp and Childs Brain 1980;6:26–38.
skull masses in pediatric patients. Childs Nerv Syst 2008;24:459–465. 1089. O’Rahilly R, Müller F. The meninges in human development. J Neuro-
1063. Nishio S, Morioka T, Murakami N, Fukui M, Inamitsu T, Ishihara S. Mel- pathol Exp Neurol 1986;45:588–608.
anotic neuroectodermal tumor of infancy at the anterior fontanel. Neu- 1090. Truwit CL, Barkovich AJ. Pathogenesis of intracranial lipoma: An MR
roradiology 1991;41:202–204. study in 42 patients. Am J Neuroradiol 1990;11:665–674.
1064. Inwards C, Unni K, Beabout J, Shives T. Solitary congenital fibromatosis 1091. Coban YK, Boran C, Omeroglu SA, Okur E. Pai syndrome: an adult
(infantile myofibromatosis) of bone. Am J Surg Pathol 1991;15:935–941. patient with bifid nose and frontal hairline marker. Cleft Palate Craniofac
1065. Moore J, Waldenmaier N, Potchen E. Congenital generalized fibromato- J 2002;40:325–328.
sis: a new management strategy provided by magnetic resonance imag- 1092. Pai GS, Levkoff AH, Leithiser RE, Jr. Median cleft of the upper lip associ-
ing. Am J Dis Child 1987;141:714–716. ated with lipomas of the central nervous system and cutaneous polyps.
1066. Queralt J, Poirier V. Solitary infantile myobibromatosis of the skull. Am J Am J Med Genet 1987;26:921–924.
Neuroradiol 1995;16:476–478. 1093. Dean B, Drayer BP, Beresini DC, Bird CR. MR imaging of pericallosal
1067. Chung E, Enzinger F. Infantile myofibromatosis. Cancer 1981;48: lipoma. Am J Neuroradiol 1988;9:929–931.
1807–1818. 1094. Hadecke J, Buchfelder M, Hans-Jürgen T, Schneyer U. Multiple intracra-
1068. Schut L, Sutton LN, Bruce DA. Vascular malformations of the scalp and nial lipomas: a case report. Neurosurg Rev 1997;20:282–287.
skull. In: Edwards MSB, Hoffman HJ, eds. Cerebral Vascular Disease 1095. Rengachary SS, Watanabe IJ. Ultrastructure and pathogenesis of intracra-
in Children and Adolescents. Baltimore: Williams and Wilkins, 1988: nial arachnoid cysts. Neuropathol Exp Neurol 1981;40:60–83.
411–422. 1096. Harsch GR, Edwards MSB, Willson CB. Intracranial arachnoid cysts in
1069. Spektor S, Weinberger G, Constantini S, Gomori JM, Beni-Adani L. Giant children. J Neurosurg 1986;64:835–842.
lateral sinus pericranii. J Neurosurg 1998;88:145–147. 1097. Naidich TP, McLone DG, Radkowski MA. Intracranial arachnoid cysts.
1070. Bollar A, Allut A, Prieto A, Gelabert M, Becerra E. Sinus pericranii: radio- Pediatr Neurosci 1986;12:112–122.
logical and etiopathological consideations. J Neurosurg 1992;77:469–472. 1098. Catala M, Poirier J. Arachnoid cysts: Histologic, embryologic and physio-
1071. Gandolfo C, Krings T, Alvarez H, et al. Sinus pericranii: diagnostic and pathologic review. Rev Neurol 1998;154:489–501.
therapeutic considerations in 15 patients. Neuroradiology 2007;49(6): 1099. Go KG, Houthoff HJ, Blaauw EH, et al. Arachnoid cysts of the sylvian
505–514. fissure. Evidence of fluid secretion. J Neurosurg 1984;60:803–810.
1072. Zabek M. Familial incidence of foramina parietalia permagna. Neuro- 1100. Becker T, Wagner M, Hoffman E, Warmuth-Metz M, Nadjmi M. Do
chirugia (Stuttg) 1987;30:25–27. arachnoid cysts grow? A retrospective CT volumetric study. Neuroradiol-
1073. Chakrabortty S, Oi S, Suzuki H, et al. Congenital frontal bone defect with ogy 1991;33:341–345.
intact overlying scalp. Childs Nerv Syst 1993;9:485–487. 1101. Gosalakkal JA. Intracranial arachnoid cysts in children: a review of
1074. Perlyn CA, Schmelzer R, Govier D, Marsh JL. Congenital scalp and calva- pathogenesis, clinical features, and management. Pediatr Neurol 2002;26:
rial deficiencies: principles for classification and surgical management. 93–98.
Plast Reconstr Surg 2005;115:1129–1141. 1102. Oberbauer R, Haase J, Pucher R. Arachnoid cysts in children: a European
1075. Reddy AT, Hedlund GL, Percy AK. Enlarged parietal foramina. Association co-operative study. Childs Nerv Syst 1992;8(5):281–286.
with cerebral venous and cortical anomalies. Neurology 2000;54:1175–1178. 1103. Schievink WI, Huston III J, Torres VE, Marsh WR. Intracranial cysts in
1076. Valente M, Valente KD, Sugayama SS, Kim CA. Malformation of corti- autosomal dominant polycystic kidney disease. J Neurosurg 1995;83:1004–
cal and vascular development in one family with parietal foramina 1007.

Barkovich_Chap05.indd 566 5/6/2011 9:01:37 PM


Chapter 5 • Congenital Malformations of the Brain and Skull 567

1104. Pierre-Kahn A, Sonigo P. Malformative intracranial cysts: diagnosis and 1132. Galland B, Taylor B, Bolton D. Prone versus supine sleep position: a
outcome. Childs Nerv Syst 2003;19(7–8):477–483. review of the physiological studies in SIDS research. J Paediatr Child
1105. Adan L, Bussières L, Dinand V, Zerah M, Pierre-Kahn A, Brauner R. Health 2002;38:332–338.
Growth, puberty and hypothalamic-pituitary function in children with 1133. Chadduck WM, Kast J, Donahue DJ. The enigma of lambdoid positional
suprasellar arachnoid cyst. Eur J Pediatr 2000;159:348–355. molding. Pediatr Neurosurg 1997;26:304–311.
1106. DiRocco C, Caldarelli M, DiTrapani G. Infratentorial arachnoid cysts in 1134. Pople IK, Sanford RA, Muhlbauer MS. Clinical presentation and management
children. Child’s Brain 1981;8:119–133. of 100 infants with occipital plagiocephaly. Pediatr Neurosurg 1996;25:1–6.
1107. Boltshauser E, Martin F, Altermatt S. Outcome in children with space-occu- 1135. Robinson S, Proctor M. Diagnosis and management of deformational
pying posterior fossa arachnoid cysts. Neuropediatrics 2002;33(3):118–121. plagiocephaly. J Neurosurg Pediatr 2009;3:284–295.
1108. Galassi E, Tognetti F, Gaist G, et al. CT scan and metrizamide cisternog- 1136. McGarry A, Dixon M, Greig R, Hamilton D, Sexton S, Smart H. Head
raphy in arachnoid cysts of the middle cranial fossa: calssification and shape measurement standards and cranial orthoses in the treatment
pathophysiological aspects. Surg Neurol 1982;17:363–369. of infants with deformational plagiocephaly. Dev Med Child Neurol
1109. Gelabert-González M, Fernandez-Villa J, Curtin-Prieto J, Garcia 2008;50:568–576.
Allut A, Martinez-Rumbo R. Arachnoid cyst rupture with subdural 1137. Huang MH, Gruss JS, Calrren SK, et al. The differential diagnosis of pos-
hygroma: report of three cases and literature review. Childs Nerv Syst terior plagiocephaly: true lambdoid sysnostosi versus positional molding.
2002;18(11):609–613. Plast Reconst Surg 1996;98:765–774.
1110. Cullis PA, Gilroy J. Arachnoid cyst with rupture into the subdural space. 1138. Lo LJ, Marsh JL, Pilgram TK, Vannier MW. Plagiocephaly: differen-
J Neurol Neurosurg Psychiatr 1983;46:454–456. tial diagnosis based on endocranial morphology. Plast Reconstr Surg
1111. Rakier A, Feinsod M. Gradual resolution of an arachnoid cyst after spon- 1996;97:282–291.
taneous rupture into the subdural space. J Neurosurg 1995;83:1085–1086. 1139. Anderson PJ, Harkness WJ, Taylor W, Jones BM, Hayward RD. Anoma-
1112. Little JR, Gomez MR, MacCarty CS. Infratentorial arachnoid cysts. J Neu- lous venous drainage in a case of nonsyndromic craniosynostosis. Childs
rosurg 1973;39:380–386. Nerv Syst 1997;13:97–100.
1113. Awaji M, Okamoto K, Nishiyama K. Magnetic resonance cisternog- 1140. Tokumaru A, Barkovich AJ, Ciricillo SF, Edwards MSB. Skull base and
raphy for preoperative evaluation of arachnoid cysts. Neuroradiology calvarial deformities: association with intracranial changes in craniofa-
2007;49:721–726. cial syndromes. Am J Neuroradiol 1996;17:619–630.
1114. Cohen Jr MM. Craniosynostosis update 1987. Am J Med Genet 1141. Jeevan DS, Anlsow P, Jayamohan J. Abnormal venous drainage in syn-
1988;4(Suppl):99–148. dromic craniosynostosis and the role of CT venography. Childs Nerv Syst
1115. Beighton P. Inherited Disorders of the Skeleton. Edinburgh: Churchill- 2008;24(12):1413–1420.
Livingstone, 1988. 1142. Lomri A, Lemonnier J, Hott M, et al. Increased calvaria cell differentiation
1116. Bixler D, Ward RE. Craniosynostosis. In: Myrianthopoulos NC, ed. Mal- and bone matrix formation induced by fibroblast growth factor receptor
formations, Vol 50. New York: Elsevier, 1987:113–128. 2 mutations in Apert syndrome. J Clin Invest 1998;101:1310–1317.
1117. Morriss-Kay GM, Wilkie AOM. Growth of the normal skull vault and its 1143. Bellus GA, Gaudenz K, Zackai EH, et al. Identical mutations in three dif-
alteration in craniosynostosis: insights from human genetics and experi- ferent fibroblast growth factor receptor genes in autosomal dominant
mental studies. J Anat 2005;207:637–653. craniosynostosis syndromes. Nat Genet 1996;14:174–176.
1118. Chumas PD, Cinalli G, Arnaud E, Marchac D, Renier D. Classification of pre- 1144. Robin NH, Muenke M, Schell U, et al. Linkage of Pfeiffer syndrome to
viously unclassified cases of craniosynostosis. J Neurosurg 1997;86:177–181. chromosome 8 centromere and evidence for genetic heterogeneity. Hum
1119. Burke MJ, Winston KR, Williams S. Normal sutural fusion and the eti- Mol Genet 1994;3:2153–2158.
ology of single sutural craniosynostosis: the microspicule hypothesis. 1145. Rutland P, Pulleyn L, Reardon W, et al. Identical mutations in the FGFR2
Pediatr Neurosurg 1995;22:241–247. gene cause both Pfeiffer and Crouzon syndrome phenotypes. Nat Genet
1120. Maxson R, Ishii M. The Bmp pathway in skull vault development. Front 1995;9:173–176.
Oral Biol 2008;12:197–208. 1146. Wilkie A, Slaney S, Oldridge M, et al. Apert syndrome results from local-
1121. Hunter AGW, Rudd NL. Craniosynostosis: I. Sagittal synostosis: its genet- ized mutations of FGFR2 and is allelic with Crouzon syndrome. Nat
ics and associated clinical findings in 214 ptients who lacked involvement Genet 1995;9:165–172.
of the coronal suture(s). Teratology 1976;14:185–193. 1147. Passos-Bueno M, Sertie A, Zatz M, Richieri-Costa A. Pfeiffer mutation is
1122. Agrawal D, Steinbok P, Cochrane D. Diagnosis of isolated sagittal synosto- an Apert patient: how wide is the spectrum of variability due to muta-
sis: are radiographic studies necessary? Childs Nerv Syst 2006;22:375–378. tions in the FGFR2 gene? Am J Med Genet 1997;71:243–245.
1123. Shipster C, Hearst D, Somerville A, Stackhouse J, Hayward R, Wade A. 1148. Kreiborg S, Barr M, Jr, Cohen MM, Jr. Cervical spine in the Apert syn-
Speech, language, and cognitive development in children with isolated drome. Am J Med Genet 1992;43:704–708.
sagittal synostosis. Dev Med Child Neurol 2003;45:34–43. 1149. Lajeunie E, Cameron R, El Ghouzzi V, et al. Clinical variability in patients
1124. Kimonis V, Gold JA, Hoffman TL, Panchal J, Boyadjiev SA. Genetics of with Apert’s syndrome. J Neurosurg 1999;90:443–447.
craniosynostosis. Semin Pediatr Neurol 2007;14:150–161. 1150. Fjørtoft M, Sevely A, Boetto S, Kessler S, Sarramon M, Rolland M. Pre-
1125. van Vlimmeren LA, van der Graaf Y, Boere-Boonekamp MM, L’Hoir MP, natal diagnosis of craniosynostosis: value of MR imaging. Neuroradiology
Helders PJM, Engelbert RHH. Risk factors for deformational plagioceph- 2007;49(6):515–521.
aly at birth and at 7 weeks of age: a prospective cohort study. Pediatrics 1151. Cohen MM, Jr, Kreiborg S. An updated pediatric perspective on the Apert
2007;119(2):e408–e418. syndrome. AJDC 1993;147:989–993.
1126. Boyadjiev SA. Genetic analysis of non-syndromic craniosynostosis. 1152. Taravath S, Tonsgard JH. Cerebral malformations in Carpenter syn-
Orthod Craniofacial Res 2007;10:129–137. drome. Pediatr Neurol 1993;9:230–234.
1127. Arnaud E, Renier D, Marchac D. Prognosis for mental function in 1153. Siegenthaler JA, Ashique AM, Zarbalis K, et al. Retinoic acid from the
scaphocephaly. J Neurosurg 1995;83:476–479. meninges regulates cortical neuron generation. Cell 2009;139:597–609.
1128. Cohen MM, Jr, McLean RE. Craniosynostosis: Diagnosis, Evaluation, and 1154. Cohen MM, Jr. Pfeiffer syndrome update, clinical subtypes, and guide-
Management. New York: Oxford University Press, 2000. lines for differential diagnosis. Am J Med Genet 1993;45:300–307.
1129. Lajeunie E, Le Merrer M, Bonaiti-Pellie C, Marchac D, Renier D. Genetic 1155. Murovic JA, Posnick JC, Drake JM, Humphreys RP, Hoffman HJ,
study of nonsyndromic coronal craniosynostosis. Am J Med Genet Hendrick EB. Hydrocephalus in Apert syndrome: a retrospective review.
1995;55:500–504. Pediatr Neurosurg 1993;19:151–155.
1130. Sawin PD, Muhonen MG, Menezes AH. Quantitative analysis of cerebro- 1156. Collmann H, Sörensen N, Krau&szlig; J. Hydrocephalus in craniosynos-
spinal fluid spaces in children with occipital plagiocephaly. J Neurosurg tosis: a review. Childs Nerv Syst 2005;21:902–912.
1996;85:428–434. 1157. Rich P, Cox T, Hayward R. The jugular foramen in complex and syndro-
1131. Pollack IF, Losken HW, Fasick P. Diagnosis and management of posterior mic craniosynostosis and its relationship to raised intracranial pressure.
plagiocephaly. Pediatrics 1997;99:180–185. Am J Neuroradiol 2003;24:45–51.

Barkovich_Chap05.indd 567 5/6/2011 9:01:37 PM


568 Pediatric Neuroimaging

1158. Cinalli G, Sainte-Rose C, Kollar EM, et al. Hydrocephalus and cranio- 1184. Gottfried M, Lavine L, Roessmann U. Neuropathological findings in
synostosis. J Neurosurg 1998;88:209–214. Wolf-Hirschhorn (4p-) syndrome. Acta Neuropathol 1981;55:163–165.
1159. Taylor WJ, Hayward RD, Lasjaunias P, et al. Enigma of raised intracranial 1185. Terplan K, Lopez E, Robinson H. Histologic structural anomalies in the
pressure in patients with complex craniosynostosis: the role of abnormal brain in trisomy 18 syndrome. Am J Dis Child 1970;119:223–252.
intracranial venous drainage. J Neurosurg 2001;94:377–385. 1186. Lang A. Trisomy 18 and cyclopia. Teratology 1976;14:195–204.
1160. Robson CD, Mulliken JB, Robertson RL, et al. Prominent basal emissary 1187. Chen H. Mosaic trisomy 19 syndrome. Ann Genet 1981;24:32–33.
foramina in syndromic craniosynostosis: correlationwith phenotypic 1188. Rethore M. Sur un cas de trisomie 19 en mosaique. Ann Genet 1981;24:
and molecular diagnoses. Am J Neuroradiol 2000;21:1707–1717. 32–33.
1161. Sandberg D, Navarro R, Blanch J, Ragheb J. Anomalous venous drainage 1189. Pi S, Fineman R, Wing S, Grunnet M, Chan G. Holoprosencephaly in a
preventing safe posterior fossa decompression in patients with Chiari mal- Down syndrome child. Am J Med Genet 1980;5:201–206.
formation type I and multisutural craniosynostosis. Report of two cases 1190. Benda C. Down Syndrome. New York: Grune & Stratton, 1969.
and review of the literature. J Neurosurg 2007;106(6 Suppl):490–494. 1191. Blackburn W. Comparative studies of infants with mosaic and complete
1162. Thompson D, Harkness W, Jones B, Hayward R. Aetiology of herniation of triploidy: an analysis of 55 cases. Birth Defects 1982;18:251–274.
the hindbrain in craniosynostosis. Pediatr Neurosurg 1997;26:288–295. 1192. Emberger J, Marty-Double C, Pincemin D, Caderas de Kerleau J. Holo-
1163. Cohen MM, Jr. Craniosynostosis: Diagnosis, Evaluation, and Management. prosencephaly by triploidy 69 XXX in a 5 month old fetus. Ann Genet
New York: Raven, 1986. 1976;19:191–193.
1164. Schinzel A. Catalogue of Unbalanced Chromosome Alterations in Man. 1193. Hasegawa T, Harada N, Ikeda K, et al. Digynic triploid infant surviving
Berlin: Walter de Gruyter, 1984. for 46 days. Am J Med Genet 1999;87:306–310.
1165. Kumar AJ, Naidich TP, Stetten G, et al. Chromosomal disorders: back- 1194. Barkovich AJ, Peck WW. MR of Zellweger syndrome. Am J Neuroradiol
ground and neuroradiology. Am J Neuroradiol 1992;13:577–593. 1997;18:1063–1070.
1166. Tamraz JC, Retmore MO, Iba-zizen MT, Lejeurn J, Cabanis EA. Contri- 1195. Volpe JJ, Adams RD. Cerebro-hepato-renal syndrome of Zellweger. An
butions of MRI to the knowledge of CNS malformations related to chro- inherited disorder of neuronal migration. Acta Neuropathol 1972;20:
mosomal aberrations. Hum Genet 1987;76:265–273. 175–198.
1167. Martich V, Ben-Ami T, Yousefzadeh DK, Roizen NJ. Hypoplastic poste- 1196. Gressens P, Baes M, Leroux P, et al. Neuronal migration disorder in Zell-
rior arch of C-1 in children with Down syndrome: a double jeopardy. weger mice is secondary to glutamate receptor dysfunction. Ann Neurol
Radiology 1992;183:125–128. 2000;48:336–343.
1168. Peuschel SM, Scola FH. Atlantoaxial instability in individuals with Down 1197. Brown G. Pyruvate dehydrogenase E1 α deficiency. J Inherited Metab Dis
syndrome: epidemiologic, radiographic and clinical studies. Pediatrics 1992;15:625–633.
1987;80:555–560. 1198. Dahl H, Hansen L, Brown R, Danks D, Rogers J, Brown G. X-linked
1169. El-Khoury GY, Clark CR, Dietz FR, Harre RG, Tozzi JE, Kathol MH. pyruvate dehydrogenase E1 alpha subunit deficiency in heterozygous
Posterior atlantooccipital subluxation in Down syndrome. Radiology females: variable manifestation of the same mutation. J Inherited Metab
1986;159:507–509. Dis 1992;15:835–847.
1170. Martel W, Tishler JM. Observations on the spine in mongoloidism. AJR 1199. Garcia C, Mcgarry P, Voirol M, Duncan C. Neurological involvement in
1966;97:630–638. the Smith-Lemli-Opitz syndrome: clinical and neuropathological fidings.
1171. Menezes AH, Ryken TC. Craniovertebral abnormalities in Down’s syn- Dev Med Child Neurol 1973;15:48–55.
drome. Pediatr Neurosurg 1992;18:24–33. 1200. Kelley R, Hennekam R. The Smith-Lemli-Opitz syndrome. J Med Genet
1172. Coria F, Quintana F, Villalba A, et al. Craniocervical abnormalities in 2000;37:321–335.
Down syndrome. Dev Med Child Neurol 1983;25:252–254. 1201. Ryan A, Bartlett K, Clayton P, et al. Smith-Lemli-Opitz syndrome: a vari-
1173. Inagaki M, Ando Y, Mito T, et al. Comparison of brain imaging and neuro- able clinical and biochemical phenotype. J Med Genet 1998;35:558–565.
pathology in cases of trisomy 18 and 13. Neuroradiology 1987;29:474–479. 1202. Trasimeni G, Di Biasi C, Iannilli M, et al. MRI in Smith-Lemli-Opitz syn-
1174. Iannaccone ST, Rosenberg RN. Principles of molecular genetics and neu- drome type I. Childs Nerv Syst 1997;13:47–49.
rologic diseases. In: Berg BO, ed. Principles of Child Neurology. New York: 1203. Paetou A, Salonen R, Hattia M. Brain pathology in the Meckel syndrome:
McGraw-Hill, 1996:461–606. a study of 59 cases. Clin Neuropathol 1985;4:56–62.
1175. Sutherland GR, Gedeon A, Kornman L, et al. Prenatal diagnosis of fragile 1204. Delous M, Baala L, Salomon R, et al. The ciliary gene RPGRIP1L is
X syndrome by direct detection of the unstable DNA sequence. N Eng J mutated in cerebello-oculo-renal syndrome (Joubert syndrome type B)
Med 1991;325:1720–1722. and Meckel syndrome. Nat Genet 2007;39:875–881.
1176. Shapiro LR, Wilmot PL, Shapiro DA, Pettersen IM, Casamassima AC. 1205. Frank V, Hollander AId, Brüchle NO, et al. Mutations of the CEP290 gene
Cytogenetic diagnosis of the fragile X syndrome: efficiency, utilization, encoding a centrosomal protein cause Meckel-Gruber syndrome. Human
and trends. Am J Med Genet 1991;38:408–410. Mutation 2008;29:45–52.
1177. Hagerman RJ. Physical and behavioural phenotypes. In: Hagerman 1206. Shigematsu H, Takashima S, Otani K, Ieshima A. Neuropathologi-
RJ, Silverman AC, eds. Fragile X Syndrome: Diagnosis, Treatment, and cal and Golgi study on a case of thanatomorphic dysplasia. Brain Dev
Research. Baltimore: Johns Hopkins University Press, 1991:32–34. 1985;7:628–632.
1178. Barnea-Goraly N, Eliez S, Hedeus M, et al. White matter tract alterations 1207. Coulter C, Leech R, Brumback R, Schaefer G. Cerebral abnormalities in
in fragile X syndrome: preliminary evidence from diffusion tensor imag- thanatophoric dysplasia. Radiat Res 1991;7:21–26.
ing. Am J Med Genet 2003;118B:81–88. 1208. Savarirayan R, Thompson E, Abbott K, Moore M. Cerebral cortical dys-
1179. Giglio S, Broman K, Matsumoto N, et al. Olfactory receptor-gene clusters, plasia and digital constriction rings in Adams-Oliver syndrome. Am J
genomic-inversion polymorphisms, and common chromosome rear- Med Genet 1999;86:15–19.
rangements. Am J Hum Genet 2001;68(4):874–883. 1209. Amor D, Leventer R, Hayllar S, Bankier A. Polymicrogyria associated
1180. Feenstra I, van Ravenswaalj CMA, van der Knaap MS, Willemsen MAAP. with scalp and limb defects: variant of Adams-Oliver syndrome. Am J
Neuroimaging in nine patients with inversion duplication of the short Med Genet 2000;93:328–334.
arm of Chromosome 8. Neuropediatrics 2006;37:83–87. 1210. Galloway W, Mowat A. Congenital microcephaly with hiatus hernia and
1181. Battaglia A, Carey JC, Cederholm P, Viskochil DH, Brothman AR, Galasso nephrotic syndrome in two sibs. J Med Genet 1968;5:319–321.
C. Natural history of Wolf-Hirschhorn syndrome: experience with 1211. Cooperstone B, Friedman A, Kaplan B. Galloway-Mowat syndrome
15 cases. Pediatrics 1999;103:830–836. of abnormal gyral patterns and glomerulopathy. Am J Med Genet
1182. Shannon NL, Maltby EL, Rigby AS, Quarrell OWJ. An epidemiological 1993;47:250–254.
study of Wolf-Hirschhorn syndrome: life expectancy and cause of mor- 1212. Kozlowski P, Sher J, Nicastri A, Rudelli R. Brain morphology in the
tality. J Med Genet 2001;38:674–679. Galloway syndrome. Clin Neuropathol 1989;8:85–91.
1183. Citoler P, Gropp A, Gullotta F. Cytogenetics and pathological observa- 1213. Robain O, Deonna T. Pachygyria and congenital nephrosis: disorder of
tions in the 4p-syndrome. Beitr Pathol 1971;143:84–96. migration and neuronal orientation. Acta Neuropathol 1983;60:137–141.

Barkovich_Chap05.indd 568 5/6/2011 9:01:37 PM


CHAPTER

The Phakomatoses
6 GILBERT VEZINA AND A. JAMES BARKOVICH

Neurofibromatosis type 1 Neurocutaneous melanosis


Clinical manifestations Clinical manifestations
Cranial and intracranial manifestations Pathological manifestations
Spinal manifestations Neuroimaging manifestations
Neurofibromatosis type 2 Incontinentia pigmenti
Clinical manifestations Clinical manifestations
Imaging of intracranial manifestations Pathological and neuroimaging manifestations
Imaging of spinal manifestations Hypomelanosis of Ito
Other forms of neurofibromatosis Clinical manifestations
Tuberous sclerosis complex Neurological manifestations
Clinical manifestations Neuropathological and neuroimaging manifestations
Cutaneous manifestations Basal cell nevus syndrome
Ocular manifestations Clinical manifestations
Intracranial manifestations Neuroimaging manifestations
Non-CNS manifestations PHACE (posterior fossa malformations, facial
Sturge-Weber disease hemangiomas, arterial anomalies, cardiac anomalies
Clinical manifestations and aortic coarctation, eye anomalies) syndrome
Intracranial pathology Clinical manifestations
Neuroimaging manifestations Neuroimaging manifestations
Extra-CNS manifestations Diffuse neonatal hemangiomatosis
von Hippel-Lindau disease Chédiak-Higashi syndrome
Clinical manifestations Progressive facial hemiatrophy (Parry-Romberg syndrome)
Ocular manifestations Epidermal nevus syndrome
Brain and spinal cord manifestations Clinical manifestations
Papillary cystadenomas of the endolymphatic sac Ocular and brain manifestations
Visceral manifestations Encephalocraniocutaneous lipomatosis
Ataxia-telangiectasia Clinical manifestations
Clinical manifestations Neuroimaging manifestations
Pathological and neuroimaging manifestations Other overgrowth syndromes
Nijmegen breakage syndrome

he phakomatoses are disorders characterized by multiple phakomatoses. Patients with several phakomatoses (and other inherited
T hamartomas and other congenital malformations affecting
mainly structures of ectodermal origin, that is, the nervous
cancer predisposition syndromes) have increased genetic susceptibil-
ity to multiple primary malignancies, which is enhanced by sensitivity
system, the skin, the retina, and the globe and its contents; visceral to ionizing radiation (1). In these disorders, listed in Table 6-1, care
organs are also involved, but in general to a lesser extent. Classically, should be used to minimize exposure to radiation during diagnosis,
four diseases were included in this group: von Recklinghausen neuro- treatment, and follow-up; MRI should be the favored imaging modal-
fibromatosis, tuberous sclerosis (Bourneville disease), retinocerebellar ity. A discussion of all of the neurocutaneous disorders that have been
angiomatosis (von Hippel-Lindau disease), and encephalotrigeminal described is beyond the scope of this book. Instead, we describe those
angiomatosis (Sturge-Weber disease). However, many other heredi- that are most commonly seen in practice and likely to be seen in mod-
tary diseases have subsequently been classified under the heading of erate to large-sized outpatient and inpatient practices.

569

Barkovich_Chap06.indd 569 5/6/2011 9:03:45 PM


570 Pediatric Neuroimaging

Phenotypic expression of the disease is extremely variable, both clini-


TABLE 6-1 Inherited Cancer Predisposition cally and radiologically; this variation has resulted in attempts to cor-
relate phenotypes with underlying genetic factors (14). Overall, no
Syndromes in Which Exposure genotype-phenotype relationships have been identified by mutational
to Ionizing Radiation Should analyses of NF1 patients (15). Genetic modifiers, and possibly environ-
be Minimized mental modifiers (at the level of the cellular milieu), unlinked to the
NF1 locus, likely contribute to the variable expressivity of the disease
(16). Diagnostic criteria are listed in Table 6-2.
Nijmegen breakage syndrome
Ataxia-telangiectasia
Clinical Manifestations (Tables 6-2 and 6-3)
Nevoid basal cell nevus syndrome
Cafe au lait spots are the first manifestation of NF1 to develop; they
Li-Fraumeni syndrome
are sometimes present at birth and usually develop at the age of 2 years
Hereditary retinoblastoma (7). Freckling within the axilla will develop later in about two thirds
Neurofibromatosis type 1 of patients (17–19). Cutaneous neurofibromas begin to appear around
the onset of puberty and increase in number throughout life. Lisch
(Modified from Reference 471) nodules, which are best seen by slit lamp examination, begin to appear
in childhood and are present in almost all affected adults (17–19).
A number of other features are characteristic of this disease, the most
important from the neuroimaging standpoint being gliomas of the
optic pathway (and other intracranial astrocytomas, which may have
NEUROFIBROMATOSIS TYPE 1 a higher incidence in patients with optic pathway tumors [20,21]),
kyphoscoliosis, sphenoid wing dysplasia, vascular dysplasias, nerve
Neurofibromatosis type 1 (NF1) is one of the most common autosomal
sheath tumors, and macrocephaly. NF1 is associated with an increased
dominant central nervous system (CNS) disorders. It has a world-
incidence of tumors, with the frequency of brain tumors (other than
wide incidence of approximately 1 per 2,500 to 3,000 individuals,
optic glioma) being 1.5% to 2.0% and that of extra-CNS tumors (most
without preference for race or sex (2–5). NF1 is the disorder initially
commonly sarcomas, pheochromocytomas, and leukemia) being 3%
described by von Recklinghausen in 1882 (5). The genetic locus has
to 5% (22). Megalencephaly is common; the cerebral enlargement is
been mapped to chromosome 17q11.2 (4). The NF1 gene produces
primarily attributable to an increase in cerebral white matter volume,
a cytoplasmic protein called neurofibromin, which is enormous and
with a smaller contribution from the gray matter (23). About 4% to 7%
appears to have many functions. In the CNS, it is expressed primarily in
of patients have epilepsy, primarily related to intracranial masses and
neurons, Schwann cells, oligodendrocytes, and astrocytes (2,3). Under-
cytoarchitectural abnormalities (24).
standing of the molecular pathogenesis of NF1 and the functions of
Cognitive impairment is found in 30% to 65% of children with NF1
the NF1 gene helps explain the extensive clinical and imaging features
and consists of a wide range of learning disabilities (most frequently
of this syndrome. Understanding the functions of NF1 also allows for
problems with attention, perception, executive functioning, and aca-
the development of targeted therapies to improve the management of
demic achievement) (17,18,25–27). The most common neuropsy-
clinical problems associated with NF1 (6,7). The functions of neurofi-
chological manifestation of NF1 in childhood is a specific learning
bromin include the following:
disability, defined as a major discrepancy between ability (intellect or
1. It appears to act as a tumor suppressor gene that functions in part
as a negative regulator of the RAS protooncogene (8).
2. It acts as a regulator of neural stem-cell proliferation, survival, and
astroglial differentiation, and is a regulator of neuroglial progeni-
tor function (9,10). Neurofibromin is required for intracellular TABLE 6-2 Criteria for Diagnosis of
cyclic AMP (cAMP) generation in both neurons and astrocytes; Type 1 Neurofibromatosis
abnormal levels of cAMP appear to be, at least in part, responsible
for the abnormalities in glial and neuronal development evident in
The diagnosis of NF1 requires two or more of the following:
NF1 patients.
3. It regulates ERK signaling in GABA release, an important pathway (1) Six or more cafe au lait spots over 5 mm in greatest diameter
involved in learning (and therefore in learning disabilities) (11). (over 15 mm in postpubertal individuals)
4. It is involved in the maintenance of the vascular wall. Neurofibro- (2) Two or more neurofibromas of any type or one or more plexi-
min is expressed in the vascular endothelium; loss of neurofibro- form neurofibroma
min likely causes smooth muscle proliferation, possibly in response
(3) Freckling in the axillary or inguinal areas
to nonspecific injury to the vascular wall (10), which can lead to
vasculopathy. (4) Optic glioma
5. It is involved in bone formation and remodeling. Neurofibromin (5) Two or more Lisch nodules (pigmented hamartomas of the iris)
is expressed in osteoblasts; it inhibits collagen synthesis, promotes
mineralization, and regulates osteoclastogenesis (103). (6) A distinctive osseous lesion such as sphenoid dysplasia or
6. It seems to be required for normal myelination by Schwann cells thinning of long bone cortex
(12). In addition, the gene for oligodendrocyte myelin glycopro- (7) A first-degree relative (parent, sibling, or offspring) with NF1
tein, a major myelin protein, is embedded within intron 27b of the
NF1 gene (13). Therefore, it is not surprising that abnormalities of (Adapted from Reference 473)
myelin/white matter are seen in patients with NF1.

Barkovich_Chap06.indd 570 5/6/2011 9:03:45 PM


Chapter 6 • The Phakomatoses 571

populations of NF1 patients, those with and those without a degree of


TABLE 6-3 Incidence of Clinical cognitive impairment. Multiple studies have attempted (with variable
success) to establish a correlation between the presence of the char-
Features of NF1 acteristic T2 bright foci seen in NF1 children and the cognitive defi-
cits evident in psychometric testing (26,28–31). The methodologies of
Feature Incidence these studies are complicated by the peculiar temporal evolution of the
Cutaneous Lesions T2 bright foci: they are absent at birth; they appear in early childhood
and reach peak numbers around age 7 to 12 years; and they essentially
>6 Café au lait spots >95% disappear by the end of the second decade of life. One of the most con-
Axillary freckling 65%–85% sistent markers of cognitive deficits between age 8 and 18 years appears
to be the presence of T2 bright foci in the thalami (26,28–32).
Cutaneous neurofibromas
Although learning disabilities are common in NF1, intellectual dis-
Age 0–9 y 15% ability affects a much smaller proportion of patients. Some mentally
Age 10–19 y 45% disabled NF1 patients have been found to have deletions of the entire
NF1 gene, leading to a distinctive phenotype that includes the devel-
Age 20–29 y 85%
opment of a large number of neurofibromas and a distinctive facial
Over age of 30 y 95% appearance (33,34). Affected patients also appear to have a greater
Lisch nodules number of structural anomalies detected in their brains by MR studies.
It is unclear whether this phenotype is the result of haploinsufficiency
Age 0–4 y 20% for the NF1 gene or whether a deletion of portions of contiguous genes
Age 5–9 y 40% contributes to the clinical picture (33,34).
Age 10–19 y 80% It is important to remember that few features of NF1 are likely
to be present in any particular patient. For example, one patient may
Over age of 20 y 95% have a normal vascular system and a normal intellect but have severe
Neurologic Lesions kyphoscoliosis, while another may have a normal spine and normal
cognition but have severe hypertension from renal artery stenosis.
Plexiform neurofibromas 30%
Because this chapter is about neuroradiology, we concentrate on the
Head and neck only 2%–4% CNS manifestations of NF1, which have an incidence of about 15%
Malignant peripheral nerve tumors 2%–3% (18,19). Because of this high incidence, with many lesions occurring at
an early age when signs and symptoms may be difficult to elicit, some
Lifetime risk 10%
centers advocate MR screening of asymptomatic children with NF1
Cognitive deficits (at least once between age 2 and 6 years); the value of such screening
Mental retardation 5% exams has not been determined in a large prospective cohort.
Learning disabilities 50%
Scoliosis 10%–26% Cranial and Intracranial Manifestations
Optic pathway gliomas 15% Optic Pathway Gliomas
Symptomatic 5% The most important primary brain abnormality in NF1 is the optic
Short stature 20%–30% pathway glioma (OPG) (18,35,36). OPG is seen in 15% to 20% of indi-
viduals with NF1, and 30% to 50% of these tumors will cause clini-
Macrocephaly 50%
cal symptoms, including vision loss and precocious puberty (32,33);
Epilepsy 5% therefore, the incidence of symptomatic optic pathway tumors is about
Other Systems 5% to 7%. Almost all OPGs present before age of 7 years; the tumor
can be isolated to a single optic nerve or can involve both optic nerves,
Pseudarthrosis of long bones 3%
the chiasm and the optic tracts (Figs. 6-1 to 6-4). Primary involvement
Sphenoid wing dysplasia 1% of the optic nerve is more common in the NF1 population, while spo-
Renal artery stenosis 1% radic OPG (patients without NF1) more commonly involves the chi-
asm and optic radiations (37). The prognosis is poorer for tumors that
Osteoporosis up to 50% involve the optic chiasm or tracts as compared to those involving only
Adapted from 3,7,19,103. the optic nerves (36,38). Irrespective of the distribution, the clinical
course of NF1-associated OPG is more indolent than in the sporadic
OPG (37,39).
Optic pathway tumors are most commonly of low histological
grade. Although those involving only the optic nerves are sometimes
aptitude) and achievement (performance) in patients with IQ scores classified as hamartomas rather than neoplasms, most OPG are pilo-
in the normal range. Specific learning disability has been estimated to cytic astrocytomas; in addition, some optic pathway tumors sponta-
occur in 30% to 45% of children with NF1, which is more than three neously involute over time in patients with NF1 (40–42). Therefore,
times the prevalence in the general population (26,27). The occurrence aggressive therapy is usually not indicated unless symptoms progress
of cognitive deficits does not seem to be associated with the clinical or radiologic evidence of progression is noted. Uncommonly, optic
severity of the disease (26). In fact, a bimodal distribution of full scale pathway tumors can be highly malignant, demanding a very aggressive
IQ scores is seen in children with NF1, suggesting that there are two treatment approach (38).

Barkovich_Chap06.indd 571 5/6/2011 9:03:45 PM


572 Pediatric Neuroimaging

FIG. 6-1. Optic pathway glioma in NF1. A. Sagittal T1-weighted image


shows a large mass (white arrows) replacing the chiasm and extending
posteriorly to the mammillary body and superiorly (black arrow) into the
fornices and septum pellucidum. B. Postcontrast axial T1-weighted image
shows enhancing tumor (white arrows) in the optic tracts, with extension
toward the mesial temporal lobes. C. Axial T2-weighted image at a higher
level reveals tumor infiltration in the optic tracts to the level of the lateral
geniculate body/posterior thalamus (white arrows). Tumor infiltration of
the fornices is also evident (black arrows).

Tumors of the optic nerve diffusely expand the nerve, producing As discussed in Chapter 7, CT is adequate for evaluating tumors
a fusiform mass. Two architectural forms can be distinguished. The of the intraorbital optic nerves. The orbital fat provides intrinsic con-
first is a diffuse expansion of the optic nerve itself without significant trast against which to visualize these lesions. Enlargement of the optic
subarachnoid tumor (Fig. 6-2C). The second is tumor infiltration of nerve is usually fusiform, although occasionally the enlargement can
the subarachnoid space, creating a rim of tumor around a relatively be rather eccentric; therefore, axial images should be obtained every
unaffected nerve (Fig. 6-3) (43,44). Postcontrast MRI using fat sup- 1 to 2 mm, preferably with helical scanning, and then reformatted
pression will distinguish the two architectural forms of optic nerve for optimal depiction of the nerves. Enlargement of the optic canals
gliomas. When the nerve is diffusely infiltrated, enhancing tumor fills will usually be seen if bone windows are used. CT cannot distinguish
the optic nerve sheath, whereas infiltration of the subarachnoid spaces the two architectural forms of optic gliomas and is less sensitive in
shows a rim of enhancing tumor around a minimally enhancing optic assessing intracranial involvement, such as involvement of the optic
nerve. Optic nerve tortuosity is commonly observed in NF1, even in chiasm. Unless the chiasm is very enlarged by gross tumor involve-
the absence of optic nerve glioma. ment, it is necessary to inject intrathecal water-soluble contrast to see

Barkovich_Chap06.indd 572 5/6/2011 9:03:45 PM


Chapter 6 • The Phakomatoses 573

FIG. 6-2. Optic pathway glioma with extensive infiltration. A. Sagittal


T1-weighted image shows a mass extending from the chiasm and hypothalamus
(arrows) extending toward the mamillary bodies and the fornices. The pons is
slightly enlarged and shows abnormal low T1 signal. B. Axial T2-weighted image
shows that T2 hyperintense tumor has infiltrated into the medial basal gan-
glia/internal capsules (white arrows) and the fornices (black arrow). The tumor
has extended posteriorly to the level of the lateral geniculate bodies. C. Axial
T2-weighted image at a lower level reveals diffuse tumor infiltration, manifested
as hyperintensity of the pons and middle cerebellar peduncles (long white arrows).
Both optic nerves (short white arrows) are enlarged with homogeneous low T2
signal.

the chiasm well. In children, approximately 3 mL of nonionic contrast slice thickness, should be obtained through the globes, optic nerves,
(concentration 150 mg of iodine per mL) is injected via lumbar punc- and optic chiasm, and followed by fat-suppressed contrast-enhanced
ture. The patient is tilted 45° head down (Trendelenburg) in the prone T1 axial and coronal images. A routine brain scan should follow to
position for approximately 2 minutes and then transported to the CT look for involvement of the optic tracts and other areas of brain. Fluid
scanner and imaged. attenuated inversion recovery (FLAIR) and T2-weighted images show
MRI is the preferred imaging modality for evaluating the entire involvement of the optic tracts as high signal intensity extending pos-
CNS in NF1, and this includes the optic nerves; moreover, the use of teriorly, often to the level of the lateral geniculate bodies (Figs. 6-1
MRI eliminates exposure of the child to ionizing radiation. Assess- and 6-2). Extension beyond the optic pathway can be identified when
ment of the precise diameter of the intraorbital optic nerves may be present: superiorly into the hypothalamus, fornices, and septum pel-
hampered by chemical shift artifact resulting from the location of the lucidum; laterally into the temporal lobes; posteriorly into the optic
nerve adjacent to orbital fat. The use of a fat suppression pulse, how- radiations; and inferiorly into the cerebral peduncles and the brain-
ever, will eliminate the chemical shift artifact and allow the nerve to be stem (Fig. 6-2). Rarely tumor can extend into the lateral ventricles.
evaluated well. The intracranial optic nerves, optic chiasm, optic tract, Care should be taken not to mistake enlarged subarachnoid spaces
and optic radiations are all beautifully demonstrated with MRI. T1 and within the optic nerve sheaths (the result of ectasia of the dura sur-
fat-suppressed T2-weighted axial and coronal sequences, using ≤3 mm rounding the optic nerves) for optic nerve tumors. The enlarged

Barkovich_Chap06.indd 573 5/6/2011 9:03:46 PM


574 Pediatric Neuroimaging

FIG. 6-3. Optic nerve tumor


with infiltration of the optic
nerve sheath. A. Postcontrast axial
T1-weighted image with fat sup-
pression shows enlarged, enhanc-
ing the left optic nerve (long white
arrow) and tumor infiltration into
the dilated perineural spaces (low
T1 signal bordered by a thin rim of
enhancement (short white arrows).
The right optic nerve (black
arrows) shows a smaller tumor.
B. Oblique sagittal T2-weighted
image of the left optic nerve with
fat suppression reveals thickening
and elongation of the nerve. The
sheath around the nerve shows
marked dilatation and hyperin-
tense signal (white arrows).

FIG. 6-4. Multivoxel spectroscopy of optic pathway tumor


in NF1. A. Axial FLAIR image shows tumor infiltration in the
chiasm/hypothalamus (long white arrow) and into the optic
tracts (short white arrows). B. Color map of the choline-to-
creatine ratio (Ch:Cr) from a long echo (TE = 270) multivoxel
spectroscopy reveals the highest ratio of choline to creatine
(red color) at the level of the chiasm/hypothalamus. Signifi-
cant Ch:Cr elevation is also evident in the thalami bilaterally
(yellow color). C. MR spectra from four different ROIs (num-
bered in image B). Highest (Ch:Cr) is evident in box 1, at the
level of the chiasm/hypothalamus. Ch:Cr levels are elevated
in the thalamus (boxes 3, 4) compared to the putamen (box 2);
this suggests the presence of microscopic tumor infiltration
in the thalamus, despite the normal FLAIR signal.

Barkovich_Chap06.indd 574 5/6/2011 9:03:48 PM


Chapter 6 • The Phakomatoses 575

perioptic spaces have signal intensity identical to cerebrospinal fluid, astrocytomas (Fig. 6-5) are most common, but other low-grade (49)
do not enhance after administration of contrast, and are nonprogres- and higher-grade tumors (47,50) also occur. Almost any part of the
sive (45). Care should also be taken not to confuse extension of OPG CNS can be affected, but the suprasellar area (see above) is the most
into the optic tracts (and beyond) with the characteristic T2 bright foci common site; the brainstem (Figs. 6-6 and 6-7) is another common
encountered in pediatric patients with NF1 (see next section). Differ- site for tumors in NF1 (51). It is of interest to note that the brainstem
entiation of the two pathologies can be very difficult; features of OPG tumors occurring in NF1 seem to differ biologically from brainstem
infiltration include contiguity, mass effect, low T1 signal on precon- tumors in other patients; most do not require therapeutic intervention
trast images, enhancement after contrast administration, and signifi- (51). In contrast to the predominance of pontine tumors in the general
cant elevations of choline (at MR spectroscopy) (Fig. 6-4) (46). population, the medulla is the most common site of brainstem tumor
origin in NF1, followed by the mesencephalon (particularly the peri-
Other Gliomas and Tumor-Like Conditions aqueductal region/tectum) and the pons. This difference in location
Astrocytomas are more common in NF1 than in the general population may partly explain the indolent course and vastly better long-term out-
(18,35,47,48); they can arise anywhere in the CNS. Juvenile pilocytic come of most brainstem tumors in NF1 (51–54). However, an indolent

FIG. 6-5. Development of a juvenile pilocytic astrocytoma in an area of extensive myelin vacuolation. A. Axial T2-weighted image of a 2-year-old child
reveals multifocal hyperintensity (white arrows) consistent with vacuolation around the fourth ventricle. This is a very typical location. B. Axial T2-weighted
image 1 year later reveals some overall increase in the load of hyperintense abnormalities. A more distinct, focal lesion (white arrow) has enlarged in the
right middle cerebellar peduncle. C and D. Axial T2-weighted (C) and postcontrast axial T1-weighted (D) images obtained at age 6 years show interval
incomplete resolution of the presumed vacuolation. A partially enhancing, tumor (white arrow) is clearly evident in the right middle cerebellar peduncle.
Pathology revealed a pilocytic astrocytoma.

Barkovich_Chap06.indd 575 5/6/2011 9:03:50 PM


576 Pediatric Neuroimaging

FIG. 6-6. Medullary tumor-like lesion in NF1, with resolution. A. Axial T2 image shows focal enlargement and increased T2 signal in the left posterior
medulla (short arrows), consistent with a small tumor. A neurofibroma is evident beyond the left hypoglossal canal (long arrow). B. Axial T2-weighted image
5 years later shows nearly complete resolution of the medullary lesion, with only slight residual thickening. The neurofibroma (arrow) at the skull base persists.

FIG. 6-7. Midbrain juvenile pilocytic astrocy-


toma (biopsy proven) in a 14-year-old adolescent
with NF1. A. Postcontrast T1-weighted image
shows a sharply marginated, intensely enhancing
mass (white arrow) in the dorsal inferior midbrain.
B. Axial FLAIR image demonstrates the well-cir-
cumscribed hyperintense mass (white arrow) to
have minimal surrounding edema. C. Single voxel
long echo (TE = 270) spectroscopy of the tumor
reveals a very high Ch:Cr (3.49; normal < 1.5) and
presence of lactate (double arrows). These MRS
findings, worrisome for a malignant tumor, are
commonly observed in pilocytic astrocytomas.
N-acetylaspartate–to-creatine ratio (NAA: Cr)
is decreased (1.68; normal > 2). The NAA:Cr is
higher than expected for a glial neoplasm, likely
secondary to contamination of the voxel by sur-
rounding (normal) brain tissue as the voxel (2 × 2
× 2 cms size) is slightly larger than the tumor.

Barkovich_Chap06.indd 576 5/6/2011 9:03:51 PM


Chapter 6 • The Phakomatoses 577

FIG. 6-8. Probable hamartomatous enlargement of the brainstem. A. Sagittal T1-weighted image reveals diffuse enlargement of the pons and, especially,
medulla (white arrows); the signal intensity is normal. B. Axial T2-weighted image shows enlargement and heterogeneously increased signal in the dorsal
right pons and middle cerebellar peduncle, with slight mass-effect on the fourth ventricle (arrow).

course is common in NF1 patients with diffuse pontine tumors, a on follow-up scans. The T2 signal abnormalities partly regress during
tumor type that typically has a very poor prognosis (see Chapter 7) adolescence, though the brainstem remains enlarged. For more discus-
(54). Moreover, tumors of the mesencephalic tectum in patients with sion of brainstem tumors, see Chapter 7.
NF1 are usually biologically benign and may regress spontaneously Other common locations for astrocytomas (other than the optic
(55). Thus, management of brainstem tumors in children with NF1 system and tectum) in children with NF1 are the cerebellum (Fig. 6-5),
is generally not aggressive unless clinical progression is seen––isolated the corpus callosum, and the cerebral hemispheres (Fig. 6-9) (35,48,56).
radiologic progression is considered less serious. Astrocytomas are discussed extensively in Chapter 7. Hamartomatous
Mild enlargement of the pons, medulla, and middle cerebel- enlargement of the hypothalamus is occasionally seen in children with
lar peduncles is occasionally observed in children with NF1 in asso- NF1 (Fig. 6-10), though usually without association with precocious
ciation with, and likely the result of, extensive myelin vacuolation puberty or gelastic seizures. They project within the walls of the hypo-
(Fig. 6-8). This hamartomatous enlargement should not be misdiag- thalamus in the lower third ventricle (intrathalamic/sessile configura-
nosed as a brainstem glioma; differentiating features from infiltrat- tion). Like hypothalamic hamartomas in children without NF1, they
ing gliomas include normal or nearly normal signal in T1-weighted are nearly isointense to gray matter on T1- and T2-weighted images
images, only modest (and usually heterogeneous) signal increase on and do not enhance (57); they should not be confused for a chiasmatic
T2 images, absence of contrast enhancement, and lack of progression or hypothalamic glioma.

FIG. 6-9. Grade 2 (fibrillary)


glioma in the left temporal
lobe of a 10-year-old child with
NF1. A. Axial T2-weighted
image shows a tiny, hyperin-
tense lesion (white arrow) in
the white matter of the poste-
rior left temporal lobe. B. Axial
T2-weighted image a year later
shows an ovoid, well-defined
T2 bright focus in the same
location (arrow), representing
the enlarged tumor.

Barkovich_Chap06.indd 577 5/6/2011 9:03:53 PM


578 Pediatric Neuroimaging

FIG. 6-10. Hypothalamic hamartoma in a 15-year-old boy with NF1. A. Sagittal T1-weighted image shows a gray matter intensity mass (short white arrows)
in the floor the third ventricle, posterior to the chiasm (long white arrow) and anterior to the mammillary bodies (black arrow). B. Coronal T2-weighted
image reveals bilateral gray matter intensity mass (black arrows) in the floor and left lateral wall of the hypothalamus.

In a small but significant number of patients with NF1, two years of life. New lesions appear from late infancy to about age
hydrocephalus is present (58–60). The site of obstruction of CSF flow of 12 years (Fig. 6-11) (61); new lesions almost never develop after
is usually the aqueduct of Sylvius, resulting from benign aqueductal age of 15 years. The lesions start to regress, starting late in the first
stenosis, from extensive vacuolation of the tissues surrounding the decade of life (62,64), so that they are almost never seen in patients
aqueduct (see next section), or from tumors (astrocytomas or hama- over the age of 20 years (Figs. 6-11 and 6-12). In the peak age group
rtomas) of the adjacent tectum or tegmentum of the mesencepha- (6–12 years), they are seen in up to 90% of NF1 children (32,65,66).
lon. The differentiation of these causes of aqueductal obstruction is A pathologic analysis has shown that these are regions of myelin
extremely difficult with CT (59). The tectal lesions usually do not show vacuolation, where the layers of myelin become separated as they spi-
significant contrast enhancement when they are small and, therefore, ral around the axon (67). As might be expected, water diffusivity is
subtle calcifications or distortions of the posterior third ventricle and increased in these areas of T2 hyperintensity to a greater degree than
proximal aqueduct must be identified. The diagnosis of a tectal glioma in other areas of the brain (68).
is relatively simple using MRI. In contrast to benign aqueductal steno- The discovery that the oligodendrocyte myelin glycoprotein gene
sis (in which the proximal aqueduct is dilated and the tectum displaced is imbedded within the NF1 gene (13) and that neurofibromin is
superiorly and thinned, see Chapter 8), gliomas enlarge the tectum and required for Schwann cell myelination (12) further supports dysgen-
may completely obliterate the aqueduct. Occasionally, the tectum may esis of myelin as the cause of the white matter abnormalities in NF1.
appear short and thick in aqueductal stenosis because of mass effect The vacuolization may develop, therefore, in regions where the myelin
on the rostral aspect of the tectum by a dilated suprapineal recess (see is inherently abnormal; presumably the return of normal T2/FLAIR
Chapter 8). In these cases, the patient should be reevaluated after ven- signal on subsequent exams reflects myelin repair or remyelination
tricular decompression. (69). The increased diffusivity of white matter (even normal appearing
white matter) in NF1 (70,71) further supports the concept of abnormal
Characteristic Hyperintense T2 Lesions and myelin as an underlying cause.
White Matter Abnormalities In view of the frequency with which these T2/FLAIR hyperintense
MRI has disclosed the presence of characteristic hyperintense T2/ foci are encountered in children with NF1, close imaging follow-up
FLAIR lesions in pediatric patients with NF1, located in the brain- or biopsy is not indicated as long as they occur in the characteristic
stem, cerebellar white matter, basal ganglia (especially the globus pal- locations; are not hypointense on T1-weighted images; and lack associ-
lidus), thalamus, internal capsule, corpus callosum, and occasionally ated mass effect, edema, and enhancement. However, a growing astro-
the corona radiata (Figs. 6-6 to 6-9) (29,61,62); the subcortical white cytoma of the basal ganglia, pons, or cerebellum, when small, may
matter and centrum semiovale are not involved. These lesions are be indistinguishable from one of these lesions on noncontrast MRI
characteristically multiple; they have little or no mass effect (unless (Fig. 6-5)––especially in cases when there are numerous, confluent T2
extensive confluence is evident, in which case mild mass effect may hyperintense foci in some of the other expected locations. In this situ-
be detected, Fig. 6-8); they do not elicit vasogenic edema, their signal ation, it is recommended that the patients have a follow-up contrast-
appears normal on T1-weighted images, and they do not enhance enhanced MR scan, including diffusion-weighted imaging, perfusion
after intravenous administration of paramagnetic contrast. Diffu- imaging, or proton MR spectroscopy, 6 months to a year after the first
sion studies show increased diffusivity; anisotropy is normal but study. A tumor will typically enhance or show progressive enlargement
transverse eigenvalues are increased (63). Intermediate echo (TE = or mass effect on adjacent structures (Fig. 6-5); it may also have locally
144 msec) proton MR spectroscopy shows nearly normal NAA/Cr reduced diffusion in its solid portion (68), have increased blood vol-
and moderately increased Cho/Cr (46). Though there is wide indi- ume (72), and show elevated choline with decreased creatine and NAA
vidual variability, these lesions are typically absent in the first year or on proton spectroscopy (46,73–76).

Barkovich_Chap06.indd 578 5/6/2011 9:03:54 PM


Chapter 6 • The Phakomatoses 579

FIG. 6-11. Evolution of character-


istic focal T2 bright signal abnor-
malities in pediatric NF1. A and B.
Axial FLAIR images through the
posterior fossa (A) and the basal
ganglia (B) in a 4-year-old child.
Multiple small hyperintense foci
are seen within the deep cerebellar
hemisphere (arrows, A). Two small
lesions are evident in the right glo-
bus pallidus (short white arrows, B),
along with ill-defined hyperinten-
sity in the left globus pallidus (long
white arrow) and the left side of the
splenium (black arrow). C and D.
Corresponding axial T2-weighted
FLAIR images in the same patient
at age 12 years. The previous signal
abnormalities in the deep cerebel-
lum have mostly resolved, with a
few new white matter foci evident
(white arrows, C). New T2 bright
foci have developed in the right
globus pallidus (long white arrow,
D) and the bilateral posterior thal-
ami (short white arrows, D); the left
globus pallidus lesion is no longer
visible.

FIG. 6-12. Partial resolution of


basal ganglia signal abnormalities in
NF1. A. Axial T2-weighted image at
age 7 years shows multiple bilateral
hyperintense abnormalities (white
arrows) in the globi pallidi and inter-
nal capsules. B. Axial T2-weighted
image 4 years later shows significant
regression of the abnormalities.

Barkovich_Chap06.indd 579 5/6/2011 9:03:55 PM


580 Pediatric Neuroimaging

FIG. 6-13. T1 prolongation in the basal ganglia in NF1. A and B. Axial T2- and T1-weighted images show typical hyperintensity on both T1- and
T2-weighted images in the bilateral globi pallidi. The T2 hyperintensity in the posteromedial thalami (white arrows) is also typical.

The characteristic T2 hyperintense foci seen in the globi pallidi of lesions described above (66). NF1-related hippocampal T2 changes
patients with NF1 have a somewhat different radiologic appearance should not be confused with hippocampal sclerosis (in which case the
than the cerebellar foci in that they are sometimes slightly hyperintense hippocampal volume is decreased) or with neoplastic infiltration.
on T1-weighted images (Fig. 6-13) (62,69,77,78). (Calculation of T1 In addition to these focal lesions, many patients with NF1 have both
values shows that the frontal white matter, caudate nuclei, putamina, an increased volume of cerebral white matter and a larger midsagittal
and thalami also have some T1 shortening [78], but it is not appar- corpus callosum area than age-matched controls (Fig. 6-15) (81,82);
ent to visual inspection in these other areas). The hyperintensity on not surprisingly, these findings are more pronounced in patients with
T1-weighted images develops after the T2 prolongation appears and
persists after T2 prolongation disappears, a time course that suggests
that the T1 shortening represents delayed or reactive formation of
myelin (69). This view is supported by the finding of increased cho-
line and decreased NAA in the thalami of patients with the charac-
teristic T2 prolongation of the globus pallidus, a finding that may
reflect remyelination and neuropil injury in regions of dysmyelination
(79). The characteristic lack of enhancement, location, bilaterality,
and T1 hyperintensity help differentiate these presumed hamartomas
from astrocytomas. However, as with all small, nonenhancing foci of
T2 prolongation in the brain, the biological behavior of white matter
lesions in NF1 cannot be predicted with complete accuracy by imaging
alone. Benign-appearing lesions can degenerate into neoplasms (80),
and neoplastic-looking lesions can shrink or disappear (40–42). Also
remember that focal lesions developing in the cerebral cortex or in the
subcortical or adjacent white matter do not represent characteristic T2
bright lesions of pediatric NF1 (Fig. 6-9) (66); if present, these should
be followed by sequential imaging, as they may potentially represent
low-grade neoplasms.
Occasionally, the hippocampi of pediatric patients with NF1
can show diffusely increased signal on T2/FLAIR images, often with
increased volume (Fig. 6-14); this finding can be unilateral or bilateral,
is not progressive, and persists throughout childhood and adolescence FIG. 6-14. High-resolution coronal FSE T2-weighted image reveals
(66). The nature of the hippocampal abnormality is not known; how- hyperintense T2 signal of both hippocampal formations (white arrows)
ever, is not likely to represent myelin vacuolation as these hippocampi (compare signal to that of adjacent temporal cortex). The right hippocam-
have a different temporal evolution than the NF1 dysmyelinating pus is brighter and larger than the left.

Barkovich_Chap06.indd 580 5/6/2011 9:03:57 PM


Chapter 6 • The Phakomatoses 581

macrocephaly (∼40% of those with NF1 [78]). In a recent study, a


larger corpus callosum was associated with significantly lower IQ (83).
It is postulated that the abnormal increase in callosal volume results
from increased myelination of callosal axons or redundancy of fibers
connecting the cerebral hemispheres (due to reduced apoptosis) (83).

Vascular Dysplasia
Dysplasia of the cerebral vasculature is increasingly recognized in chil-
dren with NF1; it develops at a young age, with prevalence of up to 6%
(84–86). The known significant incidence of vascular abnormalities in
the cerebral vasculature and in other parts of the body makes vascular
dysplasia a distinct clinical entity. Any child with NF1 who has seizures,
mental retardation, paralysis, or severe headaches may have a vascular
dysplasia, particularly in the absence of a brain tumor or hydrocepha-
lus. However, clinically, a majority of NF1 patients with vascular dys-
plasia do not have focal deficits as a result of their cerebral arteriopathy
by the time the dysplasia is diagnosed with MRI. Intracranial vascular
dysplasias can also complicate irradiation for optic nerve or optic chi-
asm gliomas (presumably a radiation arteritis; see discussion in Chap-
FIG. 6-15. Sagittal T1-weighted image shows an abnormally thick corpus ter 3). NF1-related vascular dysplasia can progress overtime (Fig. 6-16)
callosum (white arrows). The ventral pons (black arrows) is prominent. and may require revascularization surgery (84,86).

FIG. 6-16. Progressive NF1-related


vasculopathy. A. Axial T2-weighted
image at age 18 months shows nor-
mal appearing signal voids in the
basilar and proximal anterior and
middle cerebral arteries––note the
normal right MCA (arrow). B. Axial
T2-weighted image 2 years later
shows diminished size of the right
MCA signal void (white arrow).
C. Coronal reconstructed MIP
image from a 3D TOF MRA reveals
a lack of flow-related enhancement
in the right MCA (arrows), con-
sistent with an acquired complete
occlusion (or very severe stenosis).

Barkovich_Chap06.indd 581 5/6/2011 9:03:59 PM


582 Pediatric Neuroimaging

FIG. 6-17. Vascular dysplasia in NF1. A. Axial T2-weighted image shows absence of the left cavernous carotid artery. B. Arteriogram of the aortic arch
shows absence of the left common carotid artery origin and significant stenosis (arrow) of the proximal left subclavian artery.

Most commonly the dysplasia consists of intimal and smooth the possibility of “overlap” syndromes (syndromes with characteristics
muscle proliferation, with resultant stenosis or occlusion, in the carotid of both NF1 and NF2) is often raised (89,90). Although patients with
(common or internal), proximal middle cerebral, or proximal anterior features of both NF1 and NF2 clearly exist, the true implications of the
cerebral arteries (Figs. 6-16 and 6-17). Moyamoya phenomenon with overlap in affected patients are not known (91).
marked enlargement of the lenticulostriate arteries (which function as
collateral vessels) is seen in many such patients (87). (Moyamoya is Calvarial and Orbital Abnormalities
discussed further in Chapter 12.) Fusiform arterial dilation, cerebral Other cranial/intracranial manifestations of NF1 include deficiency of
aneurysms, and arteriovenous fistulas are also described, although less the sphenoid wing and of the occipital bone along the lambdoid suture.
commonly (84,87,88). The dysplasias can be difficult to detect on stan- Only sphenoid dysplasia is of any clinical importance, as it allows her-
dard CT or MR images; with MR, close inspection of the arterial signal niation of the temporal lobes into the orbit (Fig. 6-18). The pulsations
voids (Fig. 6-16), especially the distal carotid arteries and the circle of of the temporal lobe may be transmitted through the globe, thus being
Willis (to detect narrowing of the cavernous or supraclinoid carotids visible externally as pulsatile exophthalmos (or enophthalmos sec-
and proximal middle and anterior cerebral arteries), is essential in ondary to atrophy of orbital contents). The globe may be dysplastic,
patients with NF1 to avoid delays in clinical detection of these lesions; enlarged (buphthalmos), or hypoplastic. Sphenoid wing deficiency is
note that the intracranial vessels are poorly seen on FLAIR images, so nearly always associated with plexiform neurofibromas (PNs) in the
acquisition of T2-weighted spin-echo images should be strongly consid- orbit or periorbital regions (Fig. 6-18). Recent work indicates that the
ered when imaging children with NF1, particularly those with declining abnormalities of the bony orbit may progress over time, suggesting
function. If vascular dysplasia is suspected, MR (or CT) angiography that these are not simple dysplasias of bone, as previously postulated,
should be performed; conventional, catheter angiography is usu- but may in part result from erosions by the adjacent neurofibromas
ally reserved for a presurgical workup. Angiography generally shows (Fig. 6-18) or optic nerve tumors (92,93).
either severe stenosis or occlusion of the proximal cerebral vessels
(Fig. 6-17). Most modern MR scanners allow excellent angiography Neurofibromas and Plexiform Neurofibromas
of the large and medium-sized intracranial vessels, and CT angiogra- Another cause of intracranial complications in NF1 is excessive growth
phy is also excellent if helical scanners are used; therefore, these should of craniofacial PNs. PNs are locally aggressive congenital lesions com-
be the modalities of choice if angiography is to be performed. MRI posed of tortuous cords of Schwann cells, neurons, and collagen in a
is preferred over CT in order to avoid exposure to ionizing radiation disorganized intercellular matrix (48). They tend to progress along the
(see Chapter 1 for technical details). nerve of origin (usually small, unidentified nerves) into the intrac-
ranial space, causing distortion and compression of the brain. Most
Cranial Nerve Tumors commonly, PNs develop in the orbit where they act as masses, causing
Cranial nerve tumors are uncommon in NF1, which mainly causes impaired ocular movements and exophthalmos (Fig. 6-19). The neck
glial tumors and neurofibromas of peripheral nerves. Schwannomas is another common location for neurofibromas, with an estimated
are much more common in NF2 (see next section). The only cranial occurrence of 25% to 30% in patients with NF1 (Fig. 6-20) (94). Neck
nerve that is frequently involved in NF1 is the optic nerve, which has masses, their appearance, and their differential diagnoses are discussed
been discussed above and is, in fact, a diencephalic white matter tract in Chapter 7. The growth rate of PNs is unpredictable; they grow more
that has been misnamed as a cranial nerve. The other cranial nerves are quickly in young children and during adolescence (7,95,96). Soli-
rarely, if ever, involved. When schwannomas occur in patients with NF1, tary neurofibromas have slightly greater signal intensity than skeletal

Barkovich_Chap06.indd 582 5/6/2011 9:04:00 PM


Chapter 6 • The Phakomatoses 583

FIG. 6-18. Dysplasia of the


sphenoid wing with associated
plexiform neurofibroma. A. Axial
T2-weighted image shows an
extensive neurofibroma (white
arrows) infiltrating the supe-
rior orbit and the left temporal
fossa (the V1 distribution). The
greater wing of the sphenoid
bone is dysmorphic, result-
ing in anterior extension of the
contents of the left middle cra-
nial fossa. B. Axial T2-weighted
image at a slightly lower level
shows the inferiorly displaced,
proptotic left globe. Neurofi-
broma is also evident within an
enlarged pterygopalatine fossa
(V2 distribution, white arrows).

muscle on T1-weighted sequences. On T2-weighted sequences, the On both MRI and CT, PNs appear as masses that most commonly
lesions have variable signal intensity. Most commonly, the periphery arise in the region of the orbital apex or superior orbital fissure (in the
of the lesions tends to be of high T2 signal intensity with respect to distribution of the first division of the trigeminal nerve, Fig. 6-19) but
muscle, whereas the center of the lesions is often of low signal inten- can occur anywhere in the body. They tend to be of low attenuation
sity (97–99); this has been referred to as the “target sign” (Fig. 6-20) on CT and generally show little enhancement after administration of
(100). The central area of decreased T2 intensity is probably related to intravenous contrast. On MRI, the masses are heterogeneous, display-
the known dense central core of collagen within these lesions (98,99). ing mostly low signal intensity as compared with brain on T1-weighted
Collagen has a low mobile proton density and therefore is of low signal images and high signal intensity on T2-weighted images (Fig. 6-19)
intensity on T2-weighted images. (101). Enhancement after administration of paramagnetic contrast

FIG. 6-19. Plexiform neurofibroma and multiple solitary neurofibromas. A. Axial T2-weighted image shows innumerable small subcutaneous neuro-
fibromas (small open arrows). Note the “target sign” appearance of the large left infratemporal lesion (curved white arrow). In addition, PNs (large solid
white arrows) are present in the inferior orbits bilaterally and extend (large open white arrows) posteriorly into the cavernous sinuses, left greater than right.
B. Postcontrast axial T1-weighted image with fat suppression shows inconsistent enhancement of the neurofibromas. Note that enhancing tumor extends
posteriorly from the left cavernous sinus along the cisternal segment (arrow) of the fifth cranial nerve. C. Postcontrast axial T1-weighted image with fat
suppression at a level below (B) shows multiple enhancing tumors throughout the face and infratemporal fossae.

Barkovich_Chap06.indd 583 5/6/2011 9:04:01 PM


584 Pediatric Neuroimaging

FIG. 6-20. Malignant nerve sheath tumor developing within a plexiform neurofibroma in a 15-year-old adolescent who recently experienced lower neck
pain. A and B. Axial (A) and coronal (B) T2-weighted images with fat suppression reveal a mass in the right brachial plexus. Central target signs (black
arrows) are evident in the more superficial aspects of the lesion, characteristic of plexiform neurofibroma. In the deeper portion of the mass, a larger conflu-
ent area (white arrows) without target sign is evident. Malignant nerve sheath tumor was confirmed by needle biopsy of this area.

is variable, although at least a portion of the tumor usually enhances (105). The appearance of intramedullary astrocytomas in children with
(usually the central aspect, corresponding to central the T2 hypoin- NF1 is no different from that of other intramedullary astrocytomas
tense [“target”] components [Fig. 6-20]). Careful evaluation of the (see Chapter 10). In the absence of any neurologic signs or symp-
images often reveals extension of orbital tumors into the cavernous toms, the incidence of an underlying spinal cord disorder is very low
sinus, nasopharynx, or pterygomaxillary fissure (Fig. 6-19). in children with dextroscoliosis (convexity of the curvature to the
right). However, children with levoscoliosis, especially if it is rap-
idly progressive, associated with pain, or associated with neurologic
Spinal Manifestations
deficits, have a significantly higher incidence of underlying spinal
Scoliosis and Intramedullary Tumors cord abnormalities (106). Imaging of scoliosis is discussed further
Abnormal curvature of the spinal column (scoliosis, kyphosis) is the in Chapter 9.
most common skeletal abnormality reported in patients with NF1. In
Holt’s series, it was present in 32% of affected children; the incidence Dural Dysplasia and Meningoceles
increases with age (102). The abnormal curvature is usually mini- Dural ectasia is identified in 75% of patients with posterior vertebral
mal or mild in degree, and can resemble idiopathic adolescent sco- scalloping, and in 25% of those with lateral scalloping. Lateral menin-
liosis. It can be severe, in which case dystrophic curves are commonly goceles are diverticula of the thecal sac (most commonly at the tho-
encountered, with potential for rapid progression (Fig. 6-21) (103). racic level) that extend laterally through widened neural foramina.
Dystrophic curves are associated with a high incidence of paraspi- Most authorities feel that the meningoceles result from a primary
nal or other internal neurofibroma next to the vertebra. Dysplasia mesodermal dysplasia (104,107); the primary abnormality in affected
of the vertebral bodies is common, as are hypoplasia of the pedicles, patients may also be hypoplasia of the pedicles (which makes it pos-
transverse processes, and spinous processes; scalloping of the poste- sible for the dural sac to herniate laterally in response to spinal fluid
rior aspects of the vertebral bodies; and hyperplastic bone changes pressure). Weakness in the meninges allows the thecal sac to be focally
(94,104). It is not certain whether these bony anomalies result from stretched in response to CSF pulsations. The protrusions from the the-
a primary mesodermal dysplasia or are secondary to the effects of cal sac slowly erode the bony elements of the neural foramina, allowing
nerve sheath tumors. Plain film radiography is essential to demon- the meningoceles to form. The thoracic region is thought to be pri-
strate the scoliosis optimally. CT is the optimal modality for demon- marily affected because of the lesser development of the paravertebral
strating the changes of the individual vertebrae, as it shows superior muscles and the relatively high pressure difference between the nega-
bone detail. However, most of the bony changes are well visualized tive pressure of the thorax and the spinal CSF (108). The CT appear-
with high quality MR studies. ance of a lateral meningocele is that of a wide neural foramen with
When abnormal spinal curvature is present, the question often a CSF-attenuation dumbbell-shaped mass extending through it. The
arises as to whether it results from the dysplastic bone of neurofibro- corresponding vertebral body is usually markedly scalloped (109). The
matosis or from an intrinsic spinal cord lesion such as a tethered cord, MR appearance of these lesions is similar in that the neural foramen is
syringomyelia, or intrinsic spinal cord tumor. If an intrinsic spinal cord markedly widened and the spinal canal is enlarged as a result of scal-
tumor is present in a patient with NF1, it is likely to be an astrocytoma loping of the posterior vertebral body at the level of the meningocele

Barkovich_Chap06.indd 584 5/6/2011 9:04:03 PM


Chapter 6 • The Phakomatoses 585

FIG. 6-21. Rapid progression of cervical kyphosis (without surgical


intervention) in NF1. A. Sagittal T1-weighted image at age 10 years
shows a focal kyphotic deformity in the midcervical spine (black
arrows) secondary to dysplasia and erosions of the adjacent cervical
vertebrae. B. Postcontrast axial T1-weighted image with fat suppression
through the midportion of the kyphosis reveals enhancing, infiltrat-
ing plexiform neurofibroma in the prevertebral space (short arrows),
extending laterally into the right vertebral foramen (long arrow). The
anterior aspect of the vertebral body is eroded (white arrowheads). The
subarachnoid spaces (black arrowheads) along the posterior/right lat-
eral aspect of the cord are enlarged secondary to the dural ectasia and
kyphotic deformity. C. Sagittal T1-weighted image 6 years later reveals
marked progression in kyphotic angulation.

(Fig. 6-22). The meningocele is isointense with CSF on all imaging (111). Spinal neurofibromas are far less common in children with NF1
sequences. Differentiation from neurofibromas can be made by not- than in adults (110). In addition, spinal nerve sheath tumors are less
ing the absence of the central low intensity focus on the T2-weighted commonly symptomatic in NF1 (only 1%–2% are symptomatic [110])
images (98). Neurofibromas are of higher signal intensity than CSF on than in NF2 (30%–40% symptomatic [112]). Scoliosis appears to be
T1-weighted images (compare Fig. 6-22 with Fig. 6-23). more common in NF1 patients with spinal neurofibromas than in
those without (113). About 90% of these tumors are extradural, with
Nerve Sheath and Other Soft Tissue Tumors more than half being intraforaminal (110). In contrast, most nerve
Another major abnormality affecting the spine in NF1 is intraspinous sheath tumors in NF2 are intradural (114).
or paraspinous neurofibromas (48). Spinal neurofibromas develop in a There has been debate in the pathology literature as to whether
large number of patients with NF1, in all segments of the spine (110). the paraspinous tumors in NF1 are schwannomas or neurofibro-
In some families, neurofibromas are nearly entirely limited to the spine mas; most nerve sheath tumors in NF1 appear to be neurofibromas

Barkovich_Chap06.indd 585 5/6/2011 9:04:04 PM


586 Pediatric Neuroimaging

FIG. 6-22. Scoliosis, dural ectasia, and lateral meningoceles


in NF1. A. Coronal T2 weighted image shows a focal cur-
vature in the upper thoracic spine, and two moderate
sized lateral meningoceles (white arrows) extending out
of the spinal canal through widened upper thoracic neu-
ral foramen. B. Sagittal T2-weighted image shows scallop-
ing of the posterior vertebral bodies (white arrowheads).
C. Axial T2-weighted image through the level of the lower
meningocele shows displacement of the spinal cord to the
left (white arrows).

(97). Schwannomas are uncommon in NF1; conversely, neurofibro- neurofibromas can be seen in patients unaffected by NF1, patients
mas are fairly distinctive features of von Recklinghausen disease and with von Recklinghausen disease can have neurofibromas of vary-
rarely occur in any other setting. It is probably not possible to dif- ing sizes at many levels throughout the spinal canal (35,98). Patients
ferentiate these tumor types by imaging; moreover, there are argu- with NF1 typically have only a few (5 or 6) nerve sheath tumors. In
ments as to the exact pathology and pathogenesis of these tumors contradistinction, patients with NF2 typically have many (more than
(35,48,115,116). Neurofibromas are tumors of the nerve sheaths that 10) spinal nerve schwannomas (see Section “Imaging of Spinal Mani-
also involve the nerves themselves; they have a higher content of col- festations” on NF2 below).
lagen and elastin than schwannomas (115). The cell of origin of these The MR appearance of neurofibromas is that of ovoid lesions that
tumors is thought to be the fibroblast (35). Although isolated spinal may be entirely within the spinal canal, causing canal enlargement, or

Barkovich_Chap06.indd 586 5/6/2011 9:04:05 PM


Chapter 6 • The Phakomatoses 587

FIG. 6-23. Extensive plexiform and spinal neurofibromas with cord compres-
sion in NF1. A. Coronal T2-weighted image of the lumbar spine and upper
pelvis shows extensive plexiform neurofibroma (subcutaneous and muscular
spaces). Intraspinal involvement is evident at the level of the kidneys (black
arrows). B and C. Axial FSE T2-weighted image (B) and postcontrast axial SE
T1-weighted image (C) through the upper lumbar spine show large bilateral
neurofibromas (n) expanding the spinal canal and neural foramina; the intra-
dural component of the right-sided neurofibroma displaces the thecal sac to
the left (white arrows). Extensive PNs infiltrate the retroperitoneum.

may extend outward from the spinal canal through widened neural elongated in the anterior-posterior direction (Fig. 6-25). Under these
foramina (Fig. 6-23) (97,98). When small, intradural neurofibromas circumstances, the risk of damaging the cord during an attempted sur-
appear as nodules of soft tissue along the nerves roots, most commonly gical decompression is very high.
identified along the cauda equina (Fig. 6-24). Extension through the It is well established that malignant peripheral nerve sheath tumor
neural foramen is accompanied by enlargement of the neural foramina (MPNST, (neurofibrosarcomas) occurs in NF1; most arise within preex-
by pressure erosion of the bone. Neurofibromas shows a slightly greater isting PNs (35,48,117). Patients with NF1 harbor a 10% lifetime risk of
signal intensity than skeletal muscle on T1-weighted sequences and developing a MPNST (7,48,117). The risk of malignancy increases with
high intensity periphery with variable intensity center on T2-weighted age. Neurofibrosarcomas usually present with localized motor deficits
sequences (97–99), sometimes resulting in the aforementioned “tar- or pain and dysesthesias with or without evidence of a rapidly enlarg-
get sign” (Fig. 6-23) (100). On CT, the paraspinal neurofibromas have ing mass. Currently, 18F-fluorodeoxyglucose (FDG) positron emission
low attenuation when compared to muscle. Intraspinal neurofibromas tomography (PET) imaging seems to be the best way to differentiate
(which can be intrathecal or extrathecal) may displace the spinal cord benign from malignant nerve sheath tumors, with malignant tumors
or nerve roots of the cauda equina to the contralateral side of the canal showing significantly higher uptake of FDG than benign tumors (118).
(Fig. 6-23). When bilateral neurofibromas are present at a single level, The MR features of benign and malignant nerve sheath tumors are not
the cord can be compressed into a narrow, central band of tissue that is always sufficiently different to distinguish them. Both can be large and

Barkovich_Chap06.indd 587 5/6/2011 9:04:06 PM


588 Pediatric Neuroimaging

FIG. 6-24. Multiple small intraspinal


neurofibromas. A. Sagittal T2-weighted
image shows multiple small nodular soft
tissue intensity foci (white arrows) in the
subarachnoid space. B. Axial postcontrast
T1-weighted image shows that the lesion
(arrow) at the sacral level enhances.

relatively well circumscribed, and neither commonly invades adjacent


structures. MR findings indicative of a MPNST (Fig. 6-20) includes
NEUROFIBROMATOSIS TYPE 2
irregular tumor shape, unclear margins, intratumoral lobulation, pres- Clinical Manifestations
ence of high signal intensity foci on T1-weighted images, lack of target
sign, heterogeneous enhancement, and a lower rate of enhanced area Neurofibromatosis type 2 (NF2) has also been called neurofibroma-
(98,100,119). Of note, marked heterogeneity can be seen in benign tosis with bilateral acoustic schwannomas. NF2 is a separate disease
tumors as well (101), so this is not a specific imaging sign. altogether from NF1. NF2 is associated with an abnormality of chro-
mosome 22 (critical region in about 6 megabases within 22q12 [120]),
verifying that it is a separate entity from von Recklinghausen disease
(NF1), which has a locus on chromosome 17 (4,121,122). The prod-
uct of the NF2 gene is called merlin (moesin-ezrin-raxidin-like pro-
tein, also called schwannomin); it is a tumor suppressor gene that is
believed to control interactions between affected cells and surround-
ing structures (both cells and molecules) in the extracellular matrix.
Mutated merlin inhibits cell adhesion, which is important for regu-
lating cell growth, and is more soluble than normal protein, making
its interaction with the cytoskeleton unstable (123,124). A correlation
has been identified between the type of mutation of the NF2 gene
and the phenotype of the affected patients in that nonsense and frame
shift mutations cause more severe disease (younger age at onset and
diagnosis, more tumors), than missense mutations or mild deletions
(125,126).
NF2 is also autosomal dominant, but it is much less frequent than
NF1, with an estimated incidence of 1 in 50,000 (127). About 50% of
cases represent new mutations, and as many as one third are mosaic
for the underlying disease causing mutation (127). The major feature
of NF2 is the presence in nearly all affected individuals of bilateral
vestibular schwannomas (Figs. 6-26 and 6-27). Other tumors of the
CNS, particularly meningiomas and other schwannomas (Fig. 6-28),
may be present as well. (Most children with meningiomas do not
have NF2; however 72% of pediatric meningiomas harbor deletions
of the NF2 gene [128]). In contrast to the situation in adults, hearing
FIG. 6-25. Bilateral C-1 to C-2 spinal neurofibromas with spinal cord loss is an uncommon presentation in childhood; seizures (caused by
compression in NF1. Axial T2-weighted image shows neurofibromas (white meningiomas) and facial nerve palsy are much more common pre-
arrows) that compress the thecal sac and spinal cord (small white arrow- senting symptoms, as are neurologic symptoms related to brainstem
heads). An intradural neurofibroma component is evident on the right and/or spinal cord tumors (127,129,130). Tumor load tends to be
(large white arrowhead) (asterisk denotes odontoid process of C-2). extensive in pediatric patients; cranial meningiomas are seen in 60%

Barkovich_Chap06.indd 588 5/6/2011 9:04:07 PM


Chapter 6 • The Phakomatoses 589

FIG. 6-26. Small bilateral acoustic schwannomas in NF2. A. Postcontrast axial T1-weighted image shows bilateral enhancing masses (arrows) filling the
internal auditory canals. On the right, the mass extends into the vestibule (arrowhead). B. Axial steady-state acquisition T2-weighted image (FIESTA) shows
replacement of hyperintense CSF signal in the internal auditory canals by the hypointense schwannomas (arrows), and a filling defect in the right vestibule
(arrowhead) from intralabyrinthine tumor extension.

of cases, cranial schwannomas (other then vestibular) in 36%, and in number (112). When present, however, cutaneous schwannomas
spinal schwannomas and meningiomas each in 80% (130). Cutane- tend to develop during the first decade of life, before the cranial nerve
ous manifestations are much less frequent than in NF1. Only about schwannomas, which typically do not develop until the age of 10 to
25% of affected patients will have café au lait spots and, when pres- 15 years (131–133). Subcapsular cataracts are present in more than
ent, the spots are pale and few in number (< 5) (112,131). Cutane- half of patients, and may be present during childhood (112,134).
ous nerve sheath tumors (predominantly schwannomas) are seen Therefore, skin tumors and cataracts are valuable clues in the early detec-
in about 65% of affected patients, but are minimal in size and few tion of children with NF2 (131–133). The established diagnostic criteria

FIG. 6-27. Bilateral acoustic schwannomas in an adolescent


with NF2. Postcontrast axial SE T1-weighted image with fat
suppression shows bilateral nerve VIII schwannomas (white
arrows) expanding the internal auditory canals, extending
into the cerebellopontine angle cisterns, and compressing the
middle cerebellar peduncles, especially on the left.

Barkovich_Chap06.indd 589 5/6/2011 9:04:08 PM


590 Pediatric Neuroimaging

FIG. 6-28. NF2 with multiple schwannomas. A. Postcontrast axial T1-weighted image of NF2 patient reveals bilateral trigeminal schwannomas. The left-
sided schwannoma is limited to the cisternal portion of the nerve (white arrow), while the right-sided one involves the cisternal portion (small black 5) and
the trigeminal ganglion (large black 5). B. Postcontrast axial T1-weighted image shows bilateral eighth nerve schwannomas (white arrows), schwannoma of
the left temporal branch of the third division of the fifth cranial nerve (white arrowhead), and the large schwannoma involving the right trigeminal ganglion
(black 5). C. Postcontrast axial T1-weighted image at a slightly more inferior level shows, in addition to the fifth nerve tumors described in (B), a ninth nerve
schwannoma (white arrow) in the cerebellomedullary cistern. D. Postcontrast coronal T1-weighted image shows the large right-sided fifth nerve schwan-
noma (white arrows) extending through and expanding the right foramen ovale.

for NF2 are listed in Table 6-4. It should be noted that these criteria cranial nerve schwannomas and not more than one other tumor (men-
do not work well in children or in patients with no family history of ingioma or schwannoma). Type 2, also called Wishart’s form of NF2
NF2 (as many as 50% of affected patients [127,135]), and need revi- or the Wishart-Lee-Abbott form, is a more severe form characterized
sion. Indeed, less than 20% of patients with NF2 are diagnosed during by early onset and the presence of multiple tumors, including schwan-
childhood (133). However, early diagnosis has important implications, nomas, meningiomas, ependymomas, and sometimes astrocytomas.
as age at diagnosis is the strongest single predictor of relative risk of Patients with Type 2 NF2 have a higher incidence of cataracts and skin
mortality and is a useful index for patient counseling and clinical man- tumors, as well as CNS tumors, than those with Type 1 NF2 (112).
agement (136). These subgroups have proved useful for prognostic purposes; however,
Some authors have noted that NF2 can be subdivided into two large the reader should remember that, as with nearly all such groupings,
groups (137). Type 1, also called Gardner’s form of NF2, is the mildest many patients with NF2 have intermediate severity of the disease and
form of the disease with late onset. Patients have slowly growing eighth may not fall neatly into either form.

Barkovich_Chap06.indd 590 5/6/2011 9:04:10 PM


Chapter 6 • The Phakomatoses 591

The imaging characteristics of schwannomas and meningiomas are


TABLE 6-4 Criteria for Diagnosis of described in Chapter 7. If bilateral acoustic tumors are present or a
diagnosis of NF2 is suspected for other reasons, the imaging procedure
Neurofibromatosis Type 2 of choice should be contrast-enhanced MRI of the entire brain and
spine, with thin (≤3 mm) sections through the posterior fossa. Schwan-
Main Criteria: nomas and meningiomas both enhance brightly after the infusion of
(1) Bilateral tumors of the 8 cranial nerves demonstrated by intravenous contrast. Multiple very small meningiomas and schwan-
contrast-enhanced MRI. nomas that were not apparent on the precontrast scan are frequently
(2) A parent, first-degree relative, or offspring with NF2 and a identified on the postcontrast study (62,112).
unilateral eighth nerve tumor at less than age of 30 years or 2 An important point is that schwannomas and meningiomas are
of the following: unusual tumors in children and young adults (under age 30 years).
If a meningioma or schwannoma is seen in a young patient, a contrast-
• Neurofibroma enhanced MR scan should be obtained through the brain to look for other
• Meningioma asymptomatic schwannomas or meningiomas that may aid in establish-
ing the diagnosis of NF2. Regularly repeated contrast-enhanced MR
• Glioma
scans are recommended for patients in whom the disease is estab-
• Schwannoma lished, as new tumors continue to develop throughout life and surgical
• Juvenile lens opacity (posterior subcapsular cataract of or gamma knife therapy is necessary when they grow large enough to
cortical cataract) cause hydrocephalus or symptoms.
Additional Criteria:
(1) A unilateral vestibular schwannoma diagnosed before the age
Imaging of Spinal Manifestations
of 30, plus any two of the following: The characteristic spinal manifestations of NF2 are multiple paraspi-
• Meningioma nal nerve sheath tumors (mostly schwannomas with some neurofi-
bromas), intraspinal meningiomas, and intramedullary spinal cord
• Glioma tumors (112,125,130). Spinal tumors affect close to 75% of patients
• Schwannoma (112,129,130). Intraspinal and paraspinal nerve sheath tumors are
• Juvenile cortical cataract very common in patients with NF2 (97,112) and can nearly always be
detected with proper imaging technique in patients beyond the age of
• Juvenile lens opacity (posterior subcapsular cataract of 15 years. Because they have more extramedullary spinal tumors, patients
cortical cataract), or with NF2 more frequently develop symptoms of cord compression from
(2) Multiple meningiomas (2 or more) and a unilateral vestibular the multiple extramedullary tumors than do patients with NF1. Coro-
schwannoma nal imaging optimally shows the nerve root tumors and their relation to
the spinal cord. Nerve sheath tumors may be intramedullary, extramed-
(3) Multiple meningiomas (2 or more) and any 2 of the following:
ullary intraspinal, extraspinal, or may involve both the intraspinal and
• Glioma extraspinal compartments in addition to the intervening neural fora-
• Neurofibroma men. The tumors are isointense to neural tissue on T1-weighted images,
hyperintense (usually) on T2-weighted images, and enhance uniformly
• Schwannoma
after intravenous administration of paramagnetic contrast (Fig. 6-29E).
• Cataract As with NF1, axial images are helpful in evaluating any deformity of the
cord and the relationship of the tumors to the cord.
Manchester criteria (Modified NIH criteria). Adapted from References 127
Intrinsic spinal cord tumors and syringohydromyelia occur with
and 135.
an increased incidence in NF2 (105,117). The most common intrinsic
cord tumors are ependymomas, although astrocytomas and intramed-
ullary schwannomas are seen (125). These can be solitary (most often
involving the conus medullaris and filum terminale), or multiple,
The mainstay of management of NF2 is surgical removal of symp- occurring at all levels of the neural axis. Contrast-enhanced MR is the
tomatic cranial and spinal tumors. However, there are recent reports imaging modality of choice. Without the administration of contrast,
of shrinkage of vestibular schwannomas and improvement of hearing ependymomas of the spinal cord may be difficult to differentiate from
with therapies targeted to the intracellular signaling pathways involved multiloculated syringohydromyelia. Ependymomas and astrocytomas
in tumorogenesis (138). of the spinal cord are sometimes difficult to distinguish by CT and
MRI. However, the presence of a centrally located, contrast-enhancing
tumor with sharply marginated borders suggests the diagnosis of
Imaging of Intracranial Manifestations
ependymoma. Further characteristics of intramedullary neoplasms are
As a result of the absence of cutaneous and ocular manifestations, described in Chapter 10.
patients with NF2 may not develop any clinical manifestations of the Meningiomas are common in the intraspinal as well as intracranial
disease until the second, third, or even fourth decade of life (112,132). compartment in NF2 (117). As in patients without neurofibromatosis
Moreover, it may fall upon the radiologist to make the diagnosis of NF2 (see Chapter 10), spinal meningiomas are most common in the tho-
in affected patients. The characteristic intracranial abnormalities are racic region. They are intradural, extramedullary masses that displace
schwannomas of the vestibular (Figs. 6-26 and 6-27) and other cranial the spinal cord as they grow. These dural-based masses will sometimes
nerves (most commonly the oculomotor and the trigeminal nerves) cause pressure erosion of the adjacent bone. They can be identified
(Fig. 6-28) and meningiomas (often multiple, Figs. 6-28 and 6-29). as extramedullary, intradural masses on CT with intrathecal or IV

Barkovich_Chap06.indd 591 5/6/2011 9:04:11 PM


592 Pediatric Neuroimaging

FIG. 6-29. NF2 with bilateral acoustic schwannomas and multiple meningiomas. A. Axial postcontrast T1-weighted image through the posterior fossa shows
enlarged, enhancing seventh and eighth nerve complex on the left (solid white arrow). Enhancement is also seen in the genu of the left facial nerve (open white
arrow) and at the right porus acousticus (black arrow). B–D. Axial and coronal postcontrast images show multiple meningiomas, including bilateral intraven-
tricular meningiomas (arrows in B). E. Postcontrast sagittal T1-weighted image shows innumerable schwannomas arising from the cauda equina.

contrast. MRI is the preferred diagnostic modality; sagittal and coronal space (see Chapter 9) (142,143). The syrinx will often disappear after
images are essential to visualize the extent of the tumor and its rela- removal of the tumor (143,144). The cause of the syrinx cavity result-
tionship to the spinal cord and neural foramina. These lesions are usu- ing from primary spinal cord lesions is less clear. The syrinx most likely
ally isointense with cord on both T1- and T2-weighted images. They results from altered CSF dynamics; however, in some patients, the syr-
uniformly enhance after contrast infusion (139–141). inx may be caused by fluid secreted into the spinal cord and central
Syringohydromyelia has been described in association with neu- canal by the tumor (145). If syringohydromyelia is seen in neurofibro-
rofibromatosis. The syrinx is almost always a secondary phenomenon, matosis (either NF1 or NF2) and no extramedullary mass can be seen,
resulting from either a primary tumor of the spinal cord or an intra- a thin section, contrast-enhanced MRI of the spinal cord should be
dural, extramedullary mass, such as a meningioma or neurofibroma, performed to rule out an intramedullary lesion. Syringohydromyelia is
that alters the CSF dynamics of the surrounding spinal subarachnoid discussed in more detail in Chapter 9.

Barkovich_Chap06.indd 592 5/6/2011 9:04:11 PM


Chapter 6 • The Phakomatoses 593

of affected patients have normal intelligence, only 75% have epilepsy,


OTHER FORMS OF and almost any organ of the body can be affected (169). As a result,
NEUROFIBROMATOSIS more sophisticated criteria for clinical diagnosis of the disorder have
Although the previous sections have made the distinction of NF1 from been established and are listed in Table 6-5 (170). Adenoma sebaceum
NF2 seem clear, the reality is that some patients have a syndrome that is the term used to describe a nodular rash of brownish red color that
appears to be an overlap between the two forms (89,90). For this rea- is disseminated over the face; the rash typically originates in the naso-
son, a third type, NF3, has been described that combines some of the labial folds and eventually spreads to cover the nose and the middle
features of types 1 and 2 (146,147). The genetics of this group is not
clear. Moreover, Riccardi has described several other, additional, forms
of neurofibromatosis (147). It now seems clear that the other forms of
the disease are merely variations of NF1, with different types or sites of TABLE 6-5 Diagnostic Criteria for
mutations or with limited expression due to somatic mosaicism. Tuberous Sclerosis Complex
(Revised 1998)
TUBEROUS SCLEROSIS COMPLEX
Major Features
Clinical Manifestations
Facial angiofibromas or forehead plaque
Tuberous sclerosis complex (TSC) is an autosomal dominant genetic
Nontraumatic ungual or periungual fibromas
disease that is characterized by the presence of tumor-like lesions
(hamartomas) in multiple organ systems. Two separate genes have Hypomelanotic macules (more than three)
been identified that are mutated or deleted in patients with tuberous Shagreen patch (connective tissue nevus)
sclerosis. The TSC1 gene is localized to chromosome 9q34 (148,149)
Multiple retinal nodular hamartomas
and codes for a protein called hamartin, while the TSC2 gene has
been localized to chromosome 16p13.3 (150,151) and codes for a Cortical tubera
protein called tuberin; mutations can be identified in 75% to 85% Subependymal nodule
of TSC patients (15,152–155). Hamartin and tuberin interact physi-
cally in vivo, a fact that clarifies how mutations in two different genes Subependymal giant cell astrocytoma
result in the common phenotype (156,157). These proteins form a Cardiac rhabdomyoma, single or multiple
heterodimer that functions as a tumor suppressor by inhibiting the Lymphangioleiomyomatosisb
mTOR kinase cascade. Increased activation of mTOR results in disor-
ganized cellular overgrowth and abnormal differentiation (155,158– Renal angiomyolipomab
161); mutation of either protein seems to lead to both increased Minor features
protein translation and cell size enlargement (162). TSC2 mutations
Multiple randomly distributed enamel pits in dental enamel
are about five times more common than mutations of TSC1 (155).
TSC1 mutations are slightly less common in sporadic TSC cases Hamartomatous rectal polypsc
(those with no family history, about two thirds of all patients) and Bone cystsd
are somewhat more common (15%–50%) in familial cases (15). It
Affected first-degree relative
is of interest that the TSC2 gene is located only in 48 base pairs of
DNA from the gene for adult-onset polycystic kidney disease (PKD1). Cerebral white matter radial migration linesa,d
When patients have a continuous deletion of both TSC2 and PKD1, Gingival fibromas
they have a particularly severe phenotype with very early onset of
PKD1 and early renal failure (163). Nonrenal hamartomac
Few differences in the clinical phenotype have yet been noted in Retinal achromic patch
patients with one mutation as compared with the other (164), although “Confetti” skin lesions
it appears that individuals with mutations of TSC1 seem to have less
risk of intellectual impairment, a lower frequency of seizures, fewer Multiple renal cystsc
subependymal nodules and cortical tubers, fewer retinal hamartomas, Definite TSC Either two major features, or one major plus two
milder renal disease, and less severe facial angiofibromas (154,165), minor features
whereas those with TSC2 mutations probably have a higher risk of
Probable TSC One major plus one minor feature
renal cysts (166). However, some TSC2 mutations have relatively mild
phenotypes, probably because the effect of the particular mutation on Possible TSC Either one major feature or two or more minor features
tuberin function is the ultimate factor in determining the severity of a
When cerebral cortical dysplasia and cerebral white matter migration tracts
the phenotype (161). Overall, about 75% of TSC mutations are spon- occur together, they should be counted as one rather than two features of
taneous (155); however, before concluding that an affected child is a new tuberous sclerosis.
mutation, it is necessary to examine both parents thoroughly, including b
When both lymphangioleiomyomatosis and renal angiomyolipomas are
Wood light examination, neuroimaging and, if possible, chromosomal present, other features of tuberous sclerosis should be present before a definite
analysis from multiple tissues including germ cells (167). A 2% to 3% diagnosis is assigned.
c
risk of recurrence in future pregnancies remains even if both parents Histological confirmation is suggested.
d
seem unaffected (167). Radiologic confirmation is sufficient.
Classically, TSC has been characterized by the clinical triad of TSC, tuberous sclerosis complex.
mental retardation, epilepsy, and characteristic skin lesions known From (170).
as adenoma sebaceum (168). Recent experience has shown that half

Barkovich_Chap06.indd 593 5/6/2011 9:04:13 PM


594 Pediatric Neuroimaging

FIG. 6-30. In utero MRI of a


fetus with tuberous sclerosis. A.
Coronal single shot half-Fourier
RARE image through the fetal
brain shows a cortical tuber (black
arrowheads) extending from the
right frontal cortex through the
cerebral mantle to the supero-
lateral surface of the right lateral
ventricle. A subependymal hama-
rtoma (black arrowhead) is also
present. B. Coronal image through
the parietal lobe and chest shows a
large cardiac rhabdomyoma (R) in
addition to a parietal tuber (black
arrow).

of the cheeks in the infraorbital region. Increasing awareness of the neuroimaging study, as making the diagnosis of tuberous sclerosis is
condition has resulted in a revision of the estimated incidence from important and influences treatment (164). Current recommendations
approximately 1 in 100,000 patients (168,171,172) to 1 in 6,000 live are for cranial imaging every 1 to 3 years in children (and every 3–6
births (164,173). No racial or sexual predilection has been detected. months if a giant cell tumor is present) and renal ultrasound every
The intracranial abnormalities of tuberous sclerosis are postulated to 1–3 years at all ages (because renal angiomyolipomas may enlarge at a
result from an abnormal expression of genes within the cells of the ger- concerning rate) (186,187).
minal matrices of the developing brain (174,175). As a consequence,
these stem cells do not differentiate, develop, or migrate properly (176).
Cutaneous Manifestations
The result is the presence of dysplastic, disorganized cells in the sub-
ependymal region, the cortex, and along curvilinear pathways between Although not radiologically apparent, cutaneous adenomata sebaceum
them (13,174,177). is a characteristic component of tuberous sclerosis and the imager
Seizures are evident in about 90% of affected individuals. Infantile should have a general knowledge of its clinical characteristics, which
spasms or myoclonic seizures that begin in infancy or early childhood were described in the previous section. These lesions, which are histo-
are the presenting symptom of tuberous sclerosis in approximately logically classified as angiofibromas, usually develop between age of 1
80% of patients; indeed, a significant number (10%) of infants present- and 5 years. Angiofibromas also occur in other areas of the body, most
ing with infantile spasms have tuberous sclerosis (178). The infantile commonly the trunk, gingiva, and periungual regions; in these regions
spasms evolve into other seizure types, most commonly symptomatic they develop later (typically after age 5) and may continue to develop
generalized epilepsy (∼60%), partial epilepsy (∼20%), or a mixture throughout life (188). Depigmented nevi in the form of oval areas with
of partial and generalized epilepsy (∼20%) (179). The seizures often irregular margins (ash-leaf spots) occur on the trunk and extremi-
decrease in frequency with increasing age (180), particularly when the ties and are as common as angiofibromas. Depigmented nevi appear
epilepsy is partial (181). Patients with TSC can have nearly any type of sooner than the adenoma sebaceum; indeed, they are often present at
seizure; therefore, the diagnosis of tuberous sclerosis should be con- birth and are frequently the cutaneous lesions that lead to a diagnosis
sidered in any child with epilepsy. The incidence of cognitive impair- of tuberous sclerosis in children with seizures (189). In light-skinned
ment is about 50% (182). Among the impaired patients, approximately children, the depigmented nevi may be demonstrable only under an
two thirds will be moderately to severely retarded, and one third only ultraviolet light. Café au lait spots are occasionally seen in patients with
mildly to moderately affected. Cognitive impairment in TSC is a mul- tuberous sclerosis, but their incidence is similar to that in the general
tifactorial condition; a link exists between cognitive disabilities and population; their presence in isolation should not suggest a diagnosis
lesion load (based on the proportion of brain volume occupied by of neurofibromatosis (190). Rarely, patients may develop lesions of the
tubers), age at seizure onset, and history of infantile spasms (183); scalp that induce hyperostosis of the underlying calvarium; histologi-
however, these factors explain only part of the intelligence quotient cally, these seem to be epidermal inclusion cysts. The other common
variability (155). cutaneous lesions of tuberous sclerosis, shagreen patches and subun-
Neuroimaging often plays an important role in making the diag- gual fibromas, do not usually appear until after puberty and are not
nosis of TSC (184,185); characteristic abnormalities are present on discussed here.
neuroimaging studies in more than 95% of affected patients (185).
These CNS abnormalities are present from before the time of birth
Ocular Manifestations
(Fig. 6-30), whereas the cutaneous malformations, such as adenomata
sebaceum, may not develop until much later in childhood. All children Ocular findings are common in tuberous sclerosis. The most common
who have infantile spasms without a known cause should have a of these is the retinal hamartoma, an astrocytic proliferation that is

Barkovich_Chap06.indd 594 5/6/2011 9:04:13 PM


Chapter 6 • The Phakomatoses 595

FIG. 6-31. Retinal hamartoma. A. Axial noncontrast CT image shows


a subretinal exudate with a focal nodule (arrow). B. Axial T1-weighted
image shows a nodule (arrow) in the right globe that is isointense to the
subretinal exudate. C. Postcontrast axial T1-weighted image shows that
the nodule enhances moderately.

seen on or near the optic disc in 15% of affected patients (168,171). differ histologically from the cortical hamartomas (tubers) and there-
Retinal hamartomas are usually present in both eyes and are often fore behave differently on imaging studies. The subependymal nodules
multiple (180). They may not be present at birth, developing over tend to be located along the ventricular surface of the caudate nucleus,
the first few months to years of life (191,192); the affected globe may most often on the lamina of the thalamostriate sulcus immediately
be microphthalmic (191) and leukocoria may be present, leading to posterior to the foramen of Monro (171). Less commonly, the nodules
presumptive diagnoses of persistent hyperplastic primary vitreous may be detected along the frontal and temporal horns and the lateral
(see Chapter 5) or retinoblastoma (see Chapter 7). They originate as ventricular bodies, the third ventricle or the fourth ventricle.
fairly flat, semitransparent, whitish lesions. Eventually, when they turn In neonates, subependymal hamartomas can be detected by trans-
whitish gray or yellow and become nodular, they are said to resemble fontanelle sonography, on which they appear as echogenic subependy-
a clump of mulberries (193). Retinal hamartomas are seen on CT as mal masses (Fig. 6-32A). They cannot be differentiated from germinal
nodular masses originating from the retina (Fig. 6-31); when hama- matrix hemorrhages or gray matter heterotopia by cranial sonography
rtomas calcify, they can be seen as small, calcified retinal masses that alone. The imaging appearance of subependymal hamartomas on CT
may be difficult to differentiate from a retinoblastoma (191,194). The and MR changes with the age of the patient. They are rarely calcified
presence of calcified subependymal nodules in the brain will help make in the first year of life; the number of calcifications typically increases
the differentiation. On MRI, retinal hamartomas appear as solid retinal with the age of the patient (184). Thus, they may be difficult to detect
nodules that show moderate uniform enhancement after administra- on CT scans of infants (Fig. 6-33C), but become progressively easier
tion of paramagnetic contrast (Fig. 6-31). A subretinal exudate may be to identify as they calcify (Fig. 6-34, also 6-37B). On MR scans, sub-
present, leading the ophthalmologist toward a presumptive diagnosis ependymal hamartomas appear as irregular subependymal nodules
of Coats disease (see Chapter 5) (195); again, the observation of sub- that protrude into the adjacent ventricle. Their appearance changes
ependymal nodules in the brain will lead to the proper diagnosis. The as the signal of the surrounding white matter changes (177,196). In
reader should be aware that, although they are most common in tuber- fetuses and infants, who have unmyelinated white matter, the hamar-
ous sclerosis (they are seen in over half of the cases), retinal hamar- tomas are relatively hyperintense on T1-weighted images and hypoin-
tomas are seen occasionally in the other phakomatoses as well (194). tense on T2-weighted images (Figs. 6-32 and 6-33) (197); in fetuses
and premature infants, these may be mistaken for subependymal
Intracranial Manifestations hemorrhages unless other lesions of tuberous sclerosis are identified.
As the brain myelinates, the subependymal nodules gradually become
Subependymal Hamartomas (Nodules) isointense with the white matter. They are most easily visualized on
Several characteristic lesions occur in the brain in TSC. The most T1-weighted images where they contrast with low signal intensity of
common of these are subependymal hamartomas (or nodules), which the CSF. Small nodules may not be apparent on T2-weighted images.

Barkovich_Chap06.indd 595 5/6/2011 9:04:14 PM


596 Pediatric Neuroimaging

FIG. 6-32. Tuberous sclerosis in a young infant. A. Coronal


transfontanelle sonogram shows hyperechogenic subependy-
mal hamartoma (small white arrow) and hyperechogenic
cortical tubers (large white arrows). B–D. Axial T1-weighted
images show hyperintense subependymal nodules (small solid
arrows), hyperintense cortical lesions (large open arrows), and
linear transmantle hyperintensities (small open arrows) that
extend from the ventricular surface to the cortex. The trans-
mantle hyperintensities will become invisible as the white
matter myelinates. E. Axial T2-weighted image shows the large
calcified left posterior frontal tuber (arrows). The linear trans-
mantle dysplasias are more difficult to identify on T2-weighted
images.

Barkovich_Chap06.indd 596 5/6/2011 9:04:15 PM


Chapter 6 • The Phakomatoses 597

FIG. 6-33. Evolution of tuberous sclerosis in an infant.


A. Axial T1-weighted image shows a hyperintense subependymal
nodule (arrowhead) along the body of the right lateral ventricle.
Multiple hyperintense lesions (arrows), many linear in configu-
ration, are evident in the deep cortical and subcortical regions.
B. Axial T2-weighted image at 13 days of age shows the right sub-
ependymal nodule (white arrowhead). Only a few of the cortical
and subcortical abnormalities evident in the T1-weighted image
(A) are appreciated; these are denoted by white arrows. C. Non-
contrast axial CT scan at 4 months of age shows multiple sub-
ependymal nodules (arrows), which are slightly hyperdense (but
not yet calcified) compared to cortex. D and E. Axial T2-weighted
image (D) and T1-weighted image with magnetization transfer
(E) at 18 months of age. The cortical tubers are T2 hyperintense;
many are associated with broadening/clubbing of the overlying
cortex (arrowheads, D). The MT image (E) demonstrates the
tubers and multiple hyperintense linear white matter lesions
(white arrows in E). Increased T2 signal in the periatrial white
matter (small arrows, D) is not secondary to dysplasia but sec-
ondary to poor myelination in this region, as these areas are
isointense to surrounding white matter on the corresponding
MT image (E). Magnetization transfer suppresses signal from
water molecules partially bound to myelin (see Chapter 2).

Barkovich_Chap06.indd 597 5/6/2011 9:04:17 PM


598 Pediatric Neuroimaging

Subependymal Giant Cell Astrocytomas


Subependymal giant cell astrocytoma (SEGA) is a term given to an
enlarging subependymal nodule that is usually situated near the fora-
men of Monro. Anatomically, these tumors differ from the subependy-
mal hamartomas by their size and their tendency to enlarge; their
characteristic location and tendency toward enlargement usually result
in a clinical presentation of hydrocephalus (fatigue, decreased appe-
tite, morning headache, visual field deficit, or behavioral problems)
(177,187,203). Their incidence in TSC is approximately 5% to 10%
(171,177,184,187). Histologically, subependymal lesions in patients
with tuberous sclerosis appear to span a continuum between sub-
ependymal hamartomas and giant cell tumors; although some lesions
are clearly in one category or the other, many lesions have histologic
characteristics of both (168).
On imaging studies, SEGAs are identified by the demonstration
of tumor growth on serial studies (Fig. 6-35) or by the development
of hydrocephalus associated with tumors near the foramen of Monro.
Although most are identified near the foramen of Monro (Figs. 6-35
and 6-36), SEGAs can occur anywhere along the ependymal surface.
Neither signal intensity nor the presence or absence of enhancement
FIG. 6-34. Axial noncontrast CT scan shows multiple calcified subependy- is useful in making the distinction between benign hamartomas and
mal hamartomas (white arrows) along the walls of the lateral ventricles. giant cell tumors (177); although it is clear that essentially all SEGAs
A hypodense right frontal tuber can also be identified (black arrow). enhance, many hamartomas enhance, as well, so enhancement is not a
useful criterion. The size criteria by which subependymal lesions should
be diagnosed as tumors rather than nodules are still being debated
Larger subependymal nodules manifest variably low signal intensity (187). Braffman et al. (177) have proposed a size criterion, suggest-
on the T2-weighted images, depending upon the extent of calcification ing that subependymal lesions greater than 12 mm diameter should be
(177,198,199). T2*-weighted gradient-echo or susceptibility-weighted classified as giant cell tumors. However a measurement alone may not
images are optimal for showing the calcification because of the mag- truly indicate the presence of a SEGA; for example, two adjoining sub-
netic susceptibility differences of calcium and brain. After intravenous ependymal nodules of 6 mm size can be mistaken for a 12-mm SEGA.
administration of paramagnetic contrast, subependymal nodules show Therefore, it seems that demonstration of growth on serial imaging
variable enhancement; some will enhance markedly, some mildly, and (Fig. 6-35) is a more reliable criterion than absolute size.
some not at all (177,200,201). The presence or absence of enhancement SEGAs tend to grow into the ventricle and uncommonly invade
has no clinical significance. Subependymal nodules have increased dif- brain parenchyma. Rarely, they degenerate into higher-grade, or infil-
fusivity and reduced fractional anisotropy compared to surrounding trating, neoplasms (203,204). Such degeneration should be suspected
white matter (202). radiologically if the tumor is seen to invade the brain parenchyma

FIG. 6-35. Progressive enlargement of SEGA. A. Postcontrast coronal T1-weighted image in a 4-year-old child with TSC shows an enhancing SEGA (white
arrow) in the region of the left foramen of Monro. The lateral and third ventricles are mildly enlarged, unrelated to the intraventricular mass. B. Coronal
contrast-enhanced T1-weighted image 3 years later reveals significant interval growth of the mass. The ventricles are not significantly changed.

Barkovich_Chap06.indd 598 5/6/2011 9:04:19 PM


Chapter 6 • The Phakomatoses 599

FIG. 6-36. Large SEGA in a patient with TSC. A. Coronal contrast-enhanced T1-weighted image shows a large, uniformly enhancing mass (long white arrow)
in the left frontal horn. A 6-mm enhancing subependymal nodule (short white arrow) is evident in the lower right frontal horn. B. Coronal T2-weighted
image demonstrates slight heterogeneity in the appearance of the left sided SEGA; despite benign histology, a mild amount of edema surrounds the anterior
commissure (long white arrow). The right-sided subependymal nodule (short white arrow) is slightly irregular in appearance.

(interstitial edema will typically be seen in the adjacent parenchyma) hardening by the overlying calvarium, making identification of cor-
or if rapid enlargement is seen. The treatment of benign giant cell tical tubers quite difficult unless they are calcified (Fig. 6-37) (168),
tumors is controversial. There is a trend toward neurosurgical resec- even with helical scanners. The MR appearance of cortical tubers also
tion of SEGAs prior to the development of symptomatic hydrocepha- changes with age. In fetuses and neonates, they appear as gyri that
lus (and to prevent possible degeneration into a higher-grade tumor), are hyperintense compared to the surrounding unmyelinated white
as patients who present with acute obstructive hydrocephalus have a matter on T1-weighted images and hypointense to white matter on
poorer surgical outcome (187). Few surgeons believe that shunting of T2-weighted images (Figs. 6-30, 6-32 and 6-33); about 20% of affected
the obstructed lateral ventricle is sufficient treatment. Rarely, giant cell gyri are enlarged (177,209,210). The T1 hyperintensity/T2 hypointen-
tumors occur in patients with no other evidence of TSC (117); these sity may extend through the cerebral mantle to the ventricle from the
tumors have a worse prognosis for disease-free survival than the giant tuber (see next section) (197,209,210). The appearance changes as the
cell tumors of TSC (205). It has not been determined whether these white matter myelinates and becomes isointense to the lesion. In older
neoplasms represent a forme fruste of TSC or a spontaneous tumor infants, tubers have a low intensity center on T1-weighted images and
unrelated to the genetic abnormality that causes TSC. high signal intensity on T2-weighted and FLAIR images (Fig. 6-38).
Those tubers with inherent T1 shortening that are hidden as myelina-
Cerebral Tubers (Hamartomas) tion ensues may be detected in older children and adults by the use
The cerebral tubers are the most characteristic lesions of tuberous scle- of magnetization transfer techniques: after suppression of the high
rosis pathologically. Macroscopically, these hamartomas are smooth, intensity of myelination by application of a magnetization transfer
whitish, slightly raised nodules that appear as enlarged, atypically pulse, parenchymal lesions may be more apparent than on standard
shaped gyri; they may be either round or polygonal. Histologically, they T1-weighted images (Fig. 6-38C) (211,212).
consist of bizarre giant cells, dense fibrillary gliosis, and diminished, T1 images with magnetization transfer and FLAIR images are
disordered myelin sheaths (35,171); balloon cells may be seen, making the most sensitive sequences in detection of parenchymal lesions in
these lesions histologically indistinguishable from focal cortical dyspla- children and adults with TSC (213,214). The efficacy of FLAIR in the
sia type IIb (see Chapter 5) (206). Any single patient may have as few as imaging of unmyelinated neonates and infants has not been dem-
one to two or have dozens (18). They are most commonly supratento- onstrated. Although it is not clear that any advantage is gained by
rial, although 15% of affected patients have cerebellar tubers (207). The identifying every parenchymal lesion instead of most of them, FLAIR
proportion that calcifies has not been reliably determined. The num- imaging makes cerebral parenchymal lesions of tuberous sclerosis
ber of calcified cortical lesions seen on CT increases with age; by age more conspicuous in myelinated brains and, therefore, makes more
10 years, calcified cortical tubers are present in up to 50% of patients cortical lesions visible (213). However, unless 3D volumetric FLAIR is
with tuberous sclerosis (184). The cortical calcifications may be gyri- acquired, artifacts around the foramina of Monro, secondary to CSF
form, simulating the appearance of Sturge-Weber disease on CT (208). flow, make subependymal nodules in that region difficult to identify.
In infants, cortical tubers can be seen on transfontanelle ultra- Whatever sequence is used, the tubers themselves seem predomi-
sound where they appear as foci of hyperechogenicity (Fig. 6-32A). nantly subcortical, separate from the overlying cortex (Figs. 6-33 and
Neonatal and infantile cortical hamartomas appear on CT as lucen- 6-38A). The inner (medial) margins of the tubers are poorly defined
cies within broadened cortical gyri. The lucency diminishes with age, on all imaging sequences. Although cortical tubers may become
making the noncalcified cortical tubers difficult to identify in older isointense with normal white matter on T1-weighted images, they
children and adults. In adults, this difficulty is exacerbated by beam become hyperintense on the T2-weighted images in the mature brain

Barkovich_Chap06.indd 599 5/6/2011 9:04:20 PM


600 Pediatric Neuroimaging

FIG. 6-37. Cortical tuber in TSC. A. Axial T2-weighted image of a 4-month-old baby with TSC shows focal cortical hypointensity and thickening
(short white arrows) along the left superior frontal sulcus. A faint line (long white arrow) extends toward the left frontal horn, typical of the transmantle
nature of tubers. B. CT scan in the same patient at age 13 years. The left frontal tuber is hyperdense, with a focal calcification (short white arrow) at the deep-
est part of the sulcus. A small calcification (long white arrow), probably a small subependymal hamartoma, is noted in the right temporal horn.

(Fig. 6-38A) (177,198–200). The signal characteristics probably result may appear bright on T1-weighted images, presumably because of
from the diminished myelin and dense astrogliosis within them. It is T1 shortening caused by the crystals of calcium (216). Degenerated,
important to realize that the high signal intensity on the T2-weighted calcified cortical tubers will sometimes enhance after contrast admin-
images does not imply malignant degeneration. In fact, neoplastic istration and display low T2 signal. On diffusion weighted studies,
degeneration of cortical tubers is extremely rare. Cyst-like cortical cortical tubers have increased diffusivity and decreased anisotropy
tubers (based on low signal appearance on FLAIR images [Fig. 6-38B]) compared to normal white matter (202). On MR perfusion imaging,
are seen more commonly in younger patients than in older children most (>90%) cortical tubers are hypoperfused relative to mean gray
and adults (214); patients with cyst-like tubers appear to have a more matter (Fig. 6-39A–C); presence of hyperperfused tubers is associated
aggressive seizure phenotype (215). When cortical tubers calcify they with increased seizure frequency (217).

FIG. 6-38. Cortical tubers in TSC. A. Axial FSE T2-weighted image shows multiple hyperintense cortical tubers (arrows). B. Axial FLAIR T2-weighted
image at the same level reveals lower signal in the mid left convexity tuber (arrow), consistent with a cystic (microcystic) change. C. Axial SE T1-weighted
image with magnetization transfer shows cortical tubers as regions of mixed hypointensity and hyperintensity. Suppressing the hyperintensity of the white
matter allows multiple white matter lesions (arrows) to be seen, some extending from the tubers to the lateral ventricular surface.

Barkovich_Chap06.indd 600 5/6/2011 9:04:21 PM


Chapter 6 • The Phakomatoses 601

FIG. 6-39. Perfusion, diffusion,


and susceptibility imaging in TSC.
A–C. Dynamic susceptibility-
weighted perfusion imaging of a
cortical tuber. Axial FLAIR image
(A) shows a cortical tuber (white
arrows) in the left superior fron-
tal gyrus. Ovals 1 and 2 in (B)
show the locations of the voxels
that were sampled during the
dynamic perfusion study. Graph
of signal intensity versus time
(C) from voxels 1 and 2 shows
normal blood volume (area
under the curve) of the normal
left superior frontal gyrus (2)
but reduced blood volume of the
tuber (1).

Barkovich_Chap06.indd 601 5/6/2011 9:04:22 PM


602 Pediatric Neuroimaging

FIG. 6-39. (Continued) D–G. Arterial


spin labeling, diffusion, and SWI of TSC.
Axial T2-weighted image (D) shows sub-
ependymal hamartomas (white arrows)
at the right foramen of Monro and left
trigone and a large cortical tuber (t) in
the left parietal lobe. Image from arte-
rial spin labeling perfusion (E) shows
reduced blood volume (hypointensity,
black arrow) within the left parietal
tuber. Average diffusivity map (F) shows
increased diffusivity (hyperintensity,
white arrow) in the same tuber. Sus-
ceptibility-weighted image (G) shows
markedly increased susceptibility (white
arrows) of the subependymal hamar-
tomas and reduced susceptibility (prob-
ably due to reduced blood supply, black
arrow) in the left parietal tuber. (Figures
D–G courtesy Dr. Erin Schwartz, Phila-
delphia.)

When cortical tubers are solitary and no other visceral or CNS When children with TSC have medically refractory seizures, some
manifestations of TSC are identified, the question will arise as to epileptologists now recommend surgical removal, as studies have sug-
whether the lesion is hamartomatous or neoplastic. In such cases, pro- gested a marked reduction in seizure frequency if most seizures can
ton MR spectroscopy or perfusion MRI may be useful. Proton spec- be localized to a single lesion (221,222). In such cases, it is critical to
tra of tubers show normal to slightly elevated choline and slightly correlate the epileptiform activity of the brain with a single structural
diminished NAA (218,219). The myo-inositol peak is increased on lesion if the surgery is to be successful. PET scanning, using alpha
short TE (20–30 msec) spectra. In contrast, most childhood neoplasms 11-C-methyl-L-tryptophan, has proven helpful in the identification of
have marked elevation of choline and marked diminution of NAA epileptic foci in TSC (223). In addition, epileptic tubers are associated
(see Chapter 7) (76,220). However, some low-grade neoplasms may with the largest volume of hypometabolism relative to tuber size (based
have spectra very similar to tubers. In these cases, perfusion imaging on FDG-PET coregistered to MRI) and the highest ADC values in the
may be helpful. Low-grade neoplasms have blood volume very simi- underlying white matter (224). BOLD imaging (221), magnetic source
lar to that of normal white matter, but tubers are quite hypovascular imaging (225), and videotelemetry (222) may be useful to establish
(Fig. 6-39) (217). this correlation. Of note, the medial temporal lobe should be carefully

Barkovich_Chap06.indd 602 5/6/2011 9:04:23 PM


Chapter 6 • The Phakomatoses 603

FIG. 6-40. Value of magneti-


zation transfer imaging in TSC.
A. Axial FLAIR image reveals
one definite hyperintense lesion
(arrow) in the left parietal lobe. B.
Corresponding axial T1-weighted
image with magnetization trans-
fer shows multiple cortical hype-
rintense lesions (arrows) through
outthecortexandsubcorticalwhite
matter, highly suggestive of TSC.

evaluated with MR imaging as mesial temporal sclerosis may occur in these regions appear as low attenuation, well-defined regions within
patients with TSC (226). the cerebral white matter that do not enhance after intravenous con-
trast infusion. When calcification occurs, it can involve all or part of the
White Matter Lesions lesion. Partially calcified nodules will have a mixed attenuation, with
Islets consisting of a grouping of neurons and glial cells are invariably one portion being of lower attenuation than the surrounding white
present in the white matter of patients with TSC (35,171). Microscopi- matter and the other portion of extremely high attenuation because
cally, they contain bizarre cells including giant neurons and balloon of the calcification (168,184). If the lesion is calcified, the calcification
cells; the latter are dysplastic cells that may have characteristics of both may appear as low or high signal intensity on T1-weighted images,
neurons and glia. These white matter foci also contain areas of hypo- depending upon the characteristics of the calcium crystals (216). Also,
myelination similar to those seen in the cortical tubers (35,171). Many as with cortical tubers, these white matter lesions have short T1 and
of these clusters of heterotopic cells are microscopic and, therefore, T2 relaxation times in fetuses, neonates and young infants (Figs. 6-32
do not appear on imaging studies; however, they are manifested as an and 6-33) (197,209,210). As with cortical tubers, enhancement is only
increase in the diffusivity and reduced anisotropy of normal-appear- seen if the lesion has degenerated; under these conditions, calcification
ing white matter in affected patients (227). Others will present as fine is usually present.
lines extending radially across the cerebral white matter. CT fails to
demonstrate these abnormalities. On MR, these white matter lesions Parenchymal Cysts
have the same signal characteristics as cortical tubers (Figs. 6-32, An unknown percentage of patients with TSC have cyst-like structures
6-33, 6-37, 6-38, and 6-40) and, if the proper imaging plane is used, in the cerebral hemispheric white matter (231); some reports suggest
are seen to course through the entire cerebral mantle from the cor- an incidence of about 10% to 15% (213,232). The histology of these
tex to the ventricular surface. They can be identified on T1-weighted lesions is not known, but their appearance is benign. The cysts are most
images in infants as subtle hyperintense regions (Fig. 6-32A); once the commonly periventricular, but may occur nearly anywhere (Fig. 6-41).
white matter becomes well myelinated, they may be difficult to see on Their clinical significance is uncertain (231). They may grow slowly,
T1-weighted sequences. On T1-weighted images with magnetization but none are known to exhibit malignant degeneration or to show
transfer pulses (to suppress the hyperintensity of normal white matter), enough mass effect to necessitate intervention.
FLAIR images, and T2-weighted images, they appear as well-defined
linear or curvilinear areas of high signal intensity (177,198–200,211); Cerebellar Lesions
indeed, T1-weighted images with magnetization transfer are the most Cerebellar lesions have been described in TSC but they are less com-
sensitive for detecting white matter lesions and show the most lesions in mon than cerebral pathology, occurring in about 15% of patients
the myelinated brain (Figs. 6-33E and 6-40) (213). In addition, magne- (177,184,233). An incidence of 25% has been reported in a recent study
tization transfer studies show diminished myelination in TSC, even in (234); in that study, patients with cerebellar lesions had overall higher
“normal-appearing” white matter (228). This observation is supported autistic symptomatology than patients without cerebellar lesions. The
by the discovery of increased mean diffusivity and reduced fractional posterior fossa lesions are histologically similar to those of the cere-
anisotropy in normal-appearing white matter of patients with tuber- bral hemispheres, consisting of cortical tubers, heterotopic clusters
ous sclerosis (229,230). in the white matter and, occasionally, subependymal hamartomas
Larger white matter lesions have a variable appearance that (35,171,177). The cerebellar hemispheric lesions are difficult to see by
depends upon the amount of calcification within them. These lesions CT unless they are calcified because of the beam-hardening artifact that
have histologic and imaging characteristic identical to cortical tubers occurs in the posterior fossa. The CT and MR appearance of these cere-
and to focal cortical dysplasia type IIb (206) (see Chapter 5). On CT, bellar lesions is otherwise quite similar to the appearance of those in the

Barkovich_Chap06.indd 603 5/6/2011 9:04:25 PM


604 Pediatric Neuroimaging

FIG. 6-41. Subcortical


cysts in tuberous sclero-
sis. A. Axial postcontrast
T1-weighted image shows
enhancing subependymal
nodules and hypointense
cortical tubers, diagnostic
of TSC. B. More cephalad
image in the same sequence
shows parenchymal cysts
(arrows).

cerebral hemispheres (Fig. 6-42), appearing mainly subcortical, mani- distal extremities in affected patients (235–237). The pathogenesis is
festing T2 hyperintensity, and undergoing degeneration/calcification poorly understood, but defects in the media due to deficiency and
over time (177,184). They are differentiated from neoplasia by the fact fragmentation of elastic fibers have been postulated (237). Involve-
that they do not enhance; and they undergo degeneration, causing ex ment of the cerebral vasculature is very rare, but aneurysms have been
vacuo enlargement of adjacent CSF spaces (Fig. 6-42). reported and seem to involve children disproportionately (238,239).
More than half of the aneurysms have been discovered in the internal
Vascular Lesions carotid arteries or in the anterior cerebral artery distribution, loca-
Vascular lesions are rare in TSC, although angiographic studies have tions that are uncommon in children (see Chapter 12). Therefore,
demonstrated aneurysms in the kidney, liver, aorta, iliac arteries, and it has been suggested that the cerebral aneurysms that have been
reported in TSC are in some way related to the disease, rather than
being incidental (238,239). Certainly, an aneurysm should be consid-
ered a likely possibility and a cerebral angiogram performed if a child
with tuberous sclerosis presents with subarachnoid or intraparenchy-
mal hemorrhage.

Non-CNS Manifestations
Although brain, ocular, and cutaneous lesions are the hallmarks of
TSC, lesions also occur in the heart, kidneys, liver, lung, and spleen
(168). Renal hamartomas occur in 40% to 80% of patients with tuber-
ous sclerosis (171,240). Histologically, these lesions are angiomyolipo-
mas; they often arise in young adulthood and enlarge slowly. They are
usually asymptomatic, although hematuria, flank pain, or a palpable
abdominal mass may develop. Malignant degeneration is rare. On
ultrasound, angiomyolipomas are well-defined and highly echogenic.
The CT and MR appearances are remarkable for the presence of fat.
Benign rhabdomyomas of the heart (Fig. 6-30) are less common than
the renal hamartomas but are important because they can present as
a congenital cardiomyopathy (171). These tumors are always suben-
docardial and may be either circumscribed or diffuse. Because of their
serious consequences, some authors feel that all patients with TSC
should have screening echocardiography.
FIG. 6-42. Cerebellar tubers. Axial T2-weighted image shows multiple Pulmonary involvement is the next most common type of visceral
hyperintense lesions (large white arrows) in the subcortical regions of the abnormality in patients with tuberous sclerosis (168). The characteristic
lateral cerebellar hemispheres. The more superficial cortex is hypointense pulmonary lesion is called lymphangioleiomyomatosis. In this condi-
(small white arrows), likely from mineralization. The overlying subarachnoid tion, the lung parenchyma undergoes cystic changes with a myoma-
spaces (arrowheads) are enlarged secondary to degeneration of the tubers. tous proliferation of tissue and chronic fibrosis occurring in the septa

Barkovich_Chap06.indd 604 5/6/2011 9:04:25 PM


Chapter 6 • The Phakomatoses 605

between the cysts. Other types of visceral lesions include adenomas Intracranial Pathology
and lipomyomas of the liver, adenomas of the pancreas, and tumors of
the spleen (168). Bone lesions consist of multiple dense lesions in the The major pathological abnormality in Sturge-Weber disease is a
cranium and cystic changes in the metacarpals and phalanges of the meningeal tangle of vessels, commonly referred to as an angioma,
hands (35,168,171). which is ordinarily confined to the pia mater. This pathologic process
consists of multiple capillaries and small venous channels that are mat-
ted together on the surface of the brain. The arteries are involved to
STURGE-WEBER DISEASE a lesser extent and tend to undergo fibrosis. The combination of the
facial and pial angiomas has been postulated to result from persistence
Clinical Manifestations
of the architectonic features of the primordial sinusoidal vascular
Sturge-Weber disease, also known as encephalotrigeminal angioma- channels that are present between the fourth and eighth week of gesta-
tosis or meningofacial angiomatosis, is an almost entirely sporadic tion (248,249). At that time the ectoderm that will form the skin of the
developmental disorder that is marked by angiomatosis involving upper part of the face overlies that part of the neural tube destined to
the face, the choroid of the eye, and the leptomeninges. The facial form the parietooccipital parts of the cerebrum. Normally, the superfi-
nevus flammeus, formerly referred to as an angioma or port-wine cial and deep portions of the vascular system become widely separated
stain, can involve part or all of the face. Ocular and facial involve- during the extensive subsequent growth of the cerebral hemispheres
ment without cerebral involvement (241) and ocular and cerebral (248). According to Alexander, the persistence of primordial vessels
involvement without facial involvement (242) have been described may explain why facial nevi in the area of the ophthalmic division
and are considered part of the syndrome. Other clinical compo- of the fifth nerve are associated with leptomeningeal angioma in the
nents of the syndrome are seizures, hemiparesis, hemianopsia, and occipital region, nevi in the region of the maxillary division are asso-
mental retardation. Although familial cases are reported, no clear ciated with angiomas in the parietal convexity, and nevi involving all
evidence of heredity has been discovered. Both sexes are equally three divisions are associated with angiomatosis of the entire convexity
affected (243). of the hemisphere, including the frontal lobe (243). The weakness in
The facial nevus is composed of a plethora of thin-walled vessels this theory lies in the inconsistent correlation between the location of
that most closely resemble capillaries. It is present at birth, usually uni- the facial nevus and the pial angioma. For example, frontal angiomas
lateral although occasionally bilateral, and involves the middle portion can be present in association with maxillary nevi. Another possibility
of the face with no predilection for either side. The nevus does not is that the superficial venous drainage of the brain never fully devel-
change with advancing years. More and more cases are being reported ops and that the dilated capillaries and small venous channels (and
of Sturge-Weber syndrome involving the brain without a facial nevus the nevus flammeus) are dilated in an attempt to compensate. Chronic
(244–246). venous hypertension might then cause the parenchymal injury.
Patients with the Sturge-Weber syndrome generally develop nor- Cortical calcifications are another pathological finding in Sturge-
mally until they begin to have focal or generalized seizures. Infantile Weber syndrome (Fig. 6-43E). The calcifications occur exclusively in
spasms develop in approximately 90% of affected patients during the areas of the brain subjacent to the angioma. They begin in the sub-
first year of life, followed by development of tonic, atonic, or myoclonic cortical white matter and later develop in the cortex, predominantly
seizures (243,247). With time, the seizures become progressively refrac- in cortical layers two and three (243). They are found most frequently
tory to medication. They are accompanied by an increasing hemipa- in the temporoparietooccipital region of the brain but can be located
resis in about 30% of patients, often accompanied by homonymous anywhere in the cerebrum (35,243,250). Cortical calcifications can be
hemianopsia. The majority of affected patients are mentally retarded bilateral in up to 20% of patients (250). Although their cause is not
(243,247). Prognosis is poor when both cerebral hemispheres or the confidently established, they are most likely caused by the chronic isch-
majority of a single hemisphere is affected (247). emia that results from impaired venous drainage (101).

FIG. 6-43. Child with Sturge-


Weber syndrome. A. Postcontrast
axial T1-weighted image at age 1
year shows diffuse leptomeningeal
enhancement and an enlarged right
choroid plexus (black arrowhead) on
the right; minimal leptomeningeal
enhancement (white arrows) is pres-
ent in the left frontal and occipital
poles. B. Axial T2-weighted image at
age 1 year shows mild enlargement of
ventricles and subarachnoid spaces.
Myelination of the right hemisphere
and left occipital pole (where the
most leptomeningeal enhancement
was seen) is accelerated. A large sep-
tal vein (white arrow) is present on
the left.

Barkovich_Chap06.indd 605 5/6/2011 9:04:27 PM


606 Pediatric Neuroimaging

FIG. 6-43. (Continued) C. Postcontrast axial T1 image at age 7 years shows interval progression of the bilateral leptomeningeal angiomas. Multiple abnor-
mal prominent subependymal/intraventricular veins are seen as foci of signal void (white arrows). D. Axial T2-weighted image at age 7 years shows mild
asymmetric volume loss on the right and prominent subependymal veins (white arrows). E. Axial contrast-enhanced CT at age 15 years shows extensive
cortical calcification (black arrowheads) with posterior predominance. Prominent enhancing deep medullary and subependymal veins are faintly seen
(white arrows).

Neuroimaging Manifestations gradient-echo images, which are more sensitive to the difference in
magnetic susceptibility between normal brain and calcium (Fig. 6-45)
Brain (256). With the more sensitive techniques, a thin ribbon of low signal
Contrast-enhanced MR studies are the most accurate single imaging intensity will be seen in or subjacent to the cerebral cortex in the affected
studies for showing the extent of the pial angioma (251,252); contrast- areas (256); these areas seem to correlate with reduced FDG uptake on
enhanced T2-weighted FLAIR images improve detection of leptom- PET studies (256). Contrast-enhanced T1-weighted sequences demon-
eningeal disease compared to postcontrast T1-weighted images (253). strate leptomeningeal abnormalities to a greater extent than SWI; SWI
Enhancement can sometimes be seen on CT if the patient is imaged appears superior to contrast-enhanced T1 sequences in identifying the
in infancy, prior to the appearance of cortical calcifications (250). enlarged transmedullary veins, the abnormal periventricular veins, the
However, calcifications mask the enhancement as determined by CT; cortical gyriform abnormalities, and the gray/white matter junction
moreover, MRI is more sensitive to the extent of enhancement than abnormalities (Fig. 6-45) (259).
CT (254). After intravenous administration of contrast, the angioma Dynamic MR perfusion studies reveal that the brain underlying
appears on MRI as an area of enhancement that seems to fill the suba- the enhancing pial angioma is hypoperfused. Cerebral hypoperfusion
rachnoid space, covering the surfaces of the gyri and filling the corti- is predominantly due to impaired venous drainage: in most cases, per-
cal sulci (Figs. 6-43 and 6-44). Dilated deep medullary veins, through fusion imaging reveals increased mean transit time with normal bolus
which venous blood is shunted from the superficial to the deep venous arrival; in the most severely affected regions reduced regional blood
system, can be seen in the white matter under the angioma (Fig. 6-44E) flow can be observed (260,261). Proton MR spectroscopy of affected
(255,256). As demonstration of the extent of the angioma is critical in brain regions shows elevated choline (possibly related to accelerated
determining the patient’s prognosis and the necessary extent of cortical myelination), reduced NAA (probably due to cortical/white matter
resection, contrast-enhanced MRI should be performed in all patients ischemic injury), and slightly elevated lactate (probably resulting from
for whom surgery for seizure control is planned (252). It is important ongoing ischemia). However, MRS abnormalities (NAA reductions,
to note, however, that “burned-out” cases of Sturge-Weber syndrome, elevation of choline) can be observed even in normal-appearing white
in which the affected hemisphere is markedly shrunken and heavily matter, distant from the leptomeningeal angiomatosis (262); similarly,
calcified, may not show enhancement (257). The reason for this lack of increased diffusivity has been documented in normal-appearing white
enhancement is not clear, but it may relate to thrombosis of the vessels matter with diffusion-weighted imaging (263).
comprising the angioma. The choroid plexus is frequently enlarged in patients with Sturge-
The effects of the pial “angioma” (and the associated restricted cor- Weber disease (264). In young children, the degree of plexal enlarge-
tical venous drainage) upon the underlying brain can also be assessed ment shows a positive correlation with the extent of the leptomeningeal
by imaging. As mentioned earlier, calcifications occur in the cerebral angioma; that is, the more extensive the parenchymal involvement, the
cortex underlying the pial angioma; they are the most frequent CT larger the choroid plexus (265). The enlarged choroid plexus is best
finding of Sturge-Weber disease (Fig. 6-43E) (250,258). Calcifications demonstrated on contrast-enhanced T1-weighted images as abnor-
most commonly occur unilaterally over the posterior portion of the mally prominent enhancement of the choroid may be seen ipsilateral to
hemisphere, although any portion of cerebral cortex may be involved. the angioma (Figs. 6-43 and 6-44) (264). On T2-weighted and FLAIR
The calcification is sometimes difficult to see on T1-weighted MRI images, the affected choroid plexuses are enlarged and are hyperintense
studies. It is more readily identified on T2-weighted images and, espe- to brain parenchyma (264). This phenomenon is most commonly a
cially, on susceptibility-weighted imaging (SWI) or on T2*-weighted result of hyperplasia of the choroid plexus, possibly related to increased

Barkovich_Chap06.indd 606 5/6/2011 9:04:28 PM


Chapter 6 • The Phakomatoses 607

FIG. 6-44. Infant with Sturge-Weber syndrome. A. Post-


contrast axial SE T1 image at age 5 weeks shows enlarged
left choroid plexus (white arrowhead), but only minimal
pial enhancement in the left occipital pole (white arrows).
B. Axial T2-weighted image at age 5 weeks shows minimal
hypointensity of the left parietal white matter. C–E. Same
patient at 2 years of age; interval development of cortical
atrophy and loss of underlying white matter. Postcon-
trast axial SE T1 image (C) shows increased conspicuity
of the left parietal-occipital leptomeningeal angioma;
the left choroid plexus (white arrowhead) is enlarged.
Axial T2-weighted images (D and E) show atrophy and
hypointense subcortical white matter signal in the left
posterior frontal, parietal, and occipital lobes; abnormal
dilated medullary (white arrows) and subependymal
(black arrowhead) veins are seen.

Barkovich_Chap06.indd 607 5/6/2011 9:04:29 PM


608 Pediatric Neuroimaging

Cranial asymmetry often results from cerebral hemiatrophy. The


ipsilateral calvarium is thickened and the ipsilateral paranasal sinuses
and mastoid air cells are enlarged because of the lack of brain growth
on the affected side. The thickened calvarium is seen on MRI as a wid-
ening of the fat-filled high-intensity diploic space. Midline structures
are often apparently displaced toward the side of the leptomeningeal
angioma. Occasionally, an enlarged hemicranium may be seen on the
side of the angioma. This paradoxical enlargement may be a result of
subdural hematomas accumulating in the affected hemicranium sec-
ondary to the cerebral atrophy (273).
Enlarged vessels are commonly seen on CT and MR studies in the
subependymal and periventricular region of the brain (Figs. 6-43D,
6-44E, and 6-45). Although venous malformations, arteriovenous
malformations, and dural arteriovenous fistulae may be seen in asso-
ciation with Sturge-Weber syndrome (274), the more likely cause for
these enlarged deep hemispheric vessels in most patients is enlarge-
ment of the vessels of the deep venous system of the brain (269,275).
When such enlargement occurs, it is a result of marked slowing of
blood flow in, or possibly thrombosis of, the maldeveloped superfi-
cial venous system of the brain. The pial angioma is associated with
the maldevelopment of, and diminished outflow through, the super-
ficial venous system. Therefore, venous blood is shunted through the
aforementioned deep medullary veins and into the deep venous system
of the brain (269,275). This redirected venous drainage should not be
FIG. 6-45. Child with Sturge-Weber syndrome and right hemispheric mistaken for arteriovenous malformations in which enlarged feeding
atrophy. Axial SWI shows the diffuse, hypointense cortical and subcorti- arteries, as well as large veins, are present. The absence of superficial
cal calcifications posteriorly throughout the atrophic right hemisphere. venous drainage in the region of the pial angioma was an important
Dilated, hypointense medullary and subependymal veins (white arrows) are criterion in the diagnosis of Sturge-Weber disease prior to the advent
evident adjacent to the ventricle. (This figure courtesy Dr. Erin Schwartz, of CT (269,275).
Philadelphia.) PET using 18F-FDG has been used to study cerebral metabolism
in children with Sturge-Weber syndrome. In the early stages of the
disease, the affected area is typically hypermetabolic. However, hypo-
venous flow from the affected cerebral hemisphere through the deep metabolism soon ensues (276). The authors have suggested that PET
venous system and the plexus. These are sometimes referred to as is useful in surgical planning when cortical resection is contemplated
“angiomatous malformations of the choroid” (249). for treatment of intractable seizures and failure to attain develop-
In the infant with Sturge-Weber syndrome, the white matter mental milestones (276). Fusion of PET with MRI is more useful still
underlying the angioma typically shows accentuated T2 shortening (255). The value of PET, as compared to contrast-enhanced MR, to
compared to the remainder of the brain (Fig. 6-44D and E) (266). This determine the extent of resection in affected patients has not been
is most likely secondary to abnormal hypermyelination; increased FA established.
and decreased diffusivity have been demonstrated in the involved white
matter using diffusion tensor imaging (267). The increased myelina- Eye Anomalies
tion may result from abnormal venous congestion or from repeated Abnormalities of the globe are seen in about 30% of patients with
seizures (268). Another possible contribution to the T2 shortening is Sturge-Weber disease (243,277). Pathologically, affected patients have
the presence of increased deoxyhemoglobin in capillaries and veins, angiomas of the choroid and sclera. They present with ocular pain or
the result of restricted superficial venous drainage, with consequent retro-orbital pain from glaucoma or with acute visual deterioration
shunting of deoxygenated blood through the dilated deep medullary from retinal detachment. If the glaucoma begins in utero, the globe can
veins into the deep venous system (269). enlarge as a result of the increased intraocular pressure, a condition
The ipsilateral cerebral hemisphere ultimately becomes atrophic known as buphthalmos. Buphthalmos is seen on CT and MR scans as
in most, but not all, patients with Sturge-Weber disease (Figs. 6-43 an enlarged, somewhat elongated globe. The choroidal angioma can
to 6-45); atrophy is uncommon at the time of seizure onset, however also be detected by MRI as thickening of the posterior wall of the globe
(Fig. 6-44). As might be expected, atrophy is bilateral in those patients on noncontrast T1-weighted images, by crescentic high intensity in the
with bilateral angiomas (270). The white matter subjacent to the posterior globe on the first echo of T2-weighted images, and as crescen-
affected cerebral cortex appears as low density on CT scans and shows tic enhancement in the posterior globe on contrast-enhanced MRI of
T1 hypointensity and T2/FLAIR hyperintensity on MR (271). Diffu- the orbit using a fat-suppression pulse (Fig. 6-46) (278). The presence
sion tensor imaging shows increased diffusivity and reduced fractional of choroidal angioma correlates with the presence of bilateral disease
anisotropy in the affected white matter (255). These imaging charac- and with the extent of facial involvement; it does not correlate with the
teristics most likely represent atrophy and astrogliosis in the ischemic size of the intracranial pial angioma (278). Care must be taken not to
underlying brain. misinterpret chemical shift artifact (seen as crescentic high intensity
Infratentorial involvement (presenting as leptomeningeal enhance- along the posterior margin of the globe on T2-weighted images using
ment, atrophy, and developmental venous anomaly) may be more fre- a short bandwidth) as a choroidal angioma; because of this potential
quent than previously thought and has been reported in 10% to 40% pitfall, use of a fat-suppressed, contrast-enhanced sequence is most
of cases (272). reliable.

Barkovich_Chap06.indd 608 5/6/2011 9:04:30 PM


Chapter 6 • The Phakomatoses 609

cerebellar complaints, but this order of events is by no means constant;


moreover, the ocular findings are nonspecific and often do not estab-
lish a definite diagnosis. The usual sequence of events is reactive reti-
nal inflammation with exudate and hemorrhage, followed by retinal
detachment, glaucoma, cataract, and uveitis. By the time the patient
is seen because of decreasing visual acuity or eye pain, the secondary
changes of retinal detachment may mask the underlying lesion. Head-
ache, vertigo, and vomiting result from the cerebellar tumor and asso-
ciated increased intracranial pressure, when present. Other cerebellar
findings such as dysdiadochokinesis, dysmetria, and Romberg sign are
common (289). Uncommonly, patients may have complaints of spinal
cord dysfunction such as loss of sensation or impaired proprioception.
It is rare, however, for patients to come for medical attention because
of symptoms of spinal cord or visceral lesions in this disease (286).
Rarely, patients present with hearing loss resulting from tumors of the
FIG. 6-46. Angioma of the choroid, infant with Sturge-Weber syndrome. endolymphatic sac (290).
Postcontrast axial T1 image shows marked enhancement (white arrows) of
the choroid of the right ocular globe, indicating the presence of a choroidal
angioma. Ocular Manifestations
Angiomas of the retina are observed in over half of affected patients;
symptoms from these lesions are usually the earliest manifestation of
the disease (286). Retinal angiomas are best demonstrated clinically,
Extra-CNS Manifestations using indirect ophthalmoscopy and fluorescein angiography. The
Patients with Sturge-Weber syndrome do not typically have abnormal- angiomas are multiple in up to two thirds of patients and bilateral in
ities of the thoracic or abdominal viscera. However, some patients will up to 50% (286,289,291). Although CT and MR can detect the sec-
have angiomas of the viscera and extremities in conjunction with typi- ondary retinal detachment, the retinal angiomas themselves are usually
cal findings of Sturge-Weber syndrome (243,279,280). The angiomas quite small and are rarely detected by imaging studies.
can be localized or diffuse; they can be located in the intestine, kidneys,
spleen, ovaries, thyroid gland, pancreas, or lungs (243). This combina- Brain and Spinal Cord Manifestations
tion of Sturge-Weber syndrome with involvement of the viscera and
extremities is called Klippel-Trenaunay-Weber syndrome by some Cerebellar hemangioblastomas are present in more than half of patients
authors (281), while others consider the entire complex to be a part with von Hippel-Lindau disease (286,291). They are the second most
of the Sturge-Weber syndrome (279), and still others consider it an frequent source of initial symptoms and frequently recur after surgical
overlap syndrome (282). resection. Hemangioblastomas may develop in childhood before the
age of 10 years, in the teen years, or in early adulthood (291). The most
common locations are the spinal cord (50%), cerebellum (38%), brain-
VON HIPPEL-LINDAU DISEASE stem (10%), and cerebrum (2%) (Figs. 6-47 to 6-49) (292). Between
20% and 40% of the tumors are solid (35). If untreated, many solid
Clinical Manifestations
tumors will develop cysts and enlarge, but many tumors will remain
Also known as CNS angiomatosis, von Hippel-Lindau disease is an auto- stable in size or grow slowly unless cysts develop (293). When grow-
somal dominant disorder with incomplete penetrance characterized by ing, tumors exhibit a stuttering growth pattern and frequently remain
retinal angiomas; cerebellar and spinal cord hemangioblastomas; renal asymptomatic; thus, surgery based on radiologic progression alone is
cell carcinomas; endolymphatic sac tumors; pheochromocytomas; pap- often not indicated (294). Tumors associated with symptoms tend to
illary cystadenoma of the epididymis; angiomas of the liver and kidney; be larger and are more likely to be associated with peritumoral edema
and cysts of the pancreas, kidney, liver and epididymis (283–285). von or peritumoral cysts than asymptomatic tumors (295).
Hippel-Lindau disease affects approximately 1 in 40,000 individuals; When the nodules of hemangioblastoma are large enough, they
both sexes are affected equally (286–288). The diagnosis is established will show contrast enhancement on CT; however, when the nodules
if patients have more than one hemangioblastoma of the CNS, one are small, the tumor may be indistinguishable from a benign posterior
hemangioblastoma with a visceral manifestation of the disease, or one fossa cyst or, in the absence of a cyst, they may not be detected at all
manifestation of the disease and a known family history (286). The (250,296). The MR appearance of the cystic lesions is that of sharply
disease is caused by a defective tumor suppressor gene, called VHL, at marginated, peripheral cerebellar masses showing T1 hypointensity
chromosome 3p25-p26 (287); its gene product, pVHL, is involved in and T2/FLAIR hyperintensity (Figs. 6-47 and 6-48) (297,298). Precon-
cell cycle regulation and angiogenesis. Inactivation of pVHL results in trast and postcontrast T1-weighted sequences are the most useful for
overexpression of hypoxia inducible mRNAs, including VGEF (vascu- detection of hemangioblastomas (299). If a solid nodule can be seen
lar endothelial growth factor) (288). von Hippel-Lindau can be diag- within the wall of the cyst, it will often have small curvilinear areas of
nosed by molecular biologic techniques, with potential for diagnosis signal void within it, representing enlarged feeding and draining vessels
in utero. (300). Solid hemangioblastomas appear as ill-defined, solid masses that
The diagnosis of von Hippel-Lindau disease is usually established can be difficult to identify without administering paramagnetic con-
after identification of either the cerebellar or retinal tumors. Symptoms trast (Fig. 6-50) (297,298). In general, they show T1 hypointensity and
typically begin during the third or fourth decade of life; it is unusual T2/FLAIR hyperintensity, although hemorrhage is occasionally present,
for patients to present before the second half of the second decade resulting in high signal intensity on noncontrast T1-weighted images.
(283,286,289). Symptoms from the retinal angiomas typically antedate The use of MR contrast agents increases sensitivity for detection of

Barkovich_Chap06.indd 609 5/6/2011 9:04:31 PM


610 Pediatric Neuroimaging

FIG. 6-47. Solid and cystic cerebellar hemangioblastomas in von Hippel-Lindau disease. Postcontrast sagittal (A) and coronal (B) T1 images show a large
cerebellar cyst with an enhancing mural nodule in the superior-most vermis (long black arrows). A second small nodule (short black arrows) is seen in the
left cerebellar tonsil.

small lesions; solid portions of tumor enhance intensely (Figs. 6-47 autopsy data suggests that these lesions may be more common than was
to 6-50) (297,299). Angiography may still be performed in order to previously suspected (35,286,289). Indeed, MRI demonstrates a con-
show the location and size of the arteries supplying the tumor. The siderably larger number and higher incidence of spinal cord hemangio-
angiographic appearance of the hemangioblastoma is highly charac- blastomas than were previously believed to exist (Figs. 6-48 and 6-49)
teristic, showing tangles of tightly packed, wide vessels that become (298); the spinal cord is the most common location for hemangioblas-
opacified in the early arterial phase. A nonvascular (cystic) portion of tomas in this disease (292). Syringomyelia is present in the majority
the mass is often seen (301). These angiographic findings can also add of patients with spinal cord hemangioblastomas; symptoms are most
diagnostic specificity to a lesion that is detected on CT or MRI. often due to the cyst and not the solid portion of the tumor. Differen-
In the past, hemangioblastomas of the spinal cord were thought tiation of syringomyelia from tumor cysts and from cord edema can be
to be uncommon because of the rare clinical manifestations of spinal difficult, but T2-weighted images can often help differentiate cyst from
cord pathology in patients with von Hippel-Lindau disease. However, edema, as the cysts are typically of higher signal intensity and have

FIG. 6-48. Cerebellar and spinal cord hemangioblastomas in von Hippel-Lindau syndrome. A. Axial T1-weighted image shows a right cerebellar cyst (large
black arrows) with a mural nodule (small solid black arrows) containing a curvilinear signal void (small open white arrow). A second nodule (small solid white
arrows) with curvilinear signal voids is seen in the anterior left cerebellar hemisphere. B. Postcontrast image shows enhancement of the solid portions of
both hemangioblastomas. C. Sagittal T2-weighted image shows multiple curvilinear flow voids in the subarachnoid spaces and a mass (white arrows) at L-4.
Multiple other hemangioblastomas are difficult to see. D. Sagittal postcontrast T1-weighted image of the spine shows the large L-4 mass (white arrows) with
some internal flow voids (black arrowheads) and innumerable other enhancing hemangioblastomas (white arrowheads).

Barkovich_Chap06.indd 610 5/6/2011 9:04:31 PM


Chapter 6 • The Phakomatoses 611

FIG. 6-49. Medullary, cervicomedullary, and spinal cord hemangioblastomas in von Hippel-Lindau syndrome. A. Sagittal T1-weighted image through
the upper spinal cord after IV contrast infusion. An enhancing mass (black arrow) with surrounding cyst/edema (white arrowheads) is seen at the cervical-
medullary junction with exophytic extension into the cisterna magna. A second lesion (white arrow) is present at the T2 level dorsally. Both lesions were
hemangioblastomas. The more caudal lesion was not clinically suspected. B. Sagittal T2-weighted image shows the cervicomedullary mass (large white
arrows) as hypointense, a flow void (black arrowhead) adjacent to the mass, a tumor cyst (white arrowheads), and edema of the adjacent spinal cord (small
white arrows). C. Axial postcontrast T1-weighted image at the upper medullary level shows an enhancing hemangioblastoma (white arrows) in the left
inferior cerebellar peduncle.

sharp margins, whereas edema is less hyperintense and has indistinct hemangioblastomas is the enlarged feeding and draining vessels that
margins (Fig. 6-49). It is difficult to differentiate idiopathic syringomy- are manifest as serpiginous areas of signal void within and adjacent to
elia from syringomyelia secondary to hemangioblastomas without the the tumor (297,300).
use of an MR contrast agent. Following intravenous administration of Hemangioblastomas may also occur within the brainstem
paramagnetic contrast, the tumor enhances markedly, facilitating the (Fig. 6-49) or the cerebral hemispheres (250). The radiologic appear-
diagnosis (297,302). Another specific MR characteristic of spinal cord ance of these lesions is identical to that of the cerebellar or spinal cord

FIG. 6-50. Recurrent hemangioblastomas


in von Hippel-Lindau syndrome. A. Axial
noncontrast T1-weighted image shows
hypointensity (arrows) from prior surgery,
but no evidence of recurrent tumor. B. Post-
contrast scan shows multiple enhancing cer-
ebellar tumors.

Barkovich_Chap06.indd 611 5/6/2011 9:04:33 PM


612 Pediatric Neuroimaging

hemangioblastomas. They may be cystic with a solid nodule or entirely disease should prompt neuroimaging directed at the endolymphatic
solid; the solid portion contains curvilinear signal voids and shows sac. Conversely, the discovery of an endolymphatic sac tumor in a
marked contrast enhancement. The diagnosis is obviously facilitated patient under the age of 30 years should incite a search for the presence
by knowledge of the diagnosis of von Hippel-Lindau syndrome or the of ocular, cerebral, cerebellar, or renal masses. Patients who develop
presence of other hemangioblastomas. bilateral papillary angiomatous tumors of the endolymphatic sac may
be presumed to have von Hippel-Lindau disease (304).
The imaging findings of papillary cystadenomas of the endolym-
Papillary Cystadenomas of the
phatic sac are rather characteristic. The tumors originate from the
Endolymphatic Sac endolymphatic epithelium within the vestibular aqueduct, along the
Another tumor that has a high incidence in von Hippel-Lindau dis- posteromedial margin of the petrous pyramid (305). CT shows bone
ease is the papillary cystadenoma of the endolymphatic sac (303,304). destruction with scattered calcifications centered at the level of the
Although most papillary endolymphatic sac tumors appear to occur vestibular aqueduct (Fig. 6-51A). T1-weighted MR images show a het-
sporadically, patients with von Hippel-Lindau disease are at greater risk erogeneous mass that is predominantly isointense to brain with areas
for developing these tumors than the general population; the incidence of high signal intensity. T2-weighted images show a predominantly
of papillary cystadenomas of the endolymphatic sac in von Hippel- hyperintense mass with areas of low signal intensity (Fig. 6-51B and C).
Lindau disease is about 7% to 15% (290,305). Patients with endolym- Larger tumors show vascular signal voids. The tumors enhance homo-
phatic sac tumors typically present with hearing loss; therefore, the geneously or heterogeneously after administration of paramagnetic
finding of hearing loss in a patient with known von Hippel-Lindau contrast (Fig. 6-51D and E) (290,306). Angiography shows vascular

FIG. 6-51. Papillary cystadenoma of the endolymphatic


sac in von Hippel-Lindau syndrome. A. Axial noncontrast
CT scan shows bone destruction (white arrows) of the
inner ear and mastoid air cells. B. Axial 3D T2-weighted
image shows a hyperintense mass (white arrows) near the
vestibular aqueduct orifice and growing into the mastoid
air cells. C. Axial T1-weighted image shows the hetero-
geneous mass (white arrows), demonstrating peripheral
hyperintensity and central hypointensity. D and E. Axial
and coronal postcontrast T1-weighted images show that
the mass (white arrows) becomes relatively homogeneous
after administration of contrast due to enhancement of the
relatively hypointense center.

Barkovich_Chap06.indd 612 5/6/2011 9:04:35 PM


Chapter 6 • The Phakomatoses 613

tumors with supply primarily from external carotid branches, although Characteristic skin changes (mucocutaneous telangiectasias) begin
some supply from internal carotid and vertebrobasilar branches may to appear between 3 and 6 years of age. They appear first on the bulbar
be found in larger tumors (290,306). conjunctiva and subsequently on the ears, face, neck, palate, dorsum
of the hands, and antecubital and popliteal fossae. Other associated
Visceral Manifestations dermatologic changes can be seen as well (308,309). Recurrent bacte-
rial and viral sinopulmonary infections occur in most patients, lead-
The major visceral manifestation of von Hippel-Lindau disease is renal ing to bronchiectasis and pulmonary failure, the most common causes
cell carcinoma, which occurs in up to 40% of patients. These tumors of death. Malignancies develop in 10% to 15% of affected patients
are frequently multicentric or bilateral (289). They occur later in life (314). Lymphomas and leukemia predominate in younger patients,
than most of the neurologic manifestations, with the average presenta- whereas epithelial malignancies occur much more frequently in adults
tion during the fifth decade of life (286). Pheochromocytomas may be (314,315).
present in 10% to 15% of patients. Cysts occur in virtually all visceral
organs, including the liver, omentum, mesentery, spleen, adrenal gland,
and epididymis. Adenomas can occur in the liver and epididymis Pathological and Neuroimaging
(286,296). Thus, affected patients must be evaluated with abdominal Manifestations
and pelvic imaging studies as well as neural axis imaging.
The major neuropathological finding in ataxia-telangiectasia is degen-
eration of the cerebellar cortex. Purkinje cell and granule cell degrada-
ATAXIA-TELANGIECTASIA tion can be found in the vermis, the hemispheres, or both. The cause
of this neuronal degeneration and atrophy has not been fully discov-
Clinical Manifestations
ered; however, some authors have found ATM-mediated damage in the
Ataxia-telangiectasia is an autosomal recessive disorder characterized nuclei of the Purkinje and granule cells, suggesting that DNA damage
by a complex set of clinical features that include cerebellar degenera- resulting from the abnormalities in this protein are the cause (316).
tion, dilation of blood vessels, severe immune deficiencies, premature Additional CNS findings include atrophy of the cerebellar nuclei, ante-
aging, predisposition to cancer, and acute radiation sensitivity (307– rior horn cell atrophy and demyelination of the gracile fascicule in the
311). The incidence is approximately 1 per 40,000 live births (307). spinal cord, and the existence of nucleocytomegalic cells in the anterior
The gene (called ATM for ataxia-telangiectasia, mutated) has been pituitary gland (317). In addition, spongy degeneration of the cerebral
identified and is located on chromosome 11q22–23 (312). The gene cortex has been described surrounding degenerated cortical blood ves-
encodes a nuclear protein kinase that is needed for detecting DNA sels in adults with this disorder (317).
damage or for blocking cell growth until the damage is repaired; cells The major abnormalities seen on CT and MRI are cerebellar corti-
in affected patients have frequent chromosomal aberrations including cal atrophy, manifested as diminished cerebellar size, dilation of the
chromosomal breaks, rearrangements, and aneuploidy (311). Affected fourth ventricle, and increased cerebellar folial prominence (318).
patients usually present with cerebellar ataxia, which manifests itself Initially, the atrophy involves the vermis and the lateral aspect of the
when the child starts to walk. The ataxia is followed by a progressive cerebellar hemispheres, with more severe and extensive atrophy (and
neurological deterioration. Eventually, the children are confined to a more severe ataxia) developing with longer duration of the disease
wheelchair and exhibit oculomotor abnormalities, dysarthric speech, (Fig. 6-52) (319). Hemorrhage may occur as a result of rupture of
choreoathetosis, endocrine abnormalities, and myoclonic jerks parenchymal telangiectasias. Cerebral infarcts may result from emboli
(307,308,313). that are shunted through vascular malformations within the lungs.

FIG. 6-52. Ataxia telangiectasia in a 7-year-old child. A. Sagittal T1-weighted image shows mild to moderate atrophy of the cerebellar vermis, more severe
superiorly. B. Coronal FLAIR image reveals mild atrophy of the superior and lateral cerebellar hemispheres.

Barkovich_Chap06.indd 613 5/6/2011 9:04:36 PM


614 Pediatric Neuroimaging

Older patients may have multiple foci of hypointensity in the cerebral With few exceptions, individuals with NCM have congenital
white matter on T2*-weighted images, presumably secondary to multi- pigmented nevi on the head and neck or dorsal spine area, commonly
ple small hemorrhages from the abnormal blood vessels (313,320,321). with presence of multiple satellite nevi (334). Two thirds of affected
Proton MR spectroscopy acquired in the cerebellum with TE = 144 patients have a giant congenital melanocytic nevus, usually of the lum-
msec shows reduction of all metabolites with reduced NAA/Cho and bosacral region, while one third have numerous small melanocytic nevi
increased Ch/Cr ratios (vermis > hemispheres) compared with age- without a single large lesion (335). Symptomatic CNS disease typically
matched controls (321,322). develops at less than 2 years of age, while a smaller, second peak of onset
When cerebellar atrophy is an isolated finding in a young child of symptoms occurs at puberty or during adult life. The most common
with cerebellar ataxia and progressive neurologic deterioration, the neurological complications include seizures, cranial nerve dysfunction,
possible diagnosis of ataxia-telangiectasia should be raised. For a dis- and signs and symptoms of increased intracranial pressure. Hydro-
cussion of other causes of cerebellar atrophy in childhood, see the last cephalus is seen in two thirds of cases and is thought to result from
portion of Chapter 3. extensive leptomeningeal infiltration with melanosis or with develop-
ment of intracranial melanoma (331,336). Spinal involvement occurs in
approximately 20% of cases, and may result in myelopathy, radiculopa-
Nijmegen Breakage Syndrome
thy, and bowel or bladder dysfunction (335). The prognosis of symp-
The Nijmegen breakage syndrome (NBS) is a rare, autosomal recessive tomatic NCM is poor; the majority of patients die within 3 years of their
genetic syndrome that is classified as an ataxia-telangiectasia–related initial neurologic symptoms, either from CNS malignant melanoma or
disorder; thus it is included in this section. However, NBS appears to be from progressive growth of “benign” melanocytic cells (332).
a distinct disorder, both clinically and genetically (323), resulting from An asymptomatic form of NCM was originally suggested by the
mutations of the NBS1 gene at chromosome 8q21 (324). The protein finding of leptomeningeal melanocytosis on postmortem examination
product of the gene, called nibrin, is involved in repair of DNA (325,326). of two patients. Frieden et al. (337,338) recently documented a surpris-
The disease appears to be prevalent in the central European population, ingly high incidence of MR abnormalities in asymptomatic children
in whom the presence of a common founder mutation in NBS1 has been with giant cutaneous melanocytic nevi. Of 43 children with giant cuta-
found (326). Affected children present with microcephaly (OFC below neous melanotic nevi over their head and neck or spine, 23% (10/43)
third percentile at birth, with progressively declining percentile), distinct had focal areas of T1 shortening strongly suggestive of melanotic
facies (sloping forehead, receding mandible, prominent midface and deposits in one or multiple areas of the brain, particularly the temporal
nose, upward slanting of the palpebral fissures), and early growth retar- lobes (amygdala), cerebellum, pons, and/or medulla (337). Additional
dation (327,328). Intelligence is initially normal to borderline low, but findings included intracranial mesenchymal abnormalities (including
most progress to moderate intellectual disability by adolescence (329). arachnoid cyst), cerebellar hypoplasia, spinal lipomas, tethered spinal
Immunodeficiency is present, leading to recurrent infections and a pre- cord, and Chiari type I malformation. Despite the frequent finding of
disposition to cancer, predominantly lymphoma, at a young age (329). melanotic deposits, no patient went on to develop CNS melanoma,
Because of the DNA repair defects, these children are particularly sensi- with an average duration of 5 years follow-up (338). This is in striking
tive to ionizing radiation; exposure to x-rays should be avoided. contrast to the usual outcome of symptomatic NCM, where death usu-
Imaging findings seem to be composed of hypogenesis of the cor- ally occurs within 2 to 3 years. This result suggests that the brain MR
pus callosum (seen in ∼50%) and consequent diminished white matter findings in asymptomatic individuals may be more analogous to those
around the trigones and occipital horns of the lateral ventricles (329). in the skin. If so, affected patients would have a somewhat increased
Sinus disease is reportedly common and sphenoid sinus pneumatiza- risk of both cutaneous and primary CNS melanoma over their lifetime,
tion delayed (329). Searches should be made for opportunistic infec- but not necessarily an immediate dismal prognosis or even certainty of
tions and tumors, particularly lymphoma and leukemia. neurologic symptoms (338).
In 1972, Fox proposed the following criteria for diagnosing NCM:
(a) unduly large or unusually numerous pigmented nevi in association
NEUROCUTANEOUS MELANOSIS with melanosis or melanoma of the pia-arachnoid, (b) no evidence of
malignant change in any of the cutaneous lesions, and (c) no evidence
Clinical Manifestations
of malignant melanoma in any organ apart from the meninges (332).
Neurocutaneous melanosis (NCM) was first described by Rokitan- A revision of Fox’s criteria has been proposed in order to more accu-
sky, who reported about a 14-year-old girl with a giant congenital rately define the population at risk (335,339): (a) large or multiple (3
melanocytic nevus and mental retardation who developed late-onset or more) congenital nevi in association with meningeal melanosis or
hydrocephalus (330). Since that initial report, over 100 cases have been CNS melanoma, where large is defined as a diameter equal to or greater
reported (331). Most cases are sporadic. Giant congenital nevi are than 20 cm in an adult, 9 cm on the scalp of an infant, or 6 cm on
themselves a relatively rare birthmark, occurring in approximately 1 the body of an infant; (b) no evidence of cutaneous melanoma, except
in 20,000 to 50,000 live births (332). When these birthmarks are asso- in patients in whom the examined areas of the meningeal lesions are
ciated with melanosis of the leptomeninges or brain, a diagnosis of histologically benign; and (c) no evidence of meningeal melanoma,
NCM can be made. A deregulation of hepatocyte growth factor/scatter except in patients in whom examined areas of the skin are histologi-
factor (HGF/SF) signaling has been implicated in the pathogenesis of cally benign. Those cases with histologic confirmation are considered
NCM. HGF/SF is a cytokine that stimulates the proliferation, migra- definite; all others are considered provisional diagnoses.
tion, and morphogenesis of different types of cultured epithelial cells
(333). It plays an important role in the normal distribution and pro-
Pathological Manifestations
liferation of melanocytes, as transgenic mice overexpressing HGF/SF
have been found to develop multiple pigmented nevi in both the skin Pathologists may find a substantial number of benign melanotic cells
and leptomeninges (333). Moreover, abnormal expression of an HGF/ within the basilar meninges of normal patients at autopsy, as both mel-
SF receptor called Met has been immunohistochemically detected in anocytes and the basilar pia-arachnoid derive from neural crest (340–
the cutaneous nevus of an infant with NCM (333). 342). Patients with NCM, however, have orders of magnitude of more

Barkovich_Chap06.indd 614 5/6/2011 9:04:37 PM


Chapter 6 • The Phakomatoses 615

melanotic cells, which may be distributed diffusely or demonstrate show only subtle hyperdensity and are difficult to see unless they have
nodularity within the meninges (332,339). A number of potential converted into a melanoma. Leptomeningeal involvement is difficult to
explanations for the abnormal accumulation of melanotic cells in the appreciate unless malignant degeneration has occurred, in which case
meninges have been proposed, including abnormal migration of mel- contrast-enhanced images may show leptomeningeal enhancement.
anocyte precursors (343), abnormal expression of melanin-producing MRI shows foci of T1 hyperintensity, and sometimes T2 hypointen-
genes within the leptomeningeal cells (344), or rapid proliferation of sity, in the brain parenchyma or meninges (Figs. 6-53 and 6-54), sig-
“normal” melanin-producing leptomeningeal cells. The abnormal mel- nal characteristics that are compatible with the T1 and T2 shortening
anin-producing cells are often present in the perivascular spaces along effects of melanin. These deposits of melanin are more easily detected
vessels that penetrate the brain from the basilar cisterns and result in in the unmyelinated brain, as the T1 and T2 shortening caused by
parenchymal melanin deposits (339,345). white matter myelination makes the melanin less conspicuous (338).
The anterior temporal lobes and cerebellum appear to be the Thus, MR screening for melanotic deposits is best performed early in
most common locations for accumulation of melanotic cells in NCM infancy. FLAIR images show both parenchymal and leptomeningeal
(339,346–351). In the anterior temporal lobe, the amygdala seems par- melanosis as hyperintense compared to normal parenchyma (353). The
ticularly commonly affected (339,346,350–352). Other common loca- intraparenchymal foci of abnormal signal are typically 3 cm or less in
tions include the thalami, pons, cerebellum, and the base of the frontal diameter; they are most commonly seen in the anterior temporal lobe,
lobe (335,339,346,352). It is likely that the preferential spread to these cerebellum, and brainstem (Figs. 6-53 and 6-54) (352,354). The T1 and
locations is a result of their close proximity to the basilar meninges. T2 shortening is presumably the result of the presence of paramagnetic
stable free radicals in melanin (identified by electron spin resonance
studies) (355,356). The unpaired electrons in free radicals interact with
Neuroimaging Manifestations
water protons via dipole-dipole interactions with subsequent shortening
CT scans in children with NCM may show the associated hypoplasia of both T1 and T2 relaxation times (357,358). When extensive melano-
of the cerebellum or pons, but the foci of melanin-containing cells sis is present, affected children typically have hydrocephalus (Fig. 6-55).

FIG. 6-53. Neurocutaneous melanosis. A. Sagittal


T1-weighted image in a 3-month-old infant shows
hyperintensity in the basis pontis (black arrows)
and along the surface of the vermis (white arrows).
The pons and vermis are slightly small. B. Axial
T1-weighted image shows hyperintense melanin
deposits in the basis pontis (white arrows), right
amygdala (large black arrow), and cerebellar folia
(small black arrows). C. Sagittal T1-weighted image
of the same child at age 5 years shows hypoplasia of
the pons and cerebellum (arrows), but no evidence
of leptomeningeal melanosis. Melanosis becomes
more difficult to detect as the brain matures. It is
not certain whether this reflects changes in the lep-
tomeningeal melanosis or changes of the relative
signal intensities of the melanotic deposits and the
brain parenchyma.

Barkovich_Chap06.indd 615 5/6/2011 9:04:37 PM


616 Pediatric Neuroimaging

FIG. 6-54. Neurocutaneous melanosis


with extensive cerebellar involvement.
A. Axial T1-weighted image shows foci of
hyperintense melanin (white arrows) ante-
rior to the temporal horns of the lateral
ventricles. B. Coronal T1-weighted image
shows extensive hyperintensity within the
cerebellar hemispheres. Both hemispheres
are hypoplastic, the left more so than the
right.

FIG. 6-55. NCM with hydrocephalus. A. Sagittal


T1-weighted image shows severe hydrocephalus. The cer-
ebellum appears slightly small and the cisterna magna is
enlarged. B. Axial T1-weighted image shows multiple hyper-
intense deposits of melanin in the cerebellar hemispheres. C.
Axial T1-weighted image at the level of the midbrain shows
bilateral melanin deposits (white arrows) anterior to the tem-
poral horns of the lateral ventricles.

Barkovich_Chap06.indd 616 5/6/2011 9:04:38 PM


Chapter 6 • The Phakomatoses 617

Degeneration of the melanocytic accumulation into melanoma can The mutation appears to be lethal in males; in the few reported male
only be determined by the identification of progressive growth of the cases, postzygotic mosaicism for the NEMO gene is evident (366).
lesion, by the presence of surrounding edema or mass effect, or by the The characteristic cutaneous lesions are linear vesicular or bullous
presence of central necrosis (352). Some authors have identified abnor- eruptions that begin in the first 6 months of life (367). Four stages of
mal meningeal enhancement on MR studies of patients with NCM cutaneous lesions are described: stage 1 is characterized by erythema,
(359,360); when seen in patients with increased intracranial pressure, vesicles, and pustules; stage 2 by papules, verrucous lesions, and hyper-
this finding likely represents diffuse leptomeningeal spread of tumor keratosis; stage 3 by hyperpigmentation; and stage 4 by pallor, atrophy,
(360). However, enhancement is almost never seen until the melanotic and scarring (368). Ocular abnormalities are primarily retinal vascular
deposits have undergone malignant degeneration and spread through- anomalies with consequent retinal fibrosis (369); microphthalmia may
out the subarachnoid spaces (360). be seen in conjunction with persistent hyperplastic primary vitreous.
Cerebellar (primarily vermian) and brainstem hypoplasias The CNS is affected in 30% to 50% of affected patients, with resultant
(Fig. 6-53) are typically seen when melanosis is present around the epilepsy (about 25%), intellectual deficit (<10%), spastic or flaccid
cerebellum and pons, respectively (352,354). Over time, the melanotic quadriplegia, and ataxia (370,371). Symptoms may begin any time in
deposits in the leptomeninges may become cryptic to imaging detec- childhood, sometimes as early as the first week of life; neonatal enceph-
tion (Fig. 6-53C); when imaged at this stage, patients may be mistak- alopathy has been described (372,373). Affected patients become pro-
enly diagnosed as having idiopathic pontocerebellar hypoplasia, unless gressively microcephalic.
this history of large cutaneous melanotic nevi in infancy is obtained. In
up to 10% of patients with NCM and cerebellar hypoplasia, the fourth
ventricle enlarges and a diagnosis of the Dandy-Walker malformation Pathological and Neuroimaging Manifestations
can be made (335,361–363). Other findings that have been reported
include tethered spinal cord (338), spinal lipomas (usually subpial), Few reports on the neuropathology of incontinentia pigmenti have
and arachnoid cysts (both intracranial and intraspinal) (337). been published (374,375). The reports suggest the presence of zones
of neuronal loss accompanied by regions of minor cortical dysplasia
(374,375). Some authors suggest an inflammatory process, while oth-
INCONTINENTIA PIGMENTI ers have found no evidence of inflammation and suggest vasculopathy
as the cause (372,373).
Clinical Manifestations
Findings on neuroimaging studies are variable. Patients who are
Incontinentia pigmenti, or Bloch-Sulzberger syndrome, is a rare disor- neurologically normal usually have normal imaging studies (376,377).
der characterized by congenital skin lesions, dental and skeletal dyspla- Those with neurologic signs and symptoms have regions of hypoden-
sia, ocular abnormalities, and nonprogressive CNS involvement (364). sity (on CT) or T2 hyperintensity (on MRI) (Fig. 6-56) involving the
The name is derived from an apparent dislocation of melanin from cerebral cortex and subjacent white matter, either bilaterally or in the
the basal cell layer of the epidermis to the upper dermis (365). The hemisphere contralateral to the clinical symptoms (364,376). T1 hyper-
disorder is almost exclusively seen in females, with mutations localized intensity may be present in the affected cortex of young infants (Fig.
to the IKK-gamma gene (also called NEMO) at chromosome Xq28. 6-56) (372,378). Cerebellar lesions, when present, also involve the cortex

FIG. 6-56. Incontinentia pigmenti in a 3-week-old baby. A. Axial noncontrast CT scan shows edema (arrows) in the left hemisphere, the posterior right
hemisphere, and in the left thalamus. B. Axial T2-weighted image shows multifocal hyperintense lesions corresponding to the regions of abnormal low
attenuation seen on the CT image (A).

Barkovich_Chap06.indd 617 5/6/2011 9:04:40 PM


618 Pediatric Neuroimaging

FIG. 6-56. (Continued) C. Axial noncontrast T1-weighted image shows multiple foci of hypointensity and hyperintensity, likely representing encepha-
lomalacia with gliosis, calcification, or subacute blood. D. Coronal gradient-echo T2-weighted image confirms the presence of multiple small hemorrhagic
foci revealed as foci of hypointense T2* signal (arrows).

and underlying white matter and have similar signal characteristics to many different genetic disorders, and that the chromosomal abnormalities
the cerebral lesions (364). The regions that are involved can correspond disrupt expression or function of pigmentary genes (381,382).
to intervascular boundary zones (“watershed zones,” see Chapter 4), The most characteristic clinical feature of hypomelanosis of Ito is
suggesting a possible vascular cause for the lesions; other lesions are the presence of cutaneous hypopigmented zones with irregular borders,
not vascular in distribution, suggesting an inflammatory origin (376). streaks, whorls, and patches (383,384). Other skin alterations are seen
Affected neonates and young infants may also have foci of T1 hyperin- in up to 40% of affected patients and include café au lait spots, angiom-
tensity and T2 hypointensity in the periventricular white matter (Fig. atous nevi, nevus marmorata, nevus of Ota, Mongolian blue spot,
6-56), likely representing either hemorrhage or reactive astrogliosis; heterochromia of the iris or hair, diffuse alopecia, grayish-white trichor-
focal regions of T2 hyperintensity are seen in the cortex. Sequential rhexia, and mottled hair (385). Musculoskeletal abnormalities include
scans in affected infants show progressive cortical and, more promi- hemihypertrophy, kyphosis, scoliosis, hyperlordosis, rudimentary ribs,
nently, white matter cavitation/atrophy, with an end result of asym- genu recurvatum, pes valgus, and pes cavus (384). Genital and cardiac
metric ventricular enlargement and an appearance of diffuse white anomalies are found in 10% or less of affected patients (384,385).
matter atrophy (similar to end-stage white matter injury of prematu-
rity, Chapter 4) (372,378). Patients with abnormalities detected in the Neurological Manifestations
cerebral hemispheres also have thinning of the corpus callosum (365),
presumably from damage to cortical neurons and the transcallosal Most clinical interest in hypomelanosis of Ito derives from the fre-
axons emanating from them. The ocular globes may show subretinal quency and importance of the neurological complications. These con-
hemorrhage, or abnormally high signal of the vitreous on T2-weighted sist primarily of seizures and intellectual disability, both of which are
images (364). Later in infancy or childhood, microphthalmia may be present in more than half of affected patients (384,385). Seizures usu-
seen, resulting from retinal vascular abnormalities (372), possibly via a ally develop during the first year of life and consist of infantile spasms,
similar mechanism that causes the brain injury. major motor seizures, partial motor seizures, and other forms of myo-
clonic epilepsy (384,385). While some patients suffer from generalized
seizures that are well controlled with anticonvulsant therapy, many
HYPOMELANOSIS OF ITO have severe, pharmacoresistant focal seizures (386). Developmental
delay is reported in 75% of affected patients and moderate to severe
Clinical Manifestations cognitive deficits (IQ < 70) in 57%; only 20% of patients have an IQ
Hypomelanosis of Ito (formerly referred to as incontinentia pigmenti above 85 (385). Other reported neurologic manifestations include
achromians [379]) is a common, but poorly known, neurocutaneous macrocephaly, microcephaly, and hypotonia (385).
disorder. The pathogenesis of the disorder is unknown and is likely mul-
tifactorial. Autosomal dominant inheritance has been seen in some (but Neuropathological and Neuroimaging
not all) cases; frequency is equal among males and females (380). Karyo-
Manifestations
type analysis reveals a variety of chromosomal rearrangements, gener-
ally associated with mosaicisms. Therefore, most experts now believe that Pathologic studies of hypomelanosis of Ito have shown malforma-
hypomelanosis of Ito is not a distinct entity but is rather a symptom of tions of cortical development, such as heterotopia and cerebral and

Barkovich_Chap06.indd 618 5/6/2011 9:04:41 PM


Chapter 6 • The Phakomatoses 619

FIG. 6-57. Odontogenic keratocysts in BCNS. A. Coronal noncontrast CT scan in a 10-year-old child shows a large mass (arrowheads) arising from the left
maxillary alveolar ridge. An unerupted tooth (arrow) is evident superiorly. B. Coronal fast spin-echo T2-weighted image in a 7-year-old child shows a small,
heterogeneous mass (white arrows) in the anterior left maxillary ridge. The unerupted tooth (arrowhead) is evident superiorly.

cerebellar polymicrogyria (387,388). Imaging studies have shown evi- (Figs. 6-57 and 6-58). The keratocysts are typically multiple (average,
dence of atrophy and hemiatrophy (384,389), white matter alterations 3–6; range, 1–30) and involve both the mandible and maxilla (398–
(prolonged T1 and T2 relaxation times) (384), hemimegalencephaly 400). They appear after the seventh year of life, and are present in 80%
(390) and more focal cortical malformations (384), lissencephaly (389), of affected patients over the age of 20 years (398). About half of patients
callosal hypogenesis (389), and polymicrogyria with heterotopia (389) with BCNS present with swelling, 25% with mild pain, and 15% with
(imaging characteristics of these malformations are discussed and an unusual taste following keratocyst rupture, while 30% are asymp-
illustrated in Chapter 5). Focal/unilateral anomalies causing intrac- tomatic (398). The cysts tend to recur after surgery. Rarely, ameloblas-
table seizures are amenable to surgical resection (391). In addition, a toma or squamous cell carcinoma may arise from the keratocyst (393).
number of patients have normal neuroimaging studies or show only The radiologic appearance is that of a sharply marginated lesion that
enlarged perivascular spaces. Given the range of genetic, clinical, and seems to originate from the root of a tooth (Fig. 6-57). The cyst has the
neuroradiological features, it seems best to think of hypomelanosis of signal of water (low attenuation on CT, hypointense on T1-weighted
Ito not as a specific entity but as a clinical syndrome encompassing MR images, and hyperintense on T2-weighted MR images). The over-
multiple cutaneous and CNS disorders. lying cortical bone is expanded and thinned.
Early calcification of the dura mater is a common radiologic
BASAL CELL NEVUS SYNDROME manifestation of the BCNS (393). The falx cerebri is the dural struc-
ture most commonly affected (in 65%–90%), with the diaphragma
Clinical Manifestations sella (60%–80%), tentorium (20%–40%), and petroclinoid ligaments
The basal cell nevus syndrome (BCNS), also known as nevoid basal cell (20%) all commonly involved in this process. This calcification can be
carcinoma syndrome and as Gorlin syndrome, is an autosomal domi- appreciated as increased attenuation and irregular thickening of these
nant disorder characterized by five major components: numerous basal structures on CT (Fig. 6-58B and C) and as hypointensity with irregu-
cell cancers and epidermal cysts of the skin, odontogenic keratocysts lar thickening on MRI. MRI demonstrates dysgenesis or agenesis of the
of the mandible and maxilla, palmar and plantar pits, calcification of corpus callosum in approximately 10% of cases (399).
the falx cerebri, and skeletal anomalies (392,393). Macrocephaly, often Developmental anomalies of the spinal column are common, with
pronounced, is evident in 50% of cases (394). The cause is believed to kyphoscoliosis (25%–40%), cervical or thoracic spinal bifida occulta
be a mutation of the PTCH gene that is located at chromosome 9q22.3 (50%–60%), and a lack of segmentation of cervical or upper thoracic
(395). Patients most commonly are initially seen due to development of vertebral bodies (30%–40%) being the most common manifestations
basal cell carcinomas, which may appear as early as age of 2 years (396). (393). Anomalies of the spinal cord have not been reported.
Basal cell carcinomas more commonly proliferate during puberty, with Patients with BCNS have an increased incidence of medullo-
the nape of the neck being the most common location. An enormous blastoma (Fig. 6-59); the frequency of medulloblastoma in patients
number of neoplasms and anomalies have been described in this syn- with the syndrome is 4% to 20%, whereas the frequency of BCNS
drome (393,397). As this is a neuroradiology text, only manifestations among patients with medulloblastoma is 1% to 2% (401,402). Indeed,
in the face, spine, and CNS are covered. medulloblastoma may be the cause of initial presentation of the BCNS.
BCNS patients with medulloblastoma characteristically present dur-
ing the first 2 years of life (397), in contrast to the usual presentation
Neuroimaging Manifestations
between age of 5 and 9 years in the general population. Therefore, chil-
The most common manifestations seen on neuroimaging are dren who are diagnosed with medulloblastoma before the age of 4 years
odontogenic keratocysts and calcifications of the intracranial dura should be carefully examined for other signs of the BCNS; in this situation,

Barkovich_Chap06.indd 619 5/6/2011 9:04:42 PM


620 Pediatric Neuroimaging

FIG. 6-58. BCNS and multiple radiation-induced tumors


in a teenager previously treated for a childhood medullo-
blastoma. A and B. Axial noncontrast CT scans at age 16
years show extensive calcification (black arrows) of the ten-
torium cerebelli and falx cerebri. An isodense meningioma
is evident (white arrows, B). Enlarged right sylvian fissure
(white arrow, A) results from volume loss secondary to ear-
lier resection of a meningioma. C–E. MRI study at age 19
years. Axial T2-weighted image (C) and contrast-enhanced
fat-suppressed T1-weighted image (D) show a new men-
ingioma (black arrows) arising from the left petrous apex,
with surrounding edema. A surgical defect is evident in the
right temporal lobe from prior meningioma resection. A
low T2 signal lesion (white arrows, C) is evident in the pos-
terior ethmoidal region, surrounded by T2 bright sinona-
sal secretions. Coronal fat-suppressed T2-weighted image
(E) through the nasal cavity demonstrates the mass (white
arrows), which fills the left nasal cavity and most of the eth-
moid air cells; it erodes the floor of the anterior cranial fossa
(white arrowhead). Biopsy revealed a rhabdomyosarcoma.

Barkovich_Chap06.indd 620 5/6/2011 9:04:42 PM


Chapter 6 • The Phakomatoses 621

FIG. 6-59. Early diagnosis of BCNS in


a 3-year-old child. A. Axial T2-weighted
image shows a heterogeneous, low signal
intensity mass (white arrows) within the
posterior portion of the right cerebellar
hemisphere. B. Axial noncontrast CT
scan obtained preoperatively revealed
multiple small dural-based calcifications
such as this one (white arrow) in the falx
cerebri.

the presence of dural-based calcification, even small ones (Fig. 6-59B), is facial Hemangiomas, Arterial anomalies, Cardiac anomalies and aortic
highly indicative of the BCNS (403,404). At pathology, there is a prepon- coarctation, Eye anomalies, and Sternal clefting and/or Supraumbili-
derance of two histological variants: medulloblastoma with extensive cal raphe), and the latter name has become standard. Affected patients
nodularity, and desmoplastic medulloblastoma (405). The radiologic typically present as a result of their cutaneous anomaly, typically
appearance of medulloblastoma in this group of patients does not dif- hemangiomas involving the face, neck, or scalp. As many as 20% of
fer in any way from that of nonsyndromic patients (discussed in Chap- patients with segmental hemangiomas (in distinction from localized
ter 7). It should be noted that because of the abnormality of a tumor hemangiomas) meet diagnostic criteria for PHACE (OMIM 606519)
suppressor gene, it is best to avoid radiation therapy for medulloblas- (410). Hemangiomas of the upper half of the face seem to correlate
toma treatment, as it can result in the subsequent appearance (over with structural brain, cerebrovascular, and ocular anomalies, while
a 10-year period) of a large number of tumors in the radiation field hemangiomas in a mandibular distribution are associated with ventral
(Fig. 6-58) (403,404,406). Basal cell carcinomas, which are the most developmental defects, such as sternal abnormalities and supraumbili-
common secondary tumors, may be quite invasive and have a predilec- cal raphe (410). The segmental relationships of these vascular abnor-
tion for perineural spread. Therefore, contrast-enhanced MR scans should malities suggest a common metameric origin of cells originating in
be obtained and the cranial nerves should be particularly scrutinized in the neural crest (411,412). Most affected patients are neurologically
a search for enhancement. Postradiation meningiomas, schwannomas, normal but have some degree of intellectual disability (408). However,
sarcomas, and osteochondromas have also been reported (406). among those patients with structural brain anomalies, neurodevelop-
Other tumors of the CNS are much less common. Meningiomas mental abnormalities are found in 90% (413).
(Fig. 6-58) appear to have an increased incidence (397), and astro- Many authors make the point that patients with this syndrome are
cytomas, craniopharyngiomas, and oligodendrogliomas have been easily differentiated from those with the Sturge-Weber syndrome by
reported in patients with the syndrome (393). These tumors all appear both differences in the cutaneous lesions and in the intracranial vascu-
to have a similar radiologic appearance and clinical behavior to those lar anomalies. To summarize, patients with Sturge-Weber have a large
occurring in the general population (see Chapter 7). segmental facial hemangioma, most commonly involving the lateral
forehead (S2) or the maxillary region (S3) (414); about 20% of these
patients have anomalies of brain parenchyma or the blood vessels that
PHACE (POSTERIOR FOSSA feed the brain (410). Intracranially, patients with Sturge-Weber have a
MALFORMATIONS, FACIAL capillary/venous malformation that results in impaired cortical venous
HEMANGIOMAS, ARTERIAL ANOMALIES, drainage and progressive brain injury, whereas patients with the cuta-
CARDIAC ANOMALIES AND AORTIC neous hemangioma-vascular complex syndrome have persistence of
primitive arteries or absence/dysgenesis of normal major intracranial
COARCTATION, EYE ANOMALIES) arteries (408). As a result of these vascular anomalies, patients with the
SYNDROME PHACE syndrome are at a high risk for the development of cerebral
aneurysms.
Clinical Manifestations
The most common extracutaneous anomalies of these patients
Both Pascual-Castroviejo and his colleagues and Frieden et al. have involve the CNS, where structural abnormalities are present in 50% of
reported that a high percentage of patients with cutaneous vascular patients (414). Cerebellar anomalies appear to be the most common
malformations of the head and neck have associated anomalies of structural brain anomaly seen in PHACE syndrome, most commonly
the intracranial structures, particularly blood vessels (407,408,409). cerebellar hemispheric or vermian hypoplasias (410); true Dandy-
Both groups have proposed that this association of cutaneous and Walker malformations are much less common (414). Polymicrogyria
intracranial pathology constitutes a phakomatosis. Although Pascual- (415), cerebral hemispheric hypoplasia, periventricular nodular het-
Castroviejo et al. (408) have proposed that it be called the cutaneous erotopia, anomalies of the corpus callosum, anomalies of the septum
hemangioma-vascular complex syndrome, Frieden et al. (409) have pellucidum, and intracranial hemangiomas are also described (416);
suggested the term PHACES syndrome (Posterior fossa malformations, these are often ipsilateral to the facial hemangioma (414). When

Barkovich_Chap06.indd 621 5/6/2011 9:04:44 PM


622 Pediatric Neuroimaging

present, the intracranial hemangioma is most commonly located in the to the facial hemangioma), optic nerve hypoplasia/atrophy, iris vessel
internal auditory meatus ipsilateral to the facial hemangioma; ipsilat- hypertrophy, iris hypoplasia, and congenital cataracts. Reported ventral
eral cerebellar hemisphere hypoplasia may be present (416). defects include sternal clefting and supraumbilical raphe (410,413).
Cerebrovascular anomalies associated with PHACE are encoun- Otolaryngic abnormalities are also common, and include middle ear
tered in about 80% of cases and comprise a spectrum of congenital atelactasis, tympanic membrane, and airway hemangiomas (425).
and progressive large artery lesions. They can be classified into five An association between combined venous lymphatic vascular mal-
major groups: (a) hypoplasia or agenesis of major cerebral vessels, formations (CVLVMs, formerly called lymphangiomas) of the orbit
(b) persistence of embryonic vessels, (c) progressive vascular stenosis and intracranial venous malformations has been reported (426). Katz
or occlusion, (d) segmental dolichoectasia of cerebral vessels, and (e) et al. (426) found that 28% (7/25) of patients with orbital CVLVMs
anomalous vasculature (417,418). The most common arterial anoma- had venous malformations of the brain, most commonly involving the
lies (seen in up to 60% of affected patients) are saccular aneurysms, cerebellum, brainstem, and deep cerebral nuclei. The authors found
aberrant origins or courses of the internal carotid and major intracere- that those patients with associated intracranial venous malformations
bral arteries, arterial dysplasias, and persistence of fetal anastomoses. had diffuse orbital lesions with bony expansion of the orbit, extension
Agenesis of cerebral vessels, segmental stenoses, and occlusions of the of the orbital malformation through the superior orbital fissure, both
major intracranial vessels may also be detected (418–422). Hypoplasia superficial and deep components of orbital involvement, and, in the
of the A1 segment of the anterior cerebral artery is common in the nor- majority of cases, both intraconal and extraconal involvement. These
mal population (13%) but seems to be much higher in patients with venous malformations are not a part of the PHACES syndrome.
PHACE (414). Progressive occlusive vasculopathy, with a moyamoya-
like pattern, is also described (423). Several authors have suggested that
Neuroimaging Manifestations
affected children have an increased incidence of cerebral infarction
(422). Cardiac or aortic anomalies are seen in about 35% of patients, The most common anomalies are vascular and include anomalous
with aortic coarctation, patient ductus arteriosus, and ventricular sep- course or origin of the anterior cerebral arteries; persistence of the
tal defects being the most common (424). Ophthalmologic anomalies embryonic carotid-vertebrobasilar arterial connections (persistent
are seen in about 20%, most commonly microphthalmos (ipsilateral trigeminal artery is most common, Fig. 6-60); absence or hypoplasia

FIG. 6-60. PHACES syndrome in a 10-year-old girl. A. Axial


T2-weighted image shows a small right cerebellar hemisphere
(black arrowheads) and an anomalous vessel (black arrow) in the
prepontine cistern. B. Maximum intensity projection image from
a 3D time of flight angiogram shows that the anomalous vessel is a
persistent right trigeminal artery (arrowheads). C. Collapsed view
from a 3D time of flight MR angiogram shows a dysplastic right
middle cerebral artery (arrows) as well as the persistent trigeminal
artery.

Barkovich_Chap06.indd 622 5/6/2011 9:04:45 PM


Chapter 6 • The Phakomatoses 623

FIG. 6-61. PHACES syndrome. A and B. Axial contrast-enhanced fat-suppressed T1-weighted image (A) and axial T2-weighted image (B) show a large
hemangioma (arrows) infiltrating the anterolateral aspect of the right orbit and the right middle cranial fossa. The signal void of the right carotid artery is
absent. The right trigeminal cistern (black arrowheads) is enlarged. An enhancing lesion is seen in the right cerebellopontine angle cistern (white arrowhead
in A), consistent with an intracranial hemangioma (not seen in B due to isointensity with the cistern). The right cerebellar hemisphere is small. The inter-
hemispheric fissure of the cerebellum (white arrowheads in B) is too prominent, indicating hypoplasia of the vermis.

of the internal carotid artery (Fig. 6-61); absence of the external become manifest at birth or during the neonatal period (429). The
carotid artery and absence of hypoplasia of the vertebral, posterior cutaneous lesions vary from 5 to 15 mm in diameter; they range in
cerebral, and posterior inferior cerebellar arteries (418). Large, dys- number from 50 to more than 100 (430). Other commonly involved
morphic vessels (dysgenesis) may also be seen (Fig. 6-60C) (418), as organ systems include the liver, CNS, intestine, and lungs; nearly
can occlusive vasculopathies that may result in the moyamoya syn- any organ can be involved (431). If untreated, up to 60% of affected
drome (423). These vascular anomalies are typically associated with patients die in the first few months of life as a result of high out-
hemangiomas of the S1 (frontotemporal) and/or S3 (mandibular) put cardiac failure (431). Treatment options include steroid therapy,
facial segments. The most common parenchymal malformations of radiation, laser therapy, cyclophosphamide, and angiogenesis inhibi-
the PHACES syndrome are diffuse cerebellar hypoplasia (including tors (430,432).
the Dandy-Walker malformation, see Chapter 5), vermian hypoplasia Neuroimaging studies of affected patients show multiple heman-
(Fig. 6-61B), and unilateral cerebellar hemispheric hypoplasia (Figs. giomas and hemorrhages in the brain and spinal cord (429,433,434).
6-60 and 6-61) (408). Several patients have been reported to have Hemorrhage is common because the hemangiomas are composed
polymicrogyria, periventricular nodular heterotopia, and intracranial of dilated, thin-walled channels lined by a single layer of endothelial
hemangiomas (427). Anomalies of the aortic arch and cardiac mal- cells. When multiple cerebral hemorrhages are seen in a neonate with
formations are also reported (408). The major importance of recog- multiple cutaneous hemangiomas and congestive heart failure, the
nizing the existence of this syndrome is to recognize that all patients diagnosis of diffuse neonatal hemangiomatosis should be strongly
with hemangiomas of the face should have an intracranial study that considered.
includes both an imaging study of the brain (with particular attention
to the posterior fossa) and a vascular imaging study that visualizes the
aortic arch and the cervical and intracranial portions of the carotid and
CHÉDIAK-HIGASHI SYNDROME
vertebral arteries all the way to, and including, the major branches of the The Chédiak-Higashi syndrome is a rare, autosomal recessive syn-
circle of Willis (418,428). drome characterized by hypopigmentation (partial oculocutaneous
albinism), photophobia, decreased lacrimation, nystagmus, recurrent
pyogenic infections, and intermittent febrile episodes. The diagnosis is
DIFFUSE NEONATAL HEMANGIOMATOSIS established by examination of the blood, which demonstrates the pres-
Diffuse neonatal hemangiomatosis is a rare condition characterized ence of characteristic large peroxidase-positive panleukocytic granules,
by the presence of numerous progressive, rapidly growing cutane- and by identification of mutations of the CHS1 gene. Affected patients
ous hemangiomas associated with widespread visceral hemangiomas may come to attention because of recurrent infection, bleeding dis-
of the liver, lungs, gastrointestinal tract, brain, and meninges that orders, or neurologic signs and symptoms (435). Alternatively, they

Barkovich_Chap06.indd 623 5/6/2011 9:04:46 PM


624 Pediatric Neuroimaging

may present with the onset of an accelerated phase of the syndrome, in ENS, linear sebaceous nevus syndrome, and congenital hemidysplasia
which a lymphohistiocytic infiltration occurs; they may subsequently with ichthyosiform erythroderma and limb defects (CHILD syndrome)
develop erythrophagocytosis, hepatosplenomegaly, and lymphade- within the large category of ENS, while others split them into separate
nopathy (437). The neurologic signs and symptoms generally reflect groups (453).
brainstem and cerebellar involvement and include motor incoordina- The true incidence of ENS is unknown, but it is estimated that
tion, horizontal nystagmus, intention tremor, and dysmetria. Seizures, 8% to 18% of patients with epidermal nevi have systemic disorders
peripheral neuropathies, and mental retardation may also be present (451). It is not clear whether the location of the nevus correlates with
(435,436). Neuropathology shows perivascular cellular infiltrations development of the associated neurologic syndrome; some authors
with scattered microscopic gliomas in the cerebellum, brainstem, spi- have noted a high incidence of nevi on the head and neck in patients
nal cord, and peripheral nerves (438). Treatment is bone marrow trans- with neurologic and ocular symptoms (454), but others dispute this
plantation, which is effective in treating the hematologic and immune correlation (455,456).
deficits (439).
Reports of neuroimaging studies in Chédiak-Higashi syndrome
Ocular and Brain Manifestations
are few (440). In our case, the patient had delayed myelination, promi-
nent CSF spaces (probable cerebral atrophy), and nonenhancing focal Ocular involvement is present in 45% to 68% of affected patients
regions of T2 prolongation in the cerebellum and brainstem. Ballard et (455,456,457). Many different ocular anomalies have been described,
al. (440) reported similar findings in the cerebral white matter that did including optic disc colobomas, optic nerve dysplasia, retinal dyspla-
not significantly change over a 1 month follow-up period. sia, choroidal osteomas, anterior segment dysgenesis, and peripapillary
staphyloma (457).
CNS manifestations are evident in a majority of patients
PROGRESSIVE FACIAL HEMIATROPHY (Table 6-6); unilateral hemimegalencephaly is the most striking,
(PARRY-ROMBERG SYNDROME) typically developing ipsilateral to the skin lesions. Baker et al. (458)
The Parry-Romberg syndrome is a rare, acquired disorder character- found a 7% incidence of hemiatrophy, a 7% incidence of hemimega-
ized by progressive and self-limited atrophy of the skin, subcutaneous lencephaly, a 3% incidence of isolated gyral malformations, and no
tissues, and sometimes the underlying bone. It may start and stop at any posterior fossa malformations. Gurecki et al. (456) found CNS lesions
time during the first 2 to 3 decades of life, resulting in variable degrees by imaging in 20/23 (87%) who had biopsy-proven epidermal nevi.
of deformity (441). Neurologic symptoms are evident in about 15% Seizures were reported in 52%, intellectual impairment in 53%, and
of cases (442). Affected patients often develop partial epilepsy during motor impairment in 91%. Imaging findings included heterotopia,
the course of their disease (443); as this epilepsy is often refractory to callosal hypogenesis, cerebellar vermian hypogenesis, and focal corti-
medication, neuroimaging studies are frequently requested. cal malformations. Pavone et al. (454) gathered data on 63 patients
Few reports of neuroimaging in Parry-Romberg syndrome are with ENS. They found a high incidence (27%) of hemimegalenceph-
available (443–446). Cortical calcifications may be seen on CT studies aly in these patients. They also noted the presence of infarcts, atrophy,
(444,445). Most reports describe low density in the cerebral white mat- porencephaly, and calcifications in 5 of 17 patients with hemimegal-
ter and enlargement of the lateral ventricle ipsilateral to the facial atro- encephaly, suggesting that underlying vascular dysplasia predisposes
phy (445). In addition, T1 hypointensity and T2/FLAIR hyperintensity to these acquired lesions (454). Vascular dysplasias (ICA hypoplasia/
are present in the ipsilateral greater than contralateral cerebral white occlusion, cerebral and spinal arteriovenous malformations) and vas-
matter, and small cysts may be seen in the deep gray matter and cor- cular lesions (cerebral infarction, porencephaly) have frequently been
pus callosum (443,444,447). DTI fiber tractography has demonstrated reported; these observations support the theory that a vascular dyspla-
asymmetric involvement of white matter tracts (446). Interestingly, the sia is present in at least some patients with this disorder (456,459,460).
sulci of the overlying cerebral cortex are small (effaced), rather than
large, and the MR spectra in the affected hemisphere can be normal
(447), suggesting that this is not a purely atrophic process. Indeed, the
few pathologic studies suggest a perivascular infiltrate in the affected
regions (suggestive of Rasmussen encephalitis) and anti-DNA antibod-
TABLE 6-6 Neuroimaging Findings
ies are found in some patients, suggesting that this is an autoimmune in ENS
inflammatory disorder (442,449).
Primary
EPIDERMAL NEVUS SYNDROME Malformations of cortical development (hemimegalencephaly,
polymicrogyria)
Clinical Manifestations
Gliomatosis
The term epidermal nevus syndrome (ENS) refers to sporadic over- Hemiatrophy with and without parenchymal cysts
growth neurocutaneous disorders of unknown etiology characterized
by the combination of an epidermal nevus and a significant abnor- Vascular malformations
mality of the CNS, eye, or skeleton (449). The recent literature utilizes Intracranial/intraspinal lipomas
the term “epidermal nevus” to refer to slightly raised ovoid or linear
Secondary
plaques that are usually skin-colored in infancy but become more ver-
rucous and darker as time passes; these hamartomatous lesions are Porencephaly
encountered in approximately 1 in 1,000 newborns (450). The term Infarctions
encompasses a number of different histologic types of cutaneous
lesions (450,451). Other authors include the Proteus syndrome, Feuer- Atrophy
stein-Mims syndrome, nevus comedonicus syndrome, pigmented hairy

Barkovich_Chap06.indd 624 5/6/2011 9:04:47 PM


Chapter 6 • The Phakomatoses 625

have abnormally enhancing and enlarged nerve roots in the spinal


canal (461,462).

ENCEPHALOCRANIOCUTANEOUS
LIPOMATOSIS
Clinical Manifestations
Encephalocraniocutaneous lipomatosis (ECCL) is a sporadic over-
growth neurocutaneous syndrome characterized by the presence of
skin lesions and ocular and CNS anomalies; the etiology of the dis-
order is unknown. The two most common clinical markers of the
disorder are ocular choristomas (including epibulbar and limbal der-
moids) and the nevus psiloliporus (a smooth, hairless fatty tissue of
the scalp) (463). Other skin lesions encountered include nonscarring
alopecia, subcutaneous fatty masses, and aplastic scalp defects. Jaw
tumors, bone cysts, and aortic coarctation are rarely seen. About two
thirds of patients develop normally or have mild mental retardation;
about one third has severe mental retardation. Seizures are present in
about half of patients. ECCL has been considered a localized form of
Proteus syndrome. However, Proteus syndrome is a progressive disor-
FIG. 6-62. Hemimegalencephaly in the ENS. Axial T2-weighted image der; signs and symptoms of ECCL in general are present at birth and
of a 5-month-old baby shows relative enlargement of the right cerebral are nonprogressive, with the possible exception of skeletal cysts and
hemisphere with thick cortex, anomalous sulcation (large arrowheads), jaw tumors (463).
and abnormal hypointensity of periventricular and deep white matter
(arrows).
Neuroimaging Manifestations
Intracranial lipomas are encountered in the majority of the patients;
MR imaging of patients with ENS reveals developmental malforma- the cerebellopontine angle is commonly affected (464,465). Spinal
tions (Fig. 6-62). Neonatal white matter injury (with normal appearing lipomas are found in most patients and can extend over long seg-
cortex) has also been documented in ENS (Fig. 6-63), supporting the ments; spinal MRI is recommended when ECCL is suspected. Con-
theory of a vascular etiology of destructive lesions in some affected genital abnormalities of the meninges are also commonly seen, in
patients. Intracranial and intraspinal lipomas have been reported, as particular arachnoid cysts (Fig. 6-64). Vascular dysplasias (leptom-
eningeal angiomatosis [Fig. 6-64] and abnormal or excessive vessels)
and ischemic lesions (atrophy, porencephaly, hemispheric calcifica-
tions) can also be seen (464,465). Therefore, the clinical and radio-
logic manifestations of ECCL and ENS overlap significantly; a main
differential criterion is the skin lesion, which is complex in the ENS
and includes different forms of epidermal nevi, which change over
time (463).

OTHER OVERGROWTH SYNDROMES


Proteus syndrome is a complex hamartomatous disorder named after
the Greek god Proteus, who could change his form at will. It has mul-
tiple, diverse somatic manifestations that evolve over time and involve
the skeletal system, soft tissues, skin, and vascular systems (466). Partial
gigantism of the hands and/or feet, asymmetry of limbs, macrodac-
tyly, bony exostoses, soft tissue tumors (hemangioma, lymphangioma,
lipoma), and long bone overgrowth are common. Hemimegalenceph-
aly, polymicrogyria, and heterotopia are the most commonly reported
congenital anomalies of the CNS; others include abnormalities of the
corpus callosum, Dandy-Walker malformation, and periventricular
calcifications (467,468).
Cowden syndrome is an autosomal dominant multisystem disor-
der involving hamartomatous overgrowth of tissues of all three embry-
onic origins––including the CNS––and an increased predisposition to
cancers of the breast, thyroid, and endometrium. Approximately 80%
of patients have an identifiable germline mutation of the PTEN gene
FIG. 6-63. Periventricular white matter injury in ENS. Axial T1-weighted on chromosome 10 (288,469). Within the CNS, the pathognomonic
image shows multiple foci of T1 hyperintensity (open black arrows) in the feature is the presence of Lhermitte-Duclos disease (dysplastic ganglio-
periventricular white matter. cytoma of the cerebellum, see Chapter 5), although other associations

Barkovich_Chap06.indd 625 5/6/2011 9:04:47 PM


626 Pediatric Neuroimaging

FIG. 6-64. ECCL in a 3-month-old baby. A. Coronal non-


contrast T1-weighted image shows mild thickening of the
right-sided subcutaneous fat (arrows). The ipsilateral cerebral
hemisphere is atrophic, with enlargement of the lateral ventricle
and subarachnoid spaces. B. Axial post contrast T1-weighted
image with fat suppression demonstrates abnormal thicken-
ing and enhancement of the meninges (white arrows) over the
right lateral convexity. C. Axial T2-weighted image at a lower
level reveals a cyst in the right middle cranial fossa, distorting
the right temporal pole (arrows).

include macrocephaly, gray matter heterotopia, seizures, vascular REFERENCES


abnormalities, and mental retardation (470,471).
Bannayan-Riley-Ruvalcaba (BRR) syndrome also results from 1. Kleinerman RA. Radiation-sensitive genetically susceptible pediatric sub-
populations. Pediatr Radiol 2009;39(Suppl 1):S27–S31.
mutations in the PTEN gene. This autosomal dominant syndrome is
2. Datson MM, Scrable H, Norlund M, Sturbaum AK, Nissen LM, Ratner
associated with extreme macrocephaly and subtle cutaneous features
N. The protein product of the neurofibromatosis type 1 gene is expressed
(including lipomas and hemangiomata); motor delays are common. at highest abundance in neurons, Schwann cells, and oligodendrocytes.
BRR and Cowden syndrome likely represent age-related manifestations Neuron 1992;8:415–428.
of the same condition (472).

Barkovich_Chap06.indd 626 5/6/2011 9:04:47 PM


Chapter 6 • The Phakomatoses 627

3. Listernick R, Louis DN, Packer RJ, Gutmann DH. Optic pathway gliomas 30. Moore BD, Slopis JM, Schomer D, Jackson EF, Levy BM. Neuropsychologi-
in children with neurofibromatosis 1: consensus statement from the NF1 cal significance of areas of high signal intensity on brain MRIs of children
optic pathway glioma task force. Ann Neurol 1997;41:143–149. with neurofibromatosis. Neurology 1996;46:1660–1668.
4. Seizinger BR, Rouleau GA, Ozelins LJ, et al. Genetic linkage of von Reck- 31. North K, Joy P, Yuille D, et al. Specific learning disability in children with
linghausen neurofibromatosis to the nerve growth factor receptor gene. neurofibromatosis type 1: significance of MRI abnormalities. Neurology
Cell 1987;49:589–594. 1994;44:878–883.
5. Von Recklinghausen FD. Uber die multiplen Fibrome der Haut und ihre 32. Hyman SL, Gill DS, Shores EA, Steinberg A, North KN. T2 hyperintensities
Beziehung zu den multiplen neuromen. Festschrift fur Rudolph Virchow. in children with neurofibromatosis type 1 and their relationship to cogni-
Berlin: August Hirschwald, 1882. tive functioning. J Neurol Neurosurg Psychiatry 2007;78:1088–1091.
6. Gutmann DH. Neurofibromin in the brain. J Child Neurol 2002;17:592– 33. Leppig K, Kaplan P, Viskochil D, et al. Familial neurofibromatosis 1
601; discussion 602–594, 646–551. microdeletions: cosegregation with distinct facial phenotype and early
7. Williams VC, Lucas J, Babcock MA, Gutmann DH, Korf B, Maria BL. Neu- onset of cutaneous neurofibromata. Am J Med Genet 1997;73:197–201.
rofibromatosis type 1 revisited. Pediatrics 2009;123:124–133. 34. Korf B, Schneider G, Poussaint T. Structural anomalies revealed by neu-
8. Larizza L, Gervasini C, Natacci F, Riva P. Developmental abnormalities roimaging studies in the brains of patients with NF-1 and large deletions.
and cancer predisposition in neurofibromatosis type 1. Curr Mol Med Genet Med 1999;1:136–145.
2009;9:634–653. 35. DeRecondo J, Haguenau M. Neuropathologic survey of the phakomatoses
9. Dasgupta B, Gutmann DH. Neurofibromin regulates neural stem cell and allied disorders. In: Vinken PJ, Bruyn GW, eds. Handbook of Clinical Neu-
proliferation, survival, and astroglial differentiation in vitro and in vivo. J rology: The Phakomatoses, vol 14. Amsterdam: North Holland, 1972:19–71.
Neurosci 2005;25:5584–5594. 36. Fletcher WA, Imes RK, Hoyt WF. Chiasmal gliomas: appearance and long-
10. Li F, Munchhof AM, White HA, et al. Neurofibromin is a novel regulator term changes demonstrated by computerized tomography. J Neurosurg
of RAS-induced signals in primary vascular smooth muscle cells. Hum Mol 1986;65:154–159.
Genet 2006;15:1921–1930. 37. Shamji MF, Benoit BG. Syndromic and sporadic pediatric optic pathway
11. Cui Y, Costa RM, Murphy GG, et al. Neurofibromin regulation of ERK sig- gliomas: review of clinical and histopathological differences and treatment
naling modulates GABA release and learning. Cell 2008;135:549–560. implications. Neurosurg Focus 2007;23:E3.
12. Rosenbaum T, Kim HA, Boissy YL, Ling B, Ratner N. Neurofibromin, the 38. Alvord EC, Lufton S. Gliomas of the optic nerve or chiasm: outcome by
neurofibromatosis type 1 Ras-GAP, is required for appropriate P0 expres- patient’s age, tumor site, and treatment. J Neurosurg 1988;68:85–98.
sion and myelination. Ann N Y Acad Sci 1999;883:203–214. 39. Listernick R, Ferner RE, Liu GT, Gutmann DH. Optic pathway gliomas
13. Habib A, Gulcher JR, Hognason T, Zheng L, Stefansson K. The OMgp gene, in neurofibromatosis-1: controversies and recommendations. Ann Neurol
a secdond growth supressor within the NF1 gene. Oncogene 1998;16:1525– 2007;61:189–198.
1531. 40. Parazzini C, Triulzi F, Bianchini E, et al. Spontaneous involution of optic
14. Szudek J, Birch P, Riccardi V, Evans D, Friedman J. Associations of clinical pathway lesions in neurofibromatosis ytpe 1: serial contrast MR evaluation.
features in neurofibromatosis 1 (NF1). Genet Epidemiol 2000;19:429–439. Am J Neuroradiol 1995;16:1711–1718.
15. MacCollin M, Kwiatkowski D. Molecular genetic aspects of the phakoma- 41. Parsa CF, Hoyt CS, Lesser RL, et al. Spontaneous regression of optic
toses. Curr Opin Neurol 2001;14:163–169. gliomas: thirteen cases documented by serial neuroimaging. Arch Ophthal-
16. Sabbagh A, Pasmant E, Laurendeau I, et al. Unravelling the genetic basis mol 2001;119:516–529.
of variable clinical expression in neurofibromatosis 1. Hum Mol Genet 42. Piccirilli M, Lenzi J, Delfinis C, Trasimeni G, Salvati M, Raco A. Sponta-
2009;18:2768–2778. neous regression of optic pathways gliomas in three patients with neuro-
17. Berg B. Current concepts of neurocutaneous disorders. Brain Dev fibromatosis type I and critical review of the literature. Childs Nerv Syst
1991;13:9–20. 2006;22:1332–1337.
18. Berg B. Neurocutaneous disorders. In: Berg B, ed. Neurologic Manifestations 43. Stern J, DiGiacinto GV, Housepian EM. Neurofibromatosis and optic
of Pediatrics. Boston: Butterworth-Heinemann, 1992:485–498. glioma: clinical and morphological correlations. Neurosurgery 1979;4:
19. Huson SM, Harper PS, Compston DAS. Von Recklinghausen’s neurofi- 524–528.
bromatosis: A clinical and popluation study in southeast Wales. Brain 44. Stern J, Jacobiec FA, Housepian EM. The architecture of optic nerve gliomas
1988;111:1355–1381. with and without neurofibromatosis. Arch Ophthalmol 1980;98:505–548.
20. Kuenzle C, Weissert M, Roulet E, et al. Follow-up of optic gliomas in chil- 45. Lövblad KO, Remonda L, Ozdoba C, Huber P, Schroth G. Dural ectasia of
dren with neurofibromatosis type 1. Neuropediatrics 1994;25:295–300. the optic nerve sheath in neurofibromatosis type I: CT and MR features. J
21. Friedman JM, Birch P. An association between optic glioma and other Comput Assist Tomogr 1994;18:728–730.
tumors of the central nervous system in Neurofibromatosis type 1. Neuro- 46. Gonen O, Wang ZJ, Viswanathan AK, Molloy PT, Zimmerman RA. Three-
pediatrics 1997;28:131–132. dimensional multivoxel proton MR spectroscopy of the brain in children
22. Korf BR. Malignancy in Neurofibromatosis Type 1. Oncologist 2000;5: with neurofibromatosis type 1. Am J Neuroradiol 1999;20:1333–1341.
477–485 doi:10.1634/theoncologist.5–6–477. 47. Albers AC, Gutmann DH. Gliomas in patients with neurofibromatosis type
23. Cutting LE, Cooper KL, Koth CW, et al. Megalencephaly in NF1: predomi- 1. Expert Rev Neurother 2009;9:535–539.
nantly white matter contribution and mitigation by ADHD. Neurology 48. Canale DJ, Bebin J. Von Recklinghausen disease of the nervous system. In:
2002;59:1388–1394. Vinken PJ, Bruyn GW, eds. Handbook of Clinical Neurology: The Phakoma-
24. Vivarelli R, Grosso S, Calabrese F, et al. Epilepsy in neurofibromatosis 1. J toses, vol 14. Amsterdam: North Holland, 1972:132–162.
Child Neurol 2003;18:338–342. 49. Parizel P, Martin J, Van Vyre M, van den Hauwe L, De Schepper A. Cere-
25. North K. Neurofibromatosis type 1. Am J Med Genet 2000;97:119–127. bral ganglioglioma and neurofibromatosis type 1. Neuroradiology 1991;33:
26. North K, Joy P, Yuille D, Cocks N, Hutchins P. Cognitive function and aca- 357–359.
demic performance in children with neurofibromatosis type 1. Dev Med 50. Hochstrasser H, Boltshauser E, Valavanis A. Brain tumors in children with
Child Neurol 1995;37:427–436. Recklinghausen neurofibromatosis. Neurofibromatosis 1989;1:233–239.
27. Eldridge R, Denckla M, Bien E, et al. Neurofibromatosis type 1: neu- 51. Ullrich NJ, Raja AI, Irons MB, Kieran MW, Goumnerova L. Brainstem
rologic and cognitive assessment with sibling controls. Am J Dis Child lesions in neurofibromatosis type 1. Neurosurgery 2007;61:762–766; discus-
1989;143:833–837. sion 766–767.
28. Hoffman K, Harris E, Bryan E, Dencla M. Neurofibromatosis type 1: the 52. Bilaniuk L, Molloy P, Zimmerman R, et al. Neurofibromatosis type 1: brain
cognitive phenotype. J Pediatr 1994;124:51–58. stem tumors. Neuroradiology 1997;39:642–653.
29. Hyman S, Gill D, Shores E, et al. Natural history of cognitive deficits 53. Molloy PT, Bilaniuk LT, Vaughan SN, et al. Brainstem tumors in patients
and their relationship to MRI T2-hyperintensities in NF1. Neurology with neurofibromatosis type 1: a distinct clinical entity. Neurology
2003;60:1139–1145. 1995;45:1897–1902.

Barkovich_Chap06.indd 627 5/6/2011 9:04:49 PM


628 Pediatric Neuroimaging

54. Pollack IF, Shultz B, Mulvihill JJ. The management of brainstem gliomas in 78. Steen RG, Taylor JS, Langston JW, et al. Prospective evaluation of the brain
patients with neurofibromatosis 1. Neurology 1996;46:1652–1660. in asymptomatic children with neurofibromatosis type 1: relationship of
55. Kim G, Mehta M, Kucharczyk W, Blaser S. Spontaneous regression macrocephaly to T1 relaxation changes and structureal brain abnormali-
of a tectal mass in neurofibromatosis 1. Am J Neuroradiol 1998;19: ties. Am J Neuroradiol 2001;22:810–817.
1137–1139. 79. Wang PY, Kaufmann WE, Koth CW, Denckla MB, Barker PB. Thalamic
56. Mimouni-Bloch A, Kornreich L, Kaadan W, Steinberg T, Shuper A. Lesions involvement in neurofibromatosis type 1: evaluation with proton mag-
of the corpus callosum in children with neurofibromatosis 1. Pediatr Neurol netic resonance spectroscopic imaging. Ann Neurol 2000;47:477–484.
2008;38:406–410. 80. Carella A, Medicamento N. Malignant evolution of presumed genign lesion
57. Jung H, Neumaier Probst E, Hauffa BP, Partsch CJ, Dammann O. Asso- in the brain in neurofibromatosis: case report. Neuroradiology 1997;39:
ciation of morphological characteristics with precocious puberty and/ 639–641.
or gelastic seizures in hypothalamic hamartoma. J Clin Endocrinol Metab 81. Dubovsky EC, Booth TN, Vezina G, Samango-Sprouse CA, Palmer KM,
2003;88:4590–4595. Brasseux CO. MR imaging of the corpus callosum in pediatric patients
58. Hosoda K, Kanazawa Y, Tanaka J, Tamaki N, Matsumoto S. Neurofibroma- with neurofibromatosis type 1. Am J Neuroradiol 2001;22:190–195.
tosis presenting with aqueductal stenosis due to a tumor of the aqueduct: 82. Said SM, Yeh TL, Greenwood RS, Whitt JK, Tupler LA, Krishnan KR. MRI
case report. Neurosurgery 1986;19:1035–1037. morphometric analysis and neuropsychological function in patients with
59. Pou-Serradell A, Ugarte-elola A. Hydrocephalus in neurofibromatosis: neurofibromatosis. Neuroreport 1996;7:1941–1944.
contribution of MRI to its diagnosis. Neurofibromatosis 1989;2:218–226. 83. Pride N, Payne JM, Webster R, Shores EA, Rae C, North KN. Corpus Cal-
60. Afifi A, Jacoby C, Bell W, Menezes A. Aqueductal stenosis and neurofibro- losum Morphology and Its Relationship to Cognitive Function in Neuro-
matosis: a rare association. J Child Neurol 1988;3:125–130. fibromatosis Type 1. J Child Neurol 2010;25:834–841.
61. Brown EW, Riccardi VM, Mawad M, et al. MR imaging of optic path- 84. Rosser TL, Vezina G, Packer RJ. Cerebrovascular abnormalities in a popula-
ways in patients with neurofibromatosis. Am J Neuroradiol 1987;8: tion of children with neurofibromatosis type 1. Neurology 2005;64:553–555.
1031–1035. 85. Cairns AG, North KN. Cerebrovascular dysplasia in neurofibromatosis
62. Aoki S, Barkovich A, Nishimura K, et al. Neurofibromatosis types 1 and 2: type 1. J Neurol Neurosurg Psychiatry 2008;79:1165–1170.
cranial MR findings. Radiology 1989;172:527–536. 86. Rea D, Brandsema JF, Armstrong D, et al. Cerebral arteriopathy in children
63. van Englen SJPM, Krab LC, Moll HA, et al. Quantitative differentiation with neurofibromatosis Type 1. Pediatrics 2009;124(3):e476–e483.
between healthy and disordered brain matter in patients with neuro- 87. Sobata E, Ohkuma H, Suzuki S. Cerebrovascular disorders associated with
fibromatosis type I uding diffusion tensor imaging. Am J Neuroradiol von Recklinghausen’s neurofibromatosis. Neurosurgery 1988;22:544–549.
2008;29:816–822. 88. Schievink WI, Piepgras DG. Cervical vertebral artery aneurysms and arte-
64. Sevick R, Barkovich A, Edwards M, Koch T, Berg B, Lempert T. Evolution riovenous fistulae in neurofibromatosis type I: case reports. Neurosurgery
of white matter lesions in neurofibromatosis type 1: MR findings. Am J 1991;29:760–765.
Roentengenol 1992;159:171–175. 89. Michels V, Whisnant J, Garrity J, Miller G. Neurofibromatosis type 1 with
65. Szudek J, Friedman JM. Unidentified bright objects associated with fea- bilateral acoustic neuromas. Neurofibromatosis 1989;2:213–217.
tures of neurofibromatosis 1. Pediatr Neurol 2002;27:123–127. 90. Sadeh M, Martinovits G, Goldhammer Y. Occurence of both neurofibro-
66. Gill DS, Hyman SL, Steinberg A, North KN. Age-related findings on MRI in matosis 1 and 2 in the same individual with a rapidly progressive course.
neurofibromatosis type 1. Pediatr Radiol 2006;36:1048–1056. Neurology 1989;39:282–283.
67. DiPaolo DP, Zimmerman RA, Rorke LB, Zackai EH, Bilaniuk LT, Yachnis 91. Editorial. Is NF-1 always distinct from NF-2? Neurofibromatosis 1989;2:
AT. Neurofibromatosis type 1: pathologic substrate of high-signal intensity 193–194.
foci in the brain. Radiology 1995;195:721–724. 92. Jacquemin C, Bosley T, Liu D, Svedberg H, Buhaliqa A. Reassessment of
68. Eastwood JD, Fiorella DJ, MacFall JF, Delong DM, Provenzale JM, sphenoid dysplasia associated with neurofibromatosis type 1. Am J Neuro-
Greenwood RS. Increased brain apparent diffusion coefficient in children radiol 2002;23:644–648.
with neurofibromatosis type 1. Radiology 2001;219:354–358. 93. Jacquemin C, Bosley T, Svedberg H. Orbit deformities in craniofacial neu-
69. Terada H, Barkovich AJ, Edwards MSB, Ciricillo SF. Evolution of rofibromatosis type 1. Am J Neuroradiol 2003;24:1678–1682.
high-intensity basal ganglia lesions on T1-weighted MR in neurofibroma- 94. Holt GR. von Recklinhausen’s neurofibromatosis. Otolaryngol Clin North
tosis type 1. Am J Neuroradiol 1996;17:755–760. Am 1987;20:179–193.
70. Sheikh SF, Kubal WS, Anderson AW, Mutalik P. Longitudinal evaluation of 95. Dombi E, Solomon J, Gillespie AJ, et al. NF1 plexiform neurofibroma
apparent diffusion coefficient in children with neurofibromatosis type 1. growth rate by volumetric MRI: relationship to age and body weight.
J Comput Assist Tomogr 2003;27:681–686. Neurology 2007;68:643–647.
71. Tognini G, Ferrozzi F, Garlaschi G, et al. Brain apparent diffusion coef- 96. Tucker T, Friedman JM, Friedrich RE, Wenzel R, Funsterer C, Mautner VF.
ficient evaluation in pediatric patients with neurofibromatosis type 1. Longitudinal study of neurofibromatosis 1 associated plexiform neurofi-
J Comput Assist Tomogr 2005;29:298–304. bromas. J Med Genet 2009;46:81–85.
72. Cha S, Knopp E, Johnson G, Wetzel S, Litt A, Zagzag D. Intracranial mass 97. Halliday A, Sobel R, Martuza R. Benign spinal nerve sheath tumors: their
lesions: dynamic contrast-enhanced susceptibility-weighted echo-planar occurrence sporadically and in neurofibromatosis types 1 and 2. J Neuro-
perfusion MR imaging. Radiology 2002;223:11–29. surg 1991;74:248–253.
73. Broniscer A, Gajjar A, Bhargava R, et al. Brain stem involvement in children 98. Burk DL, Brunberg JA, Kanal E, et al. Spinal and paraspinal neurofibroma-
with neurofibromatosis type 1: role of magnetic resonance imaging and tosis; surface coil MR imaging at 1.5 T. Radiology 1987;162:797–801.
spectroscopy in the distinction from diffuse pontine glioma. Neurosurgery 99. Suh J-S, Abenoza P, Galloway H, Everson L, Griffiths H. Peripheral
1997;40:331–337. (extracranial) nerve tumors: correlation of MR imaging and histologic
74. Girard N, Wang Z, Erbetta A, et al. Prognostic value of proton MR spec- findings. Radiology 1992;183:341–346.
troscopy of cerebral hemisphere tumors in children. Neuroradiology 100. Bhargava R, Parham DM, Lasater OE, CHari RS, Chen G, Fletcher BD. MR
1998;40:121–125. imaging differentiation of benign and malignant peripheral nerve sheath
75. Krouwer H, Kim T, Rand S, et al. Single-voxel proton MR spectroscopy tumors: use of the target sign. Pediatr Radiol 1997;27:124–129.
of nonneoplastic brain lesions suggestive of a neoplasm. Am J Neuroradiol 101. Smirniotopoulos J, Murphy F. The phakomatoses. Am J Neurorad 1992;
1998;19:1695–1703. 13:725–736.
76. Tzika AA, Vajapeyam S, Barnes PD. Multivoxel proton MR spectroscopy 102. Holt JF. Neuhauser lecture: neurofibromatosis in children. AJR 1978;130:
and hemodynamic MR imaging of childhood brain tumors: Preliminary 615–639.
observations. Am J Neuroradiol 1997;18:203–218. 103. Elefteriou F, Kolanczyk M, Schindeler A, et al. Skeletal abnormalities in
77. Bognanno JR, Edwards MK, Lee TA, et al. Cranial MR imaging in neurofi- neurofibromatosis type 1: approaches to therapeutic options. Am J Med
bromatosis. Am J Neuroradiol 1988;9:461–467. Genet A 2009;149A:2327–2338.

Barkovich_Chap06.indd 628 5/6/2011 9:04:49 PM


Chapter 6 • The Phakomatoses 629

104. Tsirikos AI, Ramachandran M, Lee J, Saifuddin A. Assessment of vertebral 127. Evans DG. Neurofibromatosis type 2 (NF2): a clinical and molecular
scalloping in neurofibromatosis type 1 with plain radiography and MRI. review. Orphanet J Rare Dis 2009;4:16.
Clin Radiol 2004;59:1009–1017. 128. Begnami MD, Rushing EJ, Santi M, Quezado M. Evaluation of NF2 gene
105. Lee M, Rezai A, Freed D, Epstein FJ. Intramedullary spinal cord tumors in deletion in pediatric meningiomas using chromogenic in situ hybridiza-
neurofibromatosis. Neurosurgery 1996;38:32–37. tion. Int J Surg Pathol 2007;15:110–115.
106. Barnes P, Brody J, Jaramillo D, Akbar J, Emans J. Atypical idiopathic sco- 129. Nunes F, MacCollin M. Neurofibromatosis 2 in the pediatric population.
liosis: MR imaging evaluation. Radiology 1993;186:247–253. J Child Neurol 2003;18:718–724.
107. Miles J, Pennybacker J, Sheldon P. Intrathoracic meningocele: its develop- 130. Ruggieri M, Iannetti P, Polizzi A, et al. Earliest clinical manifestations and
ment and association with neurofibromatosis. J Neurol Neurosurg Psychiatr natural history of neurofibromatosis type 2 (NF2) in childhood: a study
1969;32:99–110. of 24 patients. Neuropediatrics 2005;36:21–34.
108. Reis C, Carneiro E, Fonseca J, et al. Epitheloid hemangioendothelioma and 131. da Cruz MJ, Hardy DG, Moffat DA. Clinical presentation of a group of
multiple thoracolumbar lateral meningoceles: two rare pathological enti- NF2 patients to a tertiary referral skull base unit. Br J Neurosurg 2000;14:
ties in a patient with NF1. Neuroradiology 2005;47:165–169. 101–104.
109. Naidich T, McLone D, Harwood-Nash D. Arachnoid cysts, paravertebral 132. Mautner VF, Tatagiba M, Guthoff R, Samii M, Pulst ST. Neurofibromatosis
meningoceles, and perineural cysts. In: Newton T, Potts D, eds. Com- 2 in the pediatric age group. Neurosurgery 1993;33:92–96.
puted Tomography of the Spine and Spinal Cord. San Anselmo: Clavedel, 133. Evans D, Birch J, Ramsden R. Paediatric presentation of type 2 neurofibro-
1983:388–390. matosis. Arch Dis Child 1999;81:496–499.
110. Thakkar SD, Feigen U, Mautner V-F. Spinal tumors in neurofibromatosis 134. Ragge NK, Baser ME, Klein J, et al. Ocular abnormalities in neurofibroma-
type 1: an MRI study of frequency, multiplicity, and variety. Neuroradiol- tosis 2. Am J Ophthalmol 1995;120:634–641.
ogy 1999;41:625–629. 135. Baser M, Friedman J, Wallace A, Ramsden R, Joe H, Evans D. Evalua-
111. Pascual-Castroviejo I, Pascual-Castroviejo SI, Velazquez-Fragua R, Botella P, tion of clinical diagnostic criteria for neurofibromatosis 2. Neurology
Viano J. Familial spinal neurofibromatosis. Neuropediatrics 2007;38: 2002;59:1759–1765.
105–108. 136. Baser M, Friedman J, Aeschliman D, et al. Predictors of the risk of mortal-
112. Mautner V-F, Lindenau M, Baser ME, et al. The neuroimaging and clinical ity in neurofibromatosis 2. Am J Hum Genet 2002;71:715–723.
spectrum of neurofibromatosis 2. Neurosurgery 1996;38:880–886. 137. Parry DM, Eldridge R, Kaiser-Kupfer MI, Bouzas EA, Pikus A, Patronas
113. Khong P, Goh W, Wong V, Fung C, Ooi G. MR imaging of spinal tumors N. Neurofibromatosis 2 (NF2): clinical characteristics of 63 affected indi-
in children with neurofibromatosis I. AJR Am J Roentgenol 2003;180: viduals and clinical evidence for heterogeneity. Am J Med Genet 1994;52:
413–417. 450–461.
114. Kluwe L, Pulst S-M, Koppen J, Mautner V-F. A 163-bp deletion at the 138. Mautner VF, Nguyen R, Kutta H, et al. Bevacizumab induces regression of
C-terminus of schwannoma associated with variable phenotypers of neu- vestibular schwannomas in patients with neurofibromatosis type 2. Neuro
rofibromatosis type 2. Hum Genet 1995;95:443–446. Oncol 12:14–18.
115. Donner TR, Voorhies R, Kline D. Neural sheath tumors of major nerves. 139. Serradell A. Lesions centralees dans les neurofibromatoses: correlations
J Neurosurg 1994;81:362–373. cliniques d’IRM et histopathologiques. J Radiol 1991;72:635–644.
116. Upadhyaya M, Spurlock G, Kluwe L, et al. The spectrum of somatic and 140. Sze G, Abramson A, Krol G, et al. Gd-DTPA in the evaluation of intradural
germline NF1 mutations in NF1 patients with spinal neurofibromas. extramedullary spinal disease. Am J Neurorad 1989;9:153–163.
Neurogenetics 2009;10:251–263. 141. Valk J. Gd-DTPA in MR of spinal lesions. Am J Neurorad 1989;9:345–350.
117. Russell D, Rubenstein L. Dysgenetic syndromes (phakomatoses) asso- 142. Williams B. Progress in syringomyelia. Neuro Res 1986;8:130–145.
ciated with tumors and hamartomas of the nervous system. In: Russell 143. Sherman J, Barkovich A, Citrin C. The MR appearance of syringohydro-
D, Rubenstein L, eds. Pathology of Tumors of the Nervous System, 5th ed. myelia: new observations. Am J Neuroradiol 1986;7:985–995.
Baltimore: Williams & Wilkins, 1989:766–784. 144. Castillo M, Quencer R, Green B, Montalvo B. Syringomyelia as a conse-
118. Ferner RE, Golding JF, Smith M, et al. [18F]2-fluoro-2-deoxy-D-glucose quence of compressive extramedullary lesions. Am J Neurorad 1987;8:
positron emission tomography (FDG PET) as a diagnostic tool for neu- 973–978.
rofibromatosis 1 (NF1) associated malignant peripheral nerve sheath 145. Nagahiro S, Matsukado Y, Kuratsu J, et al. Syringomyelia and syringob-
tumours (MPNSTs): a long-term clinical study. Ann Oncol 2008;19: ulbia associated with an ependymoma of the cauda equina involving the
390–394. conus medullaris: case report. Neurosurgery 1986;18:357–360.
119. Matsumine A, Kusuzaki K, Nakamura T, et al. Differentiation between 146. Riccardi V. Neurofibromatosis: clinical heterogeneity. Curr Probl Cancer
neurofibromas and malignant peripheral nerve sheath tumors in neu- 1982;7:1–34.
rofibromatosis 1 evaluated by MRI. J Cancer Res Clin Oncol 2009;135: 147. Riccardi V. The phakomatoses. In: Myrianthopoulos N, ed. Malformations,
891–900. vol 6(50). Amsterdam: Elsevier, 1987:365–380.
120. Trofatter J, MacCollin M, Rutter J, et al. A novel moesin-, ezrin-, radixin- 148. Haines JL, Short MP, Kwiatkowski DJ, et al. Localization of one gene for
like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell tuberous sclerosis within 9q32–9q34, and further evidence for heteroge-
1993;72:791–800. neity. Am J Med Genet 1991;49:764–772.
121. Seizinger BR, Martuza RL, Gusella JF. Loss of genes on chromosome 22 in 149. Harris RM, Carter NP, Griffithe B, et al. Physical mapping within the
tumorogenesis of human acoustic neuroma. Nature 1986;322:664–667. tuberous sclerosis linkage group in region 9q32–q34. Genomics 1993;15:
122. Huson SM. The different forms of neurofibromatosis. Br Med J 1987; 265–274.
294:1113–1114. 150. Kandt RS, Haines JL, Smith M, et al. Linkage of an important gene locus
123. Stokowski R, Cox D. Functional analysis of the neurofibromatosis type 2 for tuberous sclerosis to a chromosome 16 marker for polycystic kidney
protein by means of disease-causing point mutations. Am J Hum Genet disease. Nat Genet 1992;2:37–41.
2000;66:873–891. 151. Consortium ECTS. Identification and characterization of the tuberous
124. Hanemann CO. Magic but treatable? Tumours due to loss of merlin. Brain sclerosis gene on chromosome 16. Cell 1993;75:1305–1315.
2008;131:606–615. 152. Kwiatkowski DJ. Tuberous sclerosis: from tubers to mTOR. Ann Hum
125. Patronas NJ, Courcoutsakis N, Bromley CM, Katzman GL, MacCollin M, Genet 2003;67:87–96.
Parry DM. Intramedullary and spinal canal tumors in patients with neu- 153. Jones A, Shyamsundar M, Thomas M, et al. Comprehensive mutation
rofibromatosis 2: MR imaging findings and correlation with genotype. analysis of TSC1 and TSC2-and phenotypic correlations in150 families
Radiology 2001;218:434–442. with tuberous sclerosis. Am J Hum Genet 1999;64:1305–1315.
126. Parry D, MacCollin M, Kaiser-Kupfer M, et al. Germ-line mutations in the 154. Dabora S, Jozwiak S, Franz D, et al. Mutational analysis in a cohort of 224
neurofibromatosis 2 gene: correlations with disease severity and retinal tuberous sclerosis patients indicates increased severity of TSC2, compared
abnormalities. Am J Hum Genet 1996;59:529–539. with TSC1, disease in multiple organs. Am J Hum Genet 2001;68:64–80.

Barkovich_Chap06.indd 629 5/6/2011 9:04:49 PM


630 Pediatric Neuroimaging

155. Napolioni V, Moavero R, Curatolo P. Recent advances in neurobiology of 183. Jansen FE, Vincken KL, Algra A, et al. Cognitive impairment in tuber-
tuberous sclerosis complex. Brain Dev 2009;31:104–113. ous sclerosis complex is a multifactorial condition. Neurology 2008;70:
156. van Siegtenhorst M, Nellist M, Nagelkerken B, Cheadle J, Snell R, van den 916–923.
Ouweland A. Interaction between hamartin and tuberin, the TSC1 and 184. Kingsley D, Kendall B, Fitz C. Tuberous sclerosis: a clinicoradiological
TSC2 gene products. Hum Mol Genet 1998;7:1053–1057. evaluation of 110 cases with particular reference to atypical presentation.
157. Johnson M, Emelin J, Park S-H, Vinters, HV. Co-localization of TSC1 and Neuroradiology 1986;28:171–190.
TSC2 gene products in tubers of patients with tuberous sclerosis. Brain 185. Menor F, Marti-Bonmati L, Mulas F, Poyatos C, Cortina H. Neuroimaging
Pathol 1999;9:45–54. in tuberous sclerosis: a clinicoradiological evaluation in pediatric patients.
158. Astrinidis A, Henske EP. Tuberous sclerosis complex: linking growth Pediatr Radiol 1992;22:485–489.
energy signaling pathways with human disease. Oncogene 2005;24: 186. Roach E, DiMario F, Kandt R, Northrup H. Tuberous sclerosis consen-
7475–7481. sus conference: recommendations for diagnostic testing. J Child Neurol
159. Kwiatkowski DJ, Manning BD. Tuberous sclerosis: a GAP at the crossroads of 1999;14:401–407.
multiple signaling pathways. Hum Mol Genet 2005;14(Suppl 2):R251–R258. 187. Goh S, Butler W, Thiele EA. Subependymal giant cell tumors in tuberous
160. Sampson J. TSC1 and TSC2: genes that are mutated in the human genetic sclerosis complex. Neurology 2004;63:1457–1461.
disorder tuberous sclerosis. Biochem Soc Trans 2003;31:592–596. 188. Webb DW, Clarke A, Fryer A, Osborne JP. The cutaneous features of tuber-
161. Goh S, Weiss WA. Genotype-phenotype correlations in tuberous sclerosis: ous sclerosis: a population study. Br J Dermatol 1996;135:1–5.
who and how to treat. Ann Neurol 2006;60:505–507. 189. Hurwitz S, Braverman IM. White spots in tuberous sclerosis. J Pediatr
162. Meikle L, Talos DM, Onda H, et al. A mouse model of tuberous sclero- 1970;77:587–594.
sis: neuronal loss of tsc1 causes dysplastic and ectopic neurons, reduced 190. Bell SD, MacDonald DM. The prevalence of café-au-lait patches in tuber-
myelination, seizure activity, and limited survival. J Neurosci 2007;27: ous sclerosis. Clin Exp Dermatol 1985;10:562–565.
5546–5558. 191. Mullaney PB, Jacquemin C, Abboud E, Karcioglu ZA. Tuberous sclerosis in
163. Sampson JR, Maheshwar MM, Aspinwall R, et al. Renal cystic disease in infancy. J Pediatr Ophthalmol Strabismus 1997;34:372–375.
tuberous sclerosis: role of the polycystic kidney disease 1 gene. Am J Hum 192. Zimmer-Galler IE, Robertson DM. Long-term observation of retinal
Genet 1997;61:843–851. lesions in tuberous sclerosis. Am J Ophthalmol 1995;119:318–324.
164. Webb DW, Osborne JP. Tuberous sclerosis. Arch Dis Child 1995;72: 193. Nyboer JH, Robertson DM, Gomez MR. Retinal lesions in tuberous scle-
471–474. rosis. Arch Opthalmol 1976;94:1277–1280.
165. Lewis JC, Thomas HV, Murphy KC, Sampson JR. Genotype and psycho- 194. Hedges T III, Pozzi-Mucelli R, Char D, Newton T. CT demonstration of
logical phenotype in tuberous sclerosis. J Med Genet 2004;41:203–207. ocular calcification: correlations with clinical and pathological findings.
166. Sparagana S, Roach E. Tuberous sclerosis complex. Curr Opin Neurol Neuroradiology 1982;23:15–21.
2000;13:115–119. 195. Jost BF, Olk RJ. Atypical retinitis proliferans, retinal telangiectasis, and
167. Rose V, Au K-S, Pollom G, Roach E, Prashner H, Northrup H. Germ- vitreous hemorrhage in a patient with tuberous sclerosis. Retina 1986;6:
line mosaicism in tuberous sclerosis: how common? Am J Hum Genet 53–56.
1999;64:986–992. 196. Christophe C, Bartholome J, Blum D, et al. Neonatal tuberous sclerosis.
168. Gomez M. Tuberous Sclerosis, 2nd ed. New York: Raven Press, 1988. US, CT, and MR diagnosis of brain and cardiac lesions. Pediatr Radiol
169. O’Callaghan FJ, Osborne JP. Advances in the understanding of tuberous 1989;19:446–448.
sclerosis. Arch Dis Child 2000;83:140–142. 197. Baron Y, Barkovich AJ. MR imaging of tuberous sclerosis in neonates and
170. Roach E, Gomez M, Northrup H. Tuberous sclerosis complex consensus young infants. Am J Neuroradiol 1999;20:907–916.
conference: revised clinical diagnostic criteria. J Child Neurol 1998;13: 198. Altman NR, Purser RK, Post MJD. Tuberous sclerosis: characteristics at
624–628. CT and MR imaging. Radiology 1988;167:527–532.
171. Donegani G, Grattarola FR, Wildi E. Tuberous sclerosis: Bourneville dis- 199. Martin N, De Brouker T, Cambier J, Marsault C, Nahum H. MRI evalua-
ease. In: Vinken PJ, Bruyn GW, eds. Handbook of Clinical Neurology: The tion of tuberous sclerosis. Neuroradiology 1987;29:437–443.
Phakomatoses, vol 14. Amsterdam: North Holland, 1972. 200. Martin N, Debussche C, de Broucker T, Mompoint D, Marsault C, Nahum
172. Bender BL, Yunis EJ. The pathology of tuberous sclerosis. Pathol Ann H. Gadolinium-DTPA enhanced MR imaging in tuberous sclerosis. Neu-
1982;17:339–382. roradiology 1990;31:492–497.
173. Ahlsen G, Gillberg IC, Lindblom R, Gillberg C. Tuberous sclerosis in west- 201. Wippold F II, Baber W, Gado M, Tobben P, Bartnicke B. Pre- and post-
ern Sweden. A population study of cases with early childhood onset. Arch contrast MR studies in tuberous sclerosis. J Comput Assist Tomogr
Neurol 1994;51:76–81. 1992;16:69–72.
174. Seidenwurm D, Barkovich A. Understanding tuberous sclerosis. Radiology 202. Karadag D, Mentzel H-J, Gullmar D, et al. Diffusion tensor imaging
1992;183:23–24. in children and adolescents with tuberous sclerosis. Pediatric Radiol
175. Stefansson K, Wollmann R, Huttenlocher P. Lineages of cells in the central 2005;35:980.
nervous system. In: Gomez M, ed. Tuberous Sclerosis, 2nd ed. New York: 203. Tsuchida T, Kamata K, Kwamata M, et al. Brain tumors in tuberous sclero-
Raven Press, 1988:75–87. sis. Child’s Brain 1981;8:271–283.
176. Yamanouchi H, Jay V, Rutka JT, Takashima S, Becker LE. Evidence of 204. Morimoto K, Mogami H. Sequential study of subependymal giant cell
abnormal differentiation in giant cells of tuberous sclerosis. Pediatr Neurol astrocytoma associated with tuberous sclerosis. J Neurosurg 1986;65:
1997;17:49–53. 874–877.
177. Braffman B, Bilaniuk L, Naidich T, et al. MR imaging of tuberous sclerosis: 205. Shepherd CW, Scheitharu B, Gomez MR, et al. Brain tumours in tuberous
pathogenesis of this phakomatosis, use of gadopentate dimeglumine, and sclerosis. A clinicopathological study of the Mayo Clinic experience. Ann
literature review. Radiology 1992;183:227–238. NY Acad Sci 1991;615:378–379.
178. Pampiglione P, Pugh F. Infantile spasms and subsequent development of 206. Mackay M, Becker L, Chuang S, et al. Malformations of cortical devel-
tuberous sclerosis syndrome. Lancet 1975;ii:1046. opment with balloon cells: clinical and radiologic correlates. Neurology
179. Fukushima K, Inoue Y, Fujiwara T, Yagi K. Long term course of West syn- 2003;60:580–587.
drome associated with tuberous sclerosis. Epilepsia 1998;39:51–54. 207. Griffiths PD, Martland TR. Tuberous sclerosis complex: the role of neuro-
180. Hanno R, Beck R. Tuberous sclerosis. Neurol Clin 1987;5:351–360. radiology. Neuropediatrics 1997;28:244–252.
181. Ohtsuka Y, Ohmorei I, Oka E. Long term follow-up of childhood epilepsy 208. Wilms G, van Wijck E, Demaerel P, Smet M-H, Plets C, Brucher J. Gyri-
associated with tuberous sclerosis. Epilepsia 1998;39:1158–1163. form calcifications in tuberous sclerosis simulating the appearance of
182. Joinson C, O’Callaghan FJ, Osborne JP, Martyn C, Harris T, Bolton PF. Sturge-Weber disease. Am J Neuroradiol 1992;13:295–297.
Learning disability and epilepsy in an epidemiological sample of individu- 209. Simon EM, Goldstein RB, Coakley FV, et al. Fast MR Imaging of Fetal CNS
als with tuberous sclerosis complex. Psychol Med 2003;33:335–344. Anomalies in Utero. Am J Neuroradiol 2000;21:1688–1698.

Barkovich_Chap06.indd 630 5/6/2011 9:04:49 PM


Chapter 6 • The Phakomatoses 631

210. Levine D, Barnes P, Korf B, Edelman R. Tuberous sclerosis in the fetus: 233. Lagos J, Gomez M. Tuberous sclerosis: reappraisal of a clinical entity. Mayo
second trimester diagnosis of subependymal tubers with ultrafast MR Clin Proc 1967;42:26–49.
imaging. AJR Am J Roentgenol 2000;175:1067–1069. 234. Eluvathingal TJ, Behen ME, Chugani HT, et al. Cerebellar lesions in tuber-
211. Girard N, Zimmerman RA, Schnur R, Haselgrove J, Christensen K. Mag- ous sclerosis complex: neurobehavioral and neuroimaging correlates.
netization transfer in the investigation of patients with tuberous sclerosis. J Child Neurol 2006;21:846–851.
Neuroradiology 1997;39:523–528. 235. Lie JT. Cardiac, pulmonary, and vascular involvements in tuberous sclero-
212. Jeong M-G, Chung T-S, Coe C-J, Jeon T-J, Kim D-I, Joo A-Y. Applica- sis. Ann N Y Acad Sci 1991;615:58–70.
tion of magnetization transfer imaging for intracranial lesions of tuberous 236. Smulewicz JJ, Tafreshi M. Angiographic changes in tuberous sclerosis.
sclerosis. J Comput Assist Tomogr 1997;21:8–14. Angiology 1977;28:300–322.
213. Pinto Gama H, da Rocha AN, Braga FV, et al. Comparative analysis of MR 237. Calcagni G, Gesualdo F, Tamisier D, Brunelle F, Sidi D, Ou P. Arterial aneu-
sequences to detect structural brain lesions in tuberous sclerosis. Pediatr rysms and tuberous a classic but little known association. Pediatr Radiol
Radiol 2006;36:119. 2008;38:795–797.
214. Jurkiewicz E, Jozwiak S, Bekiesinska-Figatowska M, Pakula-Kosciesza I, Wal- 238. Spangler WJ, Cosgrove GR, Moumdjian RA, Montes JL. Cerebral arterial
ecki J. Cyst-like cortical tubers in patients with tuberous sclerosis complex: ectasia and tuberous sclerosis. Neurosurgery 1997;40:191–194.
MR imaging with the FLAIR sequence. Pediatr Radiol 2006;36:498–501. 239. Jones B, Tomsick T, Franz D. Guglielmi detachable coil embolization of a
215. Chu-Shore CJ, Major P, Montenegro M, Thiele E. Cyst-like tubers are asso- giant midbasilar aneurysm in a 19-month-old patient. Am J Neuroradiol
ciated with TSC2 and epilepsy in tuberous sclerosis complex. Neurology 2002;23:1145–1148.
2009;72:1165–1169. 240. Chonko AM, Weiss SM, Stein JH, et al. Renal involvement in tuberous
216. Henkelman M, Watts J, Kucharczyk W. High signal intensity in MR images sclerosis. Am J Med 1974;56:124–132.
of calcified brain tissue. Radiology 1991;179:199–206. 241. Madlom M, Hoggard N, Griffiths P, Chan J. Facial naevus flammeus with
217. Pollock JM, Whitlow CT, Tan H, Kraft RA, Burdette JH, Maldjian JA. choroidal haemangioma and without intracranial involvement. Dev Med
Pulsed arterial spin-labeled MR imaging evaluation of tuberous sclerosis. Child Neurol 2003;45:139.
Am J Neuroradiol 2009;30:815–820. 242. Sen Y, Dilber E, Odemis E, Ahmetoglu A, Aynaci F. Sturge-Weber syn-
218. Li LM, Cendes F, Bastos AC, Andermann F, Dubeau F, Arnold DL. Neu- drome in a 14-year-old girl without facial naevus. Eur J Pediatr 2002;161:
ronal metabolic dysfunction in patients with cortical developmental mal- 505–506.
formations. A proton magnetic resonance spectroscopic imaging study. 243. Alexander GL. Sturge-Weber syndrome. In: Vinken PJ, Bryun GW, eds.
Neurology 1998;50:755–759. Handbook of Clinical Neurology: The Phakomatoses, vol 14. Amsterdam:
219. Mukonoweshuro W, Wilkinson ID, Griffiths PD. Proton MR spectroscopy North Holland, 1972:223–240.
of cortical tubers in adults with tuberous sclerosis complex. Am J Neurora- 244. Gorman M, Snead O. Sturge-Weber syndrome without portwine nevus.
diol 2001;22:1920–1925. Pediatrics 1977;60:785–786.
220. Tzika AA, Vigneron DB, Dunn RS, Nelson SJ, Ball WSJ. Intracranial tumors 245. Roach E. Diagnosis and management of neurocutaneous syndromes.
in children: small single-voxel proton MR spectroscopy using short- and Semin Neurol 1988;8:83–96.
long-echo sequences. Neuroradiology 1996;38:254–263. 246. Gururaj A, Sztriha L, Johansen J, Nork M. Sturge-Weber syndrome with-
221. Karenfort M, Kruse B, Freitag H, Pannek H, Tuxhorn I. Epilepsy surgery out facial nevus: a case report and review of the literature. Acta Paediatr
outcome in children with focal epilepsy due to tuberous sclerosis complex. 2000;89:740–743.
Neuropediatrics 2002;33:255–261. 247. Pascual-Castroviejo I, Diaz-Gonzalez C, Garcia-Melian R, Gonzalez-Casado
222. Koh S, Jayakar P, Dunoyer C, et al. Epilepsy surgery in children with tuber- I, Muñoz-Hiraldo E. Sturge-Weber syndrome: study of 40 patients. Pediatr
ous sclerosis complex: presurgical evaluation and outcome. Epilepsia Neurol 1993;9:283–288.
2000;41:1206–1213. 248. Streeter G. The developmental alterations in the vascular system of the
223. Luat AF, Makki M, Chugani HT. Neuroimaging in tuberous sclerosis com- brain of the human embryo. Contr Embryol Carneg Inst 1918;8:5–38.
plex. Curr Opin Neurol 2007;20:142–150. 249. Wohlwill F, Yakovlev PI. Histopathology of meningo-facial angioma-
224. Chandra PS, Salamon N, Huang J, et al. FDG-PET/MRI coregistration and tosis (weber disease): report of four cases. J Neuropathol Exp Neurol
diffusion-tensor imaging distinguish epileptogenic tubers and cortex in 1957;16:341–364.
patients with tuberous sclerosis complex: a preliminary report. Epilepsia 250. Gardeur D, Palmieri A, Mashaly R. Cranial computed tomography in the
2006;47:1543–1549. phakomatoses. Neuroradiology 1983;25:293–304.
225. Wu JY, Sutherling WW, Koh S, et al. Magnetic source imaging localizes 251. Sperner J, Schmauser I, Bittner R, et al. MR imaging findings in children
epileptogenic zone in children with tuberous sclerosis complex. Neurology with Sturge-Weber syndrome. Neuropediatrics 1990;21:146–152.
2006;66:1270–1272. 252. Benedikt R, Brown D, Walker R, Ghaed V, Mitchell M, Geyer C. Sturge-
226. Gama HP, da Rocha AJ, Valerio RM, da Silva CJ, Garcia LA. Hippocampal Weber syndrome: cranial MR imaging with Gd-DTPA. Am J Neuroradiol
Abnormalities in an MR Imaging Series of Patients with Tuberous Sclero- 1993;14:409–415.
sis. Am J Neuroradiol 2010;31(6):1059–1062. 253. Griffiths PD, Coley SC, Romanowski CA, Hodgson T, Wilkinson ID. Con-
227. Garaci FG, Floris R, Bozzao A, et al. Increased brain apparrent diffusion trast-enhanced fluid-attenuated inversion recovery imaging for leptomen-
coefficient in tuberous sclerosis. Radiology 2004;232:461–465. ingeal disease in children. Am J Neuroradiol 2003;24:719–723.
228. Ioannidou MC, Tzoufi M, Astrakas L, Argyroupoulou MI. Magnetization 254. Griffiths PD, Boodram MB, Blaser S, Armstrong D, Gilday DL, Harwood-
transfer ratio measurements of the brain in children with tuberous sclero- Nash DC. 99m-Technetium HMPAO imaging in children with the Sturge-
sis complex. Pediatr Radiol 2005;35:1071–1074. Wever syndrome: a study of nine cases with CT and MRI correlation.
229. Makki MI, Chugani DC, Janisse J, Chugani HT. Characteristics of abnor- Neuroradiology 1997;39:219–224.
mal diffusivity in normal-appearing white matter investigated with diffu- 255. Juhasz C, Haacke EM, Hu J, et al. Multimodality imaging of cortical and
sion tensor MR imaging in tuberous sclerosis complex. Am J Neuroradiol white matter abnormalities in Sturge-Weber syndrome. Am J Neuroradiol
2007;28:1662–1667. 2007;28:900–906.
230. Arulrajah S, Ertan G, Jordan L, et al. Magnetic resonance imaging and 256. Juhasz C, Haacke EM, Hu J, et al. Multimodality imaging of cortical and
diffusion-weighted imaging of normal-appearing white matter in chil- white matter abnormalities in Sturge-Weber syndrome. Am J Neuroradiol
dren and young adults with tuberous sclerosis complex. Neuroradiology 2007;28:900–906.
2009;51:781–786. 257. Fischbein NJ, Barkovich AJ, Wu Y, Berg BO. Sturge-Weber syndrome
231. Van Tassel P, Curé J, Holden KR. Cystlike white matter lesions in tuberous with no leptomeningeal enhancement on MRI. Neuroradiology 1998;40:
sclerosis. Am J Neuroradiol 1997;18:1367–1373. 177–180.
232. Griffiths P, Bolton P, Verity C. White matter abnormalities in tuberous 258. Coulam CM, Brown LR, Reese DF. Sturge-Weber syndrome. Semin Roent-
sclerosis complex. Acta Radiol 1998;39:482–486. genol 1976;11:55–59.

Barkovich_Chap06.indd 631 5/6/2011 9:04:49 PM


632 Pediatric Neuroimaging

259. Hu J, Yu Y, Juhasz C, et al. MR susceptibility weighted imaging (SWI) 283. Choyke PL, Glenn GM, Walther MM, Patronas NJ, Linehan WM, Zbar
complements conventional contrast enhanced T1 weighted MRI in char- B. von Hippel-Lindau disease: genetic, clinical, and imaging features.
acterizing brain abnormalities of Sturge-Weber Syndrome. J Magn Reson Radiology 1995;194:629–642.
Imaging 2008;28:300–307. 284. Lindau A. Studien über Kleinhirncysten. Bau, Pathogenese und Beziehun-
260. Lin DD, Barker PB, Hatfield LA, Comi AM. Dynamic MR perfusion and gen zur Angiomatosis retinae. Acta Pathologica et Microbiologica Scandi-
proton MR spectroscopic imaging in Sturge-Weber syndrome: correlation navica 1926;(Suppl 1):1–128.
with neurological symptoms. J Magn Reson Imaging 2006;24:274–281. 285. von Hippel E. Ueber eine sehr seltene Erkrankung der Netzhaut. Albrecht
261. Evans AL, Widjaja E, Connolly DJA, Griffiths PD. Cerebral perfu- von Graefes Archiv für Ophthalmologie 1904;59:83–106.
sion abnormalities in children with Sturge-Weber syndrome shown by 286. Horton WA, Wong V, Eldridge R. Von Hippel-Lindau disease. Arch Int Med
dynamic contrast bolus magnetic resonance perfusion imaging. Pediatrics 1976;136:769–777.
2006;117:2119–2125. 287. Latif F, Tory K, Gnarra J, et al. Identification of the von Hippel-Lindau
262. Batista CE, Chugani HT, Hu J, et al. Magnetic resonance spectroscopic disease tumor suppressor gene. Science 1993;260:1317–1320.
imaging detects abnormalities in normal-appearing frontal lobe of 288. Farrell CJ, Plotkin SR. Genetic causes of brain tumors: neurofibromatosis,
patients with Sturge-Weber syndrome. J Neuroimaging 2008;18:306–313. tuberous sclerosis, von Hippel-Lindau, and other syndromes. Neurol Clin
263. Arulrajah S, Ertan G, Comi AM, Tekes A, Lin DL, Huisman TA. MRI with 2007;25:925–946, viii.
diffusion-weighted imaging in children and young adults with simultane- 289. Grossman M, Melmon KL. Von Hippel-Lindau disease. In: Vinken PJ,
ous supra- and infratentorial manifestations of Sturge-Weber syndrome. Bryun GW, eds. Handbook of Clinical Neurology: The Phakomatoses,
J Neuroradiol 2010;37:51–59. vol 14. 1972:241–259.
264. Stimac GK, Solomon MA, Newton TH. CT and MR of angiomatous mal- 290. Mukherji SK, Albernaz VS, Lo WWM, et al. Papillary endolymphatic sac
formations of the choroid plexus in patients with Sturge-Weber disease. tumors: CT, MR imaging, and angiographic findings in 20 patients. Radi-
Am J Neuroradiol 1986;7:623–627. ology 1997;202:801–808.
265. Griffiths PD, Blaser S, Boodram MB, Armstrong D, Harwood-Nash DC. 291. Huson SM, Harper PS, Hourihan MD, et al. Cerebellar hemangioblastoma
Choroid plexus size in young children with Sturge-Weber syndrome. Am J and von Hippel-Lindau disease. Brain 1986;109:1297–1310.
Neuroradiol 1996;17:175–180. 292. Wanebo J, Lonser R, Glenn G, Oldfield E. The natural history of heman-
266. Jacoby C, Yuh W, Afifi A, Bell W, Schelper R, Sato Y. Accelerated myeli- gioblastomas of the central nervous system in patients with von Hippel-
nation in early Sturge-Weber syndrome demonstrated by MR imaging. Lindau disease. J Neurosurg 2003;98:82–94.
J Comput Assist Tomogr 1987;11:226–231. 293. Slater A, Moore N, Huson S. The natural history of cerebellar heman-
267. Moritani T, Kim J, Sato Y, Bonthius D, Smoker WR. Abnormal hypermy- gioblastomas in von Hippel-Lindau disease. Am J Neuroradiol 2003;24:
elination in a neonate with Sturge-Weber syndrome demonstrated on 1570–1574.
diffusion-tensor imaging. J Magn Reson Imaging 2008;27:617–620. 294. Ammerman JM, Lonser RR, Dambrosia J, Butman JA, Oldfield EH.
268. Demerens C, Stankoff B, Logak M, et al. Introduction of myelination in Long-term natural history of hemangioblastomas in patients with
the central nervous system by electrical activity. Proc Natl Acad Sci USA von Hippel-Lindau disease: implications for treatment. J Neurosurg
1996;93:9887–9892. 2006;105:248–255.
269. Bentson JR, Wilson GH, Newton TH. Cerebral venous drainage pattern of 295. Jagannathan J, Lonser RR, Smith R, DeVroom HL, Oldfield EH. Surgi-
the Sturge-Weber syndrome. Radiology 1971;102:111–118. cal management of cerebellar hemangioblastomas in patients with von
270. Kendall B, Kingsley D. The value of CAT in craniocerebral malformations. Hippel-Lindau disease. J Neurosurg 2008;108:210–222.
Br J Radiol 1978;51:171–190. 296. Fill WL, Lamiell JM, Polk NO. The radiographic manifestations of von
271. Bilaniuk LT, Zimmerman RA, Hochman M, et al. MR of the Sturge-Weber Hippel-Lindau disease. Radiology 1979;133:289–295.
syndrome. Am J Neuroradiol 1987;8:945–950. 297. Ho V, Smirniotopoulos J, Murphy F, Rushing E. Radiologic-Pathologic
272. Adams ME, Aylett SE, Squier W, Chong W. A spectrum of unusual neu- correlation: hemangioblastoma. Am J Neuroradiol 1992;13:1343–1352.
roimaging findings in patients with suspected Sturge-Weber syndrome. 298. Sato Y, Waziri M, Smith W, et al. Hippel-Lindau disease: MR imaging.
Am J Neuroradiol 2009;30:276–281. Radiology 1988;166:241–246.
273. Enzmann DR, Hayward RW, Norman D, Dunn RP. Cranial computed 299. Leung RS, Biswas SV, Duncan M, Rankin S. Imaging features of von
tomographic scan appearance of Sturge-Weber disease: unusual presenta- Hippel-Lindau disease. Radiographics 2008;28:65–79.
tion. Radiology 1977;122:721–724. 300. Lee D, Sanches J, Mark A, Dillon W, Norman D, Newton T. Posterior fossa
274. Griffiths PD. Sturge-Weber syndrome revisited: the role of neuroimaging. hemangioblastomas: MR imaging. Radiology 1989;171:463–468.
Neuropediatrics 1996;27:284–294. 301. Wolpert SM. The neuroradiology of hemangioblastomas of the cerebel-
275. Probst FP. Vascular morphology and angiographic flow patterns in Sturge- lum. Am J Roentgenol 1970;110:56–66.
Weber angiomatosis: facts, thoughts and suggestions. Neuroradiology 302. Bydder G, Brown J, Niendorf H, Young I. Enhancement of cervical
1980;20:73–78. intraspinal tumors in MR imaging with IV Gd-DTPA. J Comput Assist
276. Chugani H, Mazziota J, Phelps M. Sturge-Weber syndrome: a study of Tomogr 1985;9:847–851.
cerebral glucose utilization with positron emission tomography. J Pediatr 303. Megerian CA, McKenna MJ, Nuss RC, et al. Endolymphatic sac tumors:
1989;114:244–253. histopathologic confirmation, clinical characterization, and implication
277. Peterman A, Hayles A, Dockerty M, Love J. Encephalotrigeminal angioma- in von Hippel-Lindau disease. Laryngoscope 1995;105:801–808.
tosis (Sturge-Weber Disease). J Am Med Assoc 1958;167:2169–2176. 304. Kempermann G, Neumann HPH, Scheremet R, et al. Deafness due to
278. Griffiths PD, Boodram MB, Blaser S, et al. Abnormal ocular enhancement in bilateral endolymphatic sac tumors in a case of von Hippel-Lindau syn-
Sturge-Weber syndrome: correlation of ocular, MR and CT findings with clin- drome. J Neurol Neurosurg Psychiatry 1996;16:318–320.
ical and intracranial imaging findings. Am J Neuroradiol 1996;17:749–754. 305. Lonser RR, Baggenstos M, Kim HJ, Butman JA, Vortmeyer AO. The ves-
279. Venes J, Linder S. Sturge-Weber-Dimitri syndrome. In: Edwards M, tibular aqueduct: site of origin of endolymphatic sac tumors. J Neurosurg
Hoffman H, eds. Cerebral Vascular Disease in Children and Adolescents. 2008;108:751–756.
Baltimore: Williams & Wilkins, 1988:337–341. 306. Lo WWM, Applegate LJ, Carberry JN, et al. Endolymphatic sac tumors:
280. Williams D III, Elster A. Cranial CT and MR in Klippel-Trenaunay-Weber radiologic diagnosis. Radiology 1993;189:199–204.
syndrome. Am J Neuroradiol 1992;13:291–294. 307. Paller AS. Ataxia-Telangiectasia. Neurol Clins 1987;5:447–449.
281. Anlar B, Yalaz K, Erzen C. Klippel-Trenaunay-Weber syndrome: a case 308. Sedgwick RP, Boder E. Ataxia-Telangiectasia. In: Vinken PJ, Bryun GW,
with cerebral and cerebellar atrophy. Neuroradiology 1988;30:360. eds. Handbook of Clinical Neurology: The Phakomatosis, vol 14. Amster-
282. Kiley M, Oxbury J, Coley S. Intracranial hypertension in Sturge-Weber/ dam: North Holland, 1972:267–334.
Klippel-Trenaunay-Weber overlap syndrome due to impairment of cere- 309. Hosking G. Ataxia-Telangiectasia. Develop Med Child Neurol 1982;24:
bral venous outflow. J Clin Neurosci 2002;9:330–333. 77–80.

Barkovich_Chap06.indd 632 5/6/2011 9:04:49 PM


Chapter 6 • The Phakomatoses 633

310. Heintz N. Ataxia telangiectasia: cell signaling, cell death, and the cell cycle. 334. Lovett A, Maari C, Decarie JC, et al. Large congenital melanocytic nevi
Curr Opin Neurol 1996;9:137–140. and neurocutaneous melanocytosis: one pediatric center’s experience.
311. Meyn SM. Ataxia-telengiectasia, cancer, and the pathobiology of the ATM J Am Acad Dermatol 2009;61:766–774.
gene. Clin Genet 1999;55. 335. Kadonaga D, Barkovich A, Edwards M, Frieden I. Neurocutaneous mel-
312. Savitsky K, Bar-Shira A, Gilad S, et al. A single ataxia-telangiectasia gene anosis in association with the Dandy-Walker malformation. Pediatr
with a product similar to PI-3 kinase. Science 1995;268:1749–1753. Dermatol 1992;9:37–43.
313. Kieslich M, Hoche F, Reichenbach J, et al. Extracerebellar MRI-lesions in 336. Bittencourt F, Marghoob A, Kopf A, Koenig K, Bart R. Large congenital
ataxia telangiectasia go along with deficiency of the GH/IGF-1 axis, mark- melanocytic nevi and the risk for development of malignant melanoma
edly reduced body weight, high ataxia scores and advanced age. Cerebel- and neurocutaneous melanocytosis. Pediatrics 2000;106:736–741.
lum 2009;9:190–197. 337. Frieden IJ, Williams ML, Barkovich AJ. Giant congenital melanocytic nevi:
314. Frizzera G, Rosai J, Dehner L, et al. Lymphoreticular disorders in primary brain magnetic resonance findings in neurologically asymptomatic chil-
immunodeficiencies: new findings based on up-to-date histologic classifi- dren. J Am Acad Dermatol 1994;31:423–429.
cation of 35 cases. Cancer 1980;46:692–699. 338. Foster RD, Williams ML, Barkovich AJ, Hoffman WY, Mathes SJ, Frieden
315. Boder E. Ataxia-telangiectasia: an overview. In: Gatti RA, Swift M, IJ. Giant congenital melanocytic nevi: the significance of neurocutaneous
eds. Ataxia-Telangiectasia: Genetics, Neuropathology, and Immunology melanosis in neurologically asymptomatic children. Plast Reconstr Surg
of a Degenerative Disease of Childhood. New York: Allen R. Liss, Inc., 2001;107:933–941.
1985:1–63. 339. Kadonaga J, Frieden I. Neurocutaneous melanosis: definition and review
316. Dar I, Biton S, Shiloh Y, Barzilai A. Analysis of the ataxia telangiectasia of the literature. Acad Dermatol 1991;24:747–755.
mutated-mediated dna damage response in murine cerebellar neurons. 340. Le Douarin N. The Neural Crest. Cambridge: Cambridge University Press,
J Neurosci 2006;26:7767–7774. 1982.
317. Kamiya M, Yamanouchi H, Yoshida T, et al. Ataxia telangiectasia with vas- 341. O’Rahilly R, Muller F. The meninges in human development. J Neuropath
cular abnormalities in the brain parenchyma: report of an autopsy case Exp Neurol 1986;45:588–608.
and literature review. Pathol Int 2001;51:271–276. 342. Yu H, Tsaur K, Chein C, et al. Neurocutaneous melanosis: electron micro-
318. Sardanelli F, Pittiglio G, Mavilio N, et al. Sindrome di Louis-Bar, atassia- scopic comparison of the pigmented melanocytic nevi of skin and menin-
teleangiectasia: quadro clinica e tomografico MR in 4 casi. Acta Neurora- geal melanosis. J Dermatol 1985;12:267–276.
diologia 1991;5:261–264. 343. Stern C, Artinger K, Bronner-Fraser M. Tissue interactions affecting the
319. Tavani F, Zimmerman RA, Berry GT, Sullivan K, Gatti R, Bingham P. Atax- migration and differentiation of neural crest cells in the chick embryo.
ia-telangiectasia: the pattern of cerebellar atrophy on MRI. Neuroradiology Development 1991;113:207–216.
2003;45:315–319. 344. Kitamuira K, Takiguchi-Hayashi K, Sezaki M, Yamamoto H, Takeuchi T.
320. Ciemins JJ, Horowitz AL. Abnormal white matter signal in ataxia telangi- Avian neural crest cells express a melanogenic trait during early migration
ectasia. Am J Neuroradiol 2000;21:1483–1485. from the neural tube: observations with the new monoclonal antibody,
321. Wallis LI, Griffiths P, Ritchie SJ, Romanowski CAJ, Darwent G, Wilkinson “MEBL-1”. Development 1992;114:367–378.
ID. Proton spectroscopy and imaging at 3T in ataxia-telangiectasia. Am J 345. Smith AB, Rushing EJ, Smirniotopoulos JG. Pigmented lesions of the
Neuroradiol 2007;28:79–83. central nervous system: radiologic-pathologic correlation. Radiographics
322. Lin DD, Crawford TO, Lederman HM, Barker PB. Proton MR spectro- 2009;29:1503–1524.
scopic imaging in ataxia-telangiectasia. Neuropediatrics 2006;37:241–246. 346. Fox H, Emery J, Goodbody R, Yates P. Neuro-cutaneous melanosis. Arch
323. Shiloh Y. Ataxia-telangiectasia and the Nijmegen breakage syndrome: Dis Child 1964;39:508–516.
related disorders but genes apart. Annu Rev Genet 1997;31:635–662. 347. Fischer S. Primary perivascular cerebral, cerebellar and leptomeningeal
324. Saar K, Chrzanowska K, Stumm M, et al. The gene for the ataxia- melanoma. Acta Psychiatr Scand 1956;31:21–34.
telangiectasia variant, Nijmegen breakage syndrome, maps to a - cM inter- 348. Adeyanju M, Reyes M, Junaid A. Fatal pneumococcal infection in neuro-
val on chromosome 8q21. Am J Hum Genet 1997;60:605–610. cutaneous melanosis. J Child Neurol 1988;3:293–294.
325. Carney J, Maser R, Olivares H, et al. The hMre11/hRad50 progein complex 349. Lamas E, Diez Lobato R, Lotelo T, Ricoy J, Castro S. Neurocutaneous
and Nijmegen breakage syndrome: linkage of double-strand break repair melanosis: report of a case and review of the literature. Acta Neurochir
to the cellular DNA damage response. Cell 1998;93:477–486. 1977;36:93–105.
326. Varon R, Vissinga C, Platzer M, et al. Nibrin, a novel DNA double-strand 350. Thomas C, Toone B, Rose P. Neurocutaneous melanosis and psychosis. Am
break repair protein, is mutated in Nijmegen breakage syndrome. Cell J Psychiatr 1988;145:649–650.
1998;93:467–476. 351. Sebag G, Dubois J, Pfister P, Brunelle F, St-Rose C. Neurocutaneous mel-
327. Weemaes C, Hustinx T, Scheres J, van Munster P, Bakkeren J, Taalman R. anosis and temporal lobe tumor in a child: MR study. Am J Neuroradiol
A new chromosomal instability disorder: the Nijmegen breakage syn- 1991;12:699–700.
drome. Acta Pediatr Scand 1981;70:557–564. 352. Barkovich A, Frieden I, Williams M. MR of neurocutaneous melanosis.
328. Chrzanowska K, Kleijer W, Krajewska-Walasek M, et al. Eleven Pol- Am J Neuroradiol 1994;15:859–867.
ish patients with microcephaly, immunodeficiency, and chromosomal 353. Hayashi M, Maeda M, Maji T, Matsubara T, Tsukahara H, Takeda K. Diffuse
instability: the Nijmegen breakage syndrome. Am J Med Genet 1995;57: leptomeningeal hyperintensity on fluid-attenuated inversion recovery MR
462–471. images in neurocutaneous melanosis. Am J Neuroradiol 2004;25:138–141.
329. Bekiesinska-Figatowska M, Chrzanowska K, Sidorska J, et al. Cranial MRI 354. Ko S-F, Wang H-S, Lui T-N, Ng S-H, Ho Y-S, Tsai C-C. Neurocutane-
in the Nijmegen breakage syndrome. Neuroradiology 2000;42:43–47. ous melanosis associated with inferior vermian hypoplasia: MR findings.
330. Rokitansky J. Ein ausgezeichneter Fall von Pigment-Mal mit ausgebreit- J Comput Assist Tomogr 1993;17:691–695.
eter Pigmentierung der inneren Hirn- und Rüchenmarkshaute. Allg Wien 355. Sealy R. Radicals in melanin biochemistry. Methods Enzymol 1984;105:
Med Z 1861;6:113–116. 479–483.
331. Pavlidou E, Hagel C, Papavasilliou A, Giouroukos S, Panteliadis C. Neuro- 356. Jimbow K, Miyake Y, Homma K, et al. Characterization of melanogen-
cutaneous melanosis: report of three cases and up-to-date review. J Child esis and morphogenesis of melanosomes by physiochemical proper-
Neurol 2008;23:1382–1391. ties of melanin and melanosomes in malignant melanoma. Cancer Res
332. Fox H. Neurocutaneous melanosis. In: Vincken P, Bruyn G, eds. The Pha- 1984;44:1128–1134.
komatoses, vol 14. New York: Elsevier, 1972:414–428. 357. Gomori J, Grossman R, Shields J, Augsburger J, Joseph P, DeSimeone D.
333. Takayama H, Nagashima Y, Hara M, et al. Immunohistochemical detec- Choroidal melanomas: correlation of NMR spectroscopy and MR imag-
tion of the c-met proto-oncogene product in the congenital melanocytic ing. Radiology 1986;158:443–445.
nevus of an infant with neurocutaneous melanosis. J Am Acad Dermatol 358. Burton D, Forsen S, Karlstrom G, Dwek R. Proton relaxation enhancement
2001;44:538–540. in biochemistry: a critical survey. Prog NMR Spectrosc 1979;13:1–45.

Barkovich_Chap06.indd 633 5/6/2011 9:04:49 PM


634 Pediatric Neuroimaging

359. Rhodes R, Rriedman H, Hatter HJ, Hockenberger B, Oakes W, Tomita T. 385. Pascual-Castroviejo I, Roche C, Martinez-Bermejo A, et al. Hypomelano-
Contrast-enhanced MR imaging of neurocutaneous melanosis. Am J Neu- sis of Ito. A study of 76 infantile cases. Brain Dev 1998;20:36–43.
roradiol 1991;12:380–382. 386. Assogba K, Ferlazzo E, Striano P, et al. Heterogeneous seizure manifesta-
360. Byrd SE, Darling CF, Tomita T, Chou P, de Leon G, Radkowski MA. MR tions in Hypomelanosis of Ito: report of four new cases and review of the
imaging of symptomatic neurocutaneous melanosis in children. Pediatr literature. Neurol Sci 2010;31:9–16.
Radiol 1997;27:39–44. 387. Fujino O, Hashimoto K, Fujita T, et al. Clinico-neuropathological study
361. Berker M, Oruckaptan H, Oge H, Benli K. Neurocutaneous melanosis of incontinentia pigmenti achromians – an autopsy case. Brain Dev
associated with Dandy-Walker malformation: case report and review of 1995;17:425–427.
the literature. Pediatr Neurosurg 2000;33:270–273. 388. Ross DL, Liwnicz BH, Chun RWM, Gilbert E. Hypomelanosis of Ito
362. Schreml S, Gruendobler B, Schreml J, et al. Neurocutaneous melanosis in (incontinentia pigmenti achromaians) –a clinico-pathologic study: mac-
association with Dandy-Walker malformation: case report and literature rocephaly and gray matter heterotopias. Neurology 1982;32:1013–1016.
review. Clin Exp Dermatol 2008;33:611–614. 389. Steiner J, Adamsbaum C, Desguerres I, et al. Hypomelanosis of Ito and
363. Marnet D, Vinchon M, Mostofi K, Catteau B, Kerdraon O, Dhellemmes P. brain abnormalities: MRI findings and literature review. Pediatr Radiol
Neurocutaneous melanosis and the Dandy-Walker complex: an uncom- 1996;26:763–768.
mon but not so insignificant association. Childs Nerv Syst 2009;25: 390. Peserico A, Battistella PA, Bertoli P, Drigo P. Unilateral hypomelanosis of
1533–1539. Ito with hemimegalencephaly. Acta Paediatr Scand 1988;77:446–447.
364. Pascual-Castroviejo I, Roche MC, Fernandez VM, et al. Incontinen- 391. Placantonakis DG, Ney G, Edgar M, Souweidane M, Hosain S, Schwartz
tia pigmenti: MR demonstration of brain changes. Am J Neuroradiol TH. Neurosurgical management of medically intractable epilepsy associ-
1994;15:1521–1527. ated with hypomelanosis of Ito. Epilepsia 2005;46:329–331.
365. Aydingöz Ü, Midia M. Central nervous system involvement in inconti- 392. Gorlin RJ, Sedano HL. The multiple nevoid basal cell carcinoma syndrome
nentia pigmenti: cranial MRI of two siblings. Neuroradiology 1998;40: revisited. Birth Defects 1971;7:140–148.
364–366. 393. Gorlin RJ. Nevoid basal cell carcinoma syndrome. Dermatol Clin
366. Fusco F, Fimiani G, Tadini G, Michele D, Ursini MV. Clinical diagnosis of 1995;13:113–125.
incontinentia pigmenti in a cohort of male patients. J Am Acad Dermatol 394. Kimonis V, Goldstein A, Pastakia B, et al. Clinical manifestations in 105
2007;56:264–267. persons with nevoid basal cell carcinoma syndrome. Am J Med Genet
367. Garrod AE. Peculiar pigmentation of the skin in an infant. Trans Clin Soc 1997;69:299–308.
Lond 1906;39:216–224. 395. Johnson RL, Rothman AL, Xie J, et al. Human homolog of patched,
368. Landy S, Donnai D. Incontinentia pigmenti (Bloch-Sulzberger syndrome). a candidate gene for the basal cell nevus syndrome. Science 1996;272:
J Med Genet 1993;30:53–59. 1668–1671.
369. Brodsky MC, Baker RS, Hamed LM. Pediatric Neuro-ophthalmology. 396. Shanley S, Ratcliffe J, Hockey A, et al. Nevoid basal cell carcinoma syndrome:
New York: Springer, 1996. review of 118 affected individuals. Am J Med Genet 1994;50:282–290.
370. Rosman NP. Incontinentia pigmenti. In: Gomez MR, ed. Neurocutaneous 397. Gorlin RJ. Nevoid basal-cell carcinoma syndrome. Medicine 1987;66:
Diseases: A Practical Approach. Stoneham: Butterworths, 1987:293–300. 98–113.
371. Phan TA, Wargon O, Turner AM. Incontinentia pigmenti case series: clini- 398. Evans DGR, Ladusans E, Rimmer S, et al. Complications of the naevoid
cal spectrum of incontinentia pigmenti in 53 female patients and their basal cell carcinoma syndrome: results of a population based study. J Med
relatives. Clin Exp Dermatol 2005;30:474–480. Genet 1993;30:460–464.
372. Hennel SJ, Ekert PG, Volpe JJ, Inder TE. Insights into the pathogenesis of 399. Kimonis VE, Mehta SG, Digiovanna JJ, Bale SJ, Pastakia B. Radiological
cerebral lesions in incontinentia pigmenti. Pediatr Neurol 2003;29:148. features in 82 patients with nevoid basal cell carcinoma (NBCC or Gorlin)
373. Shuper A, Bryan RN, Singer HS. Destructive encephalopathy in inconti- syndrome. Genet Med 2004;6:495–502.
nentia pigmenti: a primary disorder? Pediatr Neurol 1990;2:137–140. 400. Palacios E, Serou M, Restrepo S, Rojas R. Odontogenic keratocysts in
374. O’Doherty NY, Norman RM. Incontinentia pigmenti (Bloch-Sulzberger nevoid basal cell carcinoma (Gorlin’s) syndrome: CT and MRI evaluation.
syndrome) with cerebral malformation. Dev Med Child Neurol Ear Nose Throat J 2004;83:40–42.
1986;10:168–174. 401. Evans DGR, Farndon PA, Burnell LD, et al. The incidence of Gorlin
375. Hauw JJ, Perié G, Bonette J, Escourelle R. Les lesions cerebrales de syndrome in 173 consecutive cases of medulloblastoma. Br J Cancer
l’incontinentia pigmenti (a propos d’un cas anatomique). Acta Neuro- 1991;64:959–961.
pathol 1977;38:159–162. 402. Lacombe D, Chateil JF, Fontan D, et al. Medulloblastoma in the nevoid
376. Pascual-Castroviejo I, Pascual-Pascual SI, Velazquez-Fragua R, Martinez basal cell carcinoma syndrome. Genet Counsel 1990;1:273–277.
V. Incontinentia pigmenti: clinical and neuroimaging findings in a series 403. Stavrou T, Dubovsky EC, Reaman GH, Goldstein AM, Vezina G. Intracra-
of 12 patients. Neurologia 2006;21:239–248. nial calcifications in childhood medulloblastoma: relation to nevoid basal
377. Bryant SA, Rutledge SL. Abnormal white matter in a neurologically intact cell carcinoma syndrome. Am J Neuroradiol 2000;21:790–794.
child with incontinentia pigmenti. Pediatr Neurol 2007;36:199–201. 404. Crawford JR, Rood BR, Rossi CT, Vezina G. Medulloblastoma associated
378. Shah SN, Gibbs S, Upton CJ, Pickworth FE, Garioch JJ. Incontinentia with novel PTCH mutation as primary manifestation of Gorlin syndrome.
pigmenti associated with cerebral palsy and cerebral leukomalacia: a case Neurology 2009;72:1618.
report and literature review. Pediatr Dermatol 2003;20:491–494. 405. Garre ML, Cama A, Bagnasco F, et al. Medulloblastoma variants: age-
379. Ito M. Incontinentia pigmentia achromians. Singular case of nevus depig- dependent occurrence and relation to Gorlin syndrome–a new clinical
mentosus systemacitus. Tohoku J Exp Med 1952;55:57–59. perspective. Clin Cancer Res 2009;15:2463–2471.
380. Editorial. Hypomelanosis of Ito. Lancet 1992;339:651–652. 406. O’Malley S, Weitman D, Olding M, Sekhar L. Multiple neoplasms follow-
381. Fritz B, Kuster W, Orstavik K, Naumova A, Spranger J, Rehder H. Pigmen- ing craniospinal irradiation for medulloblastoma in a patient with nevoid
tary mosaicism in hypomelanosis of Ito. Further evidence for functionary basal cell carcinoma syndrome. J Neurosurg 1997;86:286–288.
disomy of Xp. Hum Genet 1998;103:441–449. 407. Pascual-Castroviejo I. The association of extracranial and intracranial
382. Taibjee SM, Bennett DC, Moss C. Abnormal pigmentation in hypomel- vascular malformations in children. Can J Neurol Sci 1985;12:139–148.
anosis of Ito and pigmentary mosaicism: the role of pigmentary genes. Br 408. Pascual-Castroviejo I, Viaño J, Moreno F, et al. Hemangiomas of the head,
J Dermatol 2004;151:269–282. neck, and chest with associated vascular and brain anomalies: a complex
383. Glover MT, Brett EM, Atherton DJ. Hypomelanosis of Ito: spectrum of the neurocutaneous syndrome. Am J Neuroradiol 1996;17:461–471.
disease. J Pediatr 1989;115:75–80. 409. Frieden IJ, Reese V, Cohen D. PHACE syndrome. The association of poste-
384. Pascual-Castroviejo I, Lopez-Rodriguez L, Medina M, Salamanca-Maesso rior fossa brain malformations, hemangiomas, arterial anomalies, coarc-
C, Herrero CR. Hypomelanosis of Ito. Neurological complications in 34 tation of the aorta and cardiac defects, and eye anomalies. Arch Dermatol
cases. Can J Neurol Sci 1988;15:124–129. 1996;132:307–311.

Barkovich_Chap06.indd 634 5/6/2011 9:04:49 PM


Chapter 6 • The Phakomatoses 635

410. Metry DW, Haggstrom AN, Drolet BA, et al. A prospective study of PHACE 435. Petit, R.E. and K.G. Berdal, Chédiak-Higashi syndrome: neurologic appearance.
syndrome in infantile hemangiomas: demographic features, clinical find- Arch Neurol, 1984. 41: p. 1001–1002.
ings, and complications. Am J Med Genet Part A 2006;140A:975–986. 436. Sheramata W, Kott HS, Cyr DP. The Chédiak-Higashi-Steinbrinck syn-
411. Krings T, Geibprasert S, Luo CB, Bhattacharya JJ, Alvarez H, Lasjaunias drome. Arch Neurol 1971;25:289–294.
P. Segmental neurovascular syndromes in children. Neuroimaging Clin N 437. Blume RS, Wolff SM. The Chédiak-Higashi syndrome: studies in four
Am 2007;17:245–258. patients and a review of the literature. Medicine 1972;51:247–280.
412. Castillo M. PHACES syndrome: from the brain to the face via the neural 438. Sung JH, Meyers JP, Stadlan EM, et al. Neuropathological changes in
crest cells. Am J Neuroradiol 2008;29:814–815. Chédiak-Higashi disease. J Neuropathol Exp Neurol 1969;28:86–118.
413. Metry DW, Dowd CF, Barkovich AJ, Frieden IJ. The many faces of PHACE 439. Kaplan J, De Domenico I, Ward DM. Chediak-Higashi syndrome. Curr
syndrome. J Pediatr 2001;139:117–123. Opin Hematol 2008;15:22–29.
414. Oza VS, Wang E, Berenstein A, et al. PHACES association: a neuroradio- 440. Ballard R, Tien RD, Nohria V, Juel V. The Chédiak-Higashi syndrome: CT
logic review of 17 patients. Am J Neuroradiol 2008;29:807–813. and MR findings. Pediatr Radiol 1994;24:266–267.
415. Aeby A, Guerrini R, David P, Rodesch G, Raybaud C, Van Bogaert P. Facial 441. Archambault L, Fromm NK. Progressive facial hemiatrophy. Arch Neurol
hemangioma and cerebral corticovascular dysplasia: a syndrome associ- Psychiatry 1932;7:529–584.
ated with epilepsy. Neurology 2003;60:1030–1032. 442. Carreno M, Donaire A, Barcelo MI, et al. Parry Romberg syndrome and
416. Judd CD, Chapman PR, Koch B, Shea CJ. Intracranial infantile hemangiomas linear scleroderma in coup de sabre mimicking Rasmussen encephalitis.
associated with PHACE syndrome. Am J Neuroradiol 2007;28:25–29. Neurology 2007;68:1308–1310.
417. Baccin C, Krings T, Álvarez H, Ozanne A, Lasjaunias P. A report of two 443. Dupont S, Catala M, Hasboun D, Semah F, Baulac M. Progressive facial
cases with dolichosegmental intracranial arteries as a new feature of hemiatrophy and epilepsy: a common underlying dysgenetic mechansim.
PHACES syndrome. Childs Nerv Syst 2007;23:559–567. Neurology 1997;48:1013–1018.
418. Hess CP, Fullerton HJ, Metry DW, et al. Cervical and intracranial arte- 444. Cory RC, Clayman DA, Faillace WJ, McKee SW, Gama CH. Clinical and
rial anomalies in 70 patients with PHACE syndrome. Am J Neuroradiol. radiologic findings in progressive facial hemiatrophy (Parry-Romberg
2010;31:1980–1986. Syndrome). Am J Neuroradiol 1997;18:751–758.
419. Heyer GL, Dowling MM, Licht DJ, et al. The cerebral vasculopathy of 445. Asher SW, Berg BO. Progressive hemifacial atrophy. Report of three cases
PHACES syndrome. Stroke 2008;39:308–316. and computed tomographic findings. Arch Neurol 1982;39:44–46.
420. Bhattacharya JJ, Luo CB, Alvarez H, Rodesch G, Pongpech S, Lasjaunias 446. Moon WJ, Kim HJ, Roh HG, Oh J, Han SH. Diffusion tensor imaging
PL. PHACES syndrome: a review of eight previously unreported cases and fiber tractography in Parry-Romberg syndrome. Am J Neuroradiol
with late arterial occlusions. Neuroradiology 2004;46:227–233. 2008;29:714–715.
421. Weon Y-C, Chung J-I, Kim H-J, Byun HS. Agenesis of bilateral internal 447. Okumura A, Ikuta T, Tsuji T, et al. Parry-Romberg syndrome with a
carotid arteries and posterior fossa abnormality in a patient with facial clinically silent white matter lesion. Am J Neuroradiol 2006;27:1729–1731.
capillary hemangioma: presumed incomplete phenotypic expression of 448. Longo D, Paonessa A, Specchio N, et al. Parry-Romberg Syndrome and
PHACE syndrome. Am J Neuroradiol 2005;26:2635–2639. Rasmussen Encephalitis: Possible Association. Clinical and Neuroimaging
422. Drolet BA, Dohil M, Golomb MR, et al. Early stroke and cerebral vas- Features. J Neuroimaging 2009.
culopathy in children with facial hemangiomas and PHACE syndrome. 449. Pupillo G, Andermann F, Dubeau F. Linear scleroderma and intractable
Pediatrics 2006;117:959–964. epilepsy: neuropathologic evidence for a chronic inflammatory process.
423. Burrows PE, Robertson RL, Mulliken JB, et al. Cerebral vasculopathy and Ann Neurol 1996;39:277–278.
neurologic sequelae in infants with cervicofacial hemangioma: report of 450. Rogers M, McCrossin I, Commens C. Epidermal nevi and the epidermal
eight patients. Radiology 1998;207:601–608. nevus syndrome. A review of 131 cases. J Am Acad Dermatol 1989;20:
424. Rao RP, Drolet BA, Holland KE, Frommelt PC. PHACES association: a 476–488.
vasculocutaneous syndrome. Pediatr Cardiol 2008;29:793–799. 451. Vidaurri-de la Cruz H, Tamayo-Sanchez L, Duran-McKinster C, de la Luz
425. Rudnick EF, Chen EY, Manning SC, Perkins JA. PHACES syndrome: Orozco-Covarrubias M, Ruiz-Maldonado R. Epidermal nevus syndromes:
otolaryngic considerations in recognition and management. Int J Pediatr clinical findings in 35 patients. Pediatr Dermatol 2004;21:432–439.
Otorhinolaryngol 2009;73:281–288. 452. Solomon LM, Esterly NB. Epidermal and other congenital organoid nevi.
426. Katz SE, Rootman J, Vangveeravong S, Graeb D. Combined venous lym- Curr Probl Pediatr 1975;6:1–56.
phatic malformations of the orbit (so-called lymphangiomas): associa- 453. Happle R. How many epidermal nevus syndromes exist? A clinicogenetic
tion with noncontiguous intracranial vascular anomalies. Ophthalmology classification. J Am Acad Dermatol 1991;25:550–556.
1998;105:176–184. 454. Pavone L, Curatolo P, Rizzo R, et al. Epidermal nevus syndrome: a neu-
427. Poindexter G, Metry DW, Barkovich AJ, Frieden IJ. The expanding spec- rologic variant with hemimegalencephaly, gyral malformation,mental
trum of PHACE syndrome: ventricular heterotopia, endocrine dysfunc- retardation, seizures, and facial hemihypertrophy. Neurology 1991;41:
tionn, and intracerebral hemangiomas. Pediatr Neurol 2007;36:402–406. 266–271.
428. Giardini A, Gholam C, Khambadkone S, Kostolny M. Need for compre- 455. Zaremba J. Jadassohn’s naevus phakomatosis: a study based on a review of
hensive vascular assessment before surgical repair of aortic coarctation in thirty-seven cases. J Ment Defic Res 1978;22:103–123.
PHACES syndrome. Pediatr Cardiol 31:291–293. 456. Gurecki PJ, Holden KR, Sahn EE, Dyer DS, Cure JK. Developmental neural
429. Al-Kaabi A, Yanofsky R, Bunge M, Hyman J, Rafay MF. Diffuse heman- abnormalities and seizures in epidermal nevus syndrome. Dev Med Child
giomatosis with predominant central nervous system involvement. Pedi- Neurol 1996;38:716–723.
atr Neurol 2009;40:54–57. 457. Barth PG, Valk J, Kalsbeek GL, Blom A. Organoid nevus syndrome (linear
430. Stratte EG, Tope WD, Johnson CL, et al. Multimodal management of dif- nevus sebaceus of Jadassohn): clinical and radiological study of a case.
fuse neonatal hemangiomatosis. J Am Acad Dermatol 1996;34:337–342. Neuropädiatrie 1977;8:418–428.
431. Goitz LE, Rudikoff J, O’Meara OP. Diffuse neonatal hemangiomatosis. 458. Baker ES, Ross PA, Baumann RJ. Neurologic complications of the epider-
Pediatr Dermatol 1986;3:145–152. mal nevus syndrome. Arch Neurol 1987;44:227–232.
432. Vlahovic A, Simic R, Djokic D, Ceran C. Diffuse neonatal hemangioma- 459. Kara B, Inan N, Bayramgurler D, Altintas O, Akbulut A. Epidermal nevus
tosis treatment with cyclophosphamide: a case report. J Pediatr Hematol syndrome with azygos anterior cerebral artery. Pediatr Neurol 2008;39:
Oncol 2009;31:858–860. 283–285.
433. Poirier VC, Ablin DS, Frank EH. Diffuse neonatal hemangiomatosis: a case 460. Canyigit M, Oguz KK. Epidermal nevus syndrome with internal carotid
report. Am J Neuroradiol 1990;11:1097–1099. artery occlusion and intracranial and orbital lipomas. Am J Neuroradiol
434. Balaci E, Sumner TE, Auringer ST, Cox TD. Diffuse neonatal hemangiom- 2006;27:1559–1561.
atosis with extensive involvement of the brain and cervical spinal cord. 461. Mall V, Heinen F, Uhl M, Wellens E, Korinthenberg R. CNS lipoma in patients
Pediatr Radiol 1999;29:441–443. with epidermal nevus syndrome. Neuropediatrics 2000;31:175–179.

Barkovich_Chap06.indd 635 5/6/2011 9:04:49 PM


636 Pediatric Neuroimaging

462. Booth TN, Rollins NK. MR imaging of the spine in epidermal nevus 468. Dietrich RB, Glidden DE, Roth GM, Martin RA, Demo DS. The
syndrome. Am J Neuroradiol 2002;23:1607–1610. Proteus syndrome: CNS manifestations. Am J Neuroradiol 1998;19:
463. Moog U. Encephalocraniocutaneous lipomatosis. J Med Genet 2009;46: 987–990.
721–729. 469. Pilarski R. Cowden syndrome: a critical review of the clinical literature.
464. Parazzini C, Triulzi F, Russo G, Mastrangelo M, Scotti G. Encephal- J Genet Couns 2009;18:13–27.
ocraniocutaneous lipomatosis: complete neuroradiologic evaluation and 470. Padberg G, Schot J, Vielvoye G, et al. Lhermitte-Duclos disease and
follow-up of two cases. Am J Neuroradiol 1999;20:173–176. Cowden disease: a single phakomatosis. Ann Neurol 1991;29:517–523.
465. Moog U, Jones MC, Viskochil DH, Verloes A, Van Allen MI, Dobyns WB. 471. Williams III D, Elster A, Ginsberg L, Stanton C. Recurrent Lhermitte-
Brain anomalies in encephalocraniocutaneous lipomatosis. Am J Med Duclos disease: report of two cases and association with Cowden’s disease.
Genet A 2007;143A:2963–2972. Am J Neuroradiol 1992;13:287–290.
466. Biesecker L. The multifaced challenges of Proteus syndrome. JAMA 472. Lynch NE, Lynch SA, McMenamin J, Webb D. Bannayan-Riley-Ruvalcaba
2001;285:2240–2243. syndrome: a cause of extreme macrocephaly and neurodevelopmental
467. Griffiths PD, Welch R, Gardner-Medwin D, Gholkar A, McAllister V. The delay. Arch Dis Child 2009;94:553–554.
radiological features of hemimegalencephaly including three cases associ- 473. Neurofibromatosis. Conference statement. National Institutes of Health
ated with proteus syndrome. Neuropediatrics 1994;25:140–144. Consensus Development Conference. Arch Neurol 1988;45:575–578.

Barkovich_Chap06.indd 636 5/6/2011 9:04:50 PM


CHAPTER

Intracranial, Orbital, and Neck


7 Masses of Childhood
CHARLES RAYBAUD AND A. JAMES BARKOVICH

Introduction Pineal region masses


Incidence and severity of brain tumors in Children Extraparenchymal tumors
Changing concepts of the biology of brain tumors Brain tumors in the first year of life
Clinical features of brain tumors Radiation-induced tumors of the central nervous system
Techniques of imaging pediatric brain tumors Tumors of the head and neck in childhood
Imaging characteristics used to identify brain tumors Ocular tumors
Posterior fossa tumors Extraocular orbital masses
Intraparenchymal tumors Masses of the face and neck
Extraparenchymal tumors
Supratentorial tumors
Tumors of the cerebral hemispheres
Sellar and suprasellar tumors

those in which the tumor could not be resected. Of note, the OS rate
INTRODUCTION continues to decrease in children (but not in infants) beyond 5 and
Incidence and Severity of Brain even 10 years (5). In one report, the risk of untimely death is increased
Tumors in Children 13-fold in adult survivors of childhood CNS malignancies; among chil-
dren surviving more than 5 years, more than 25% will die within 30
Brain CNS tumors represent the second most common type of cancer years of their diagnosis (by definition, before reaching the age of 49
(17% of all childhood cancer) and are the second most common cause years) (21). The most common cause of death is recurrence or progres-
of cancer deaths (25%) in children, being exceeded only by leukemia sion of the disease (61%); others include subsequent neoplasms (9%),
(1–3). There are approximately 20,500 primary brain tumors diagnosed cardiac disease (3%), and pulmonary disease (3%) (21). The risk of
in the United States each year, and of these, 3,750 (or 18.3%) occur in early death is highest in survivors of medulloblastoma/PNET. In addi-
patients less than 19 years of age (4). This corresponds to an incidence tion, neurological, neurocognitive, and endocrinological functions are
rate of 4.5/100,000 per year in persons aged 18 years or younger. A similar commonly altered in childhood CNS tumor survivors who have been
incidence of 4.2/100,000 per year was reported in Sweden (1984–2005) subjected to surgery, heavy radiotherapy, and chemotherapy (21–25).
before the age of 15 years (5). In most large series, supratentorial tumors
(including the third ventricular tumors) and infratentorial tumors
occur in nearly equal frequency (6–9). However, supratentorial tumors Changing Concepts of the Biology
are more common in the first 2 or 3 years of life, whereas infratento- of Brain Tumors
rial tumors predominate from age 4 to 10 years (7–16). Both locations
develop tumors with nearly equal frequency in children older than 10 Traditionally, tumors have been considered to result from “de-
years. Whereas metastases, glioblastoma multiforme (GBM), and men- differentiation” of mature cells. In 1997, it was recognized in leukemia
ingiomas represent most of the tumor types encountered in adults, that all tumoral cells formed a hierarchy apparently derived from a
these are uncommon in children. On the contrary, embryonal/primitive primitive hematopoietic cell (26). In a similar way, it was recognized
neuroectodermal/medulloblastoma and pilocytic astrocytomas are the that brain tumors reproduce a hierarchy of deviant neuroepithelial
most common tumors before age 9 years, and gliomas grade I to III cells that are unable to proliferate, while the tumor growth results
between 10 and 19 years. In addition, a multiplicity of diverse, uncom- from the activity of a small component of stem cell–like tumoral
mon tumor types occur in 36.5% (17) (Table 7-1) to 47% (18); this cells that have been called Brain Tumor Stem Cells (BTSC) (1,27).
diversity sometimes makes a precise diagnosis uncertain. During brain development, normal stem cells (NSC) proliferate in
Overall survival (OS) of brain tumors in children is 70% at 10 years, the early neural tube, each one producing two daughter cells (sym-
but this obviously includes benign tumors for which complete resec- metric division) and, thus, increasing the cellularity of the develop-
tion is possible (5). In fact, OS is very low for the diffuse intrinsic pon- ing brain. During the development of the cerebral cortex (from week
tine glioma (<10%–15% survival at 5 years, mean survival 11 months) 7), each NSC produces one progenitor cell and one daughter NSC
(19,20), higher in patient with medulloblastoma (53% at 10 years) (5), that remains within the proliferative pool (asymmetric division). The
and about 85% to 90% in children with low-grade gliomas, including progenitor cells give rise to the radial glial cells (future astrocytes),

637

Barkovich_Chap07.indd 637 5/6/2011 9:15:29 PM


638 Pediatric Neuroimaging

neurons, oligodendrocytes, and ependymocytes. Neuronal produc-


TABLE 7-1 Percentages of Histological tion mostly stops about mid-gestation, and glial mass production
decreases after the first years of life. However, quiescent NSC (called
Types of Tumors involving type B cells) persist in the subventricular zone (SVZ, see the section
the CNS at Hospital for on “Malformations of Cortical Development” in Chapter 5 for fur-
Sick Children ther discussion of the ventricular zone and SVZ) in the region of the
lateral ventricle and are able to periodically generate multipotent
progenitor cells (called type C cells). C cells give rise to more mature
Astrocytoma 39.4
transient progenitor cells (called type A cells), which, in turn, pro-
Low grade (WHO I-II) 32.3 duce neurons that renew the granule cells of the dentate gyrus and
High grade (WHO III-IV) 7.1 the interneurons of the olfactory bulbs indefinitely (27). These per-
sisting NSC are also able to replace damaged glial cells (astrocytes,
Embryonal 15.4
oligodendrocytes) in the setting of diseases such as ischemic necrosis
Medulloblastoma 10.6 and demyelination.
PNET 3.9 Rather than a mass of dedifferentiated mature neural cells, a
brain tumor now is perceived as a deviant reproduction of the normal
ATRT 0.9
development (1,27,28). As mentioned above, it has been recognized
Ependymoma 7.0 that only a few stem cell–like tumoral cells (called BTSC, brain cancer
Classic (WHO II) 4.9 stem cells [CSC], or brain cancer propagating cells [CPC]) have the
growth and the invasiveness characteristics of a tumor. These BTSC
Anaplastic (WHO III) 1.7 have many characteristics of NSC: proliferation, self-renewal, and
Myxopapillary (WHO I) 0.3 multipotency. In the same way as transplanted NSC reproduce nor-
Oligodendroglioma (WHO II) 1.7 mal brain cellularity, transplanted BTSC reproduce a tumor with the
same characteristics as the original tumor. Just as the NSC give rise
Craniopharyngioma 6.8 to progenitors that migrate away from the SVZ, the BTSC give rise
Neuronal, neuronal-glial 6.8 to tumors cells that may proliferate at a distance from the site where
Ganglioglioma 4.7 they originate. BTSC have the same molecular markers as the NSC
(CD133+ mainly) and respond to the same signaling proteins (nota-
DNET 1.9 bly EGFR, Notch, BMP, Shh, Wnt) (1,27–29). The cell type that devel-
Neurocytoma 0.2 ops into BTSC has not been identified. It is likely to be a precursor cell
Choroid plexus tumors 2.3 from the ventricular or SVZ. As it is estimated that it requires 4 to 7
mutations before a normal somatic cell acquires a cancerous pheno-
Papilloma (WHO I) 1.3 type, the life of the transient dividing progenitors before they divide
Carcinoma (WHO III) 1.0 might be too short (12 hours) to be affected by these mutations; in
Pineal 0.5 contrast, the life of a NSC is long (28 days), which might make it a
more likely candidate (27). Other factors that might be important in
Germ cell tumors 3.1 brain tumor development include presence of multiple genetic muta-
Pituitary 1.0 tions (in neurofibromatosis type 1 [NF1], the combined mutation of
Mesodermal 9.7 the Nf1 gene on one hand, and the mutation of the p53 gene or the
loss of the CDKN2A locus on the other hand, increase the forma-
Non meningothelial 5.0 tion of astrocytomas [27]) or modification of its environment. The
Meningioma 1.7 local stromal microenvironment, especially the vascularity, may play
Langerhans cell histiocytosis 3.0 a significant role in the development of tumors (the “seed and soil”
concept [30]). Perhaps more important is the realization that NSC are
Radiculo-neural 3.1 genetically programmed to respond to the environment within spe-
Schwannoma 1.8 cific segments of the neural tube. The CSC that derive from those seg-
ments are also genetically programmed to remain there. This suggests
Neurofibroma 1.3
that tumors such as ependymomas or pilocytic astrocytomas located
Miscellaneous in different CNS segments (e.g., hindbrain and forebrain) may have
Subependymoma a fully identical histology while being, molecularly, entirely different
tumors, with different natural history, clinical response to treatment,
Ganglioneuroma
and prognosis (31–33). Clearly, a better understanding of these fac-
Hemangiopericytoma tors will have an enormous impact of diagnosis and treatment of
Hemangioblastoma tumors in the future.
Primary CNS lymphoma
Primary CNS melanoma Clinical Features of Brain Tumors
Nearly all pediatric brain neoplasms form masses, which infiltrate
ATRT, atypical teratoid-rhabdoid tumor; DNET, dysplastic neuroectodermal
tumor; PNET, primitive neuroectodermal tumor; WHO, World Health or compress the adjacent parenchyma and result in local functional
Organization tumor grade. impairment. By occupying space in the cranial cavity and eliciting
From (17). edema, the tumor causes increased intracranial pressure, which may be
exacerbated by hydrocephalus if the tumor develops close to the mid-

Barkovich_Chap07.indd 638 5/6/2011 9:15:29 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 639

TABLE 7-2 Indications for Neuroimaging in Pediatric Headache


1. Persistent headaches of <6 mo duration that do not respond to medical treatment
2. Headache associated with abnormal neurologic findings
3. Persistent headaches without family history of migraine
4. Persistent headache associated with substantial episodes of confusion, disorientation, or emesis
5. Headaches that awaken a child repeatedly from sleep or occur immediately on awakening
6. Family or medical history of disorders that may predispose the child to central nervous system lesions and clinical or laboratory findings
suggestive of CNS involvement
From (34).

line and restricts CSF circulation. All these factors combine to stretch sensitive than CT for the detection of tumor spread via the suba-
axonal pathways, distort and herniate segments of brain, compress the rachnoid spaces, which is particularly common in pediatric tumors
vascular bed, and produce neurological impairment. Therefore, the (35–37).
cardinal clinical features of brain tumors are increased intracranial For MR evaluation, T1, T2, and FLAIR imaging are the basic
pressure (with or without hydrocephalus or macrocrania), seizures, sequences, acquired in three planes, for example T1 sagittal, T2 coro-
neurological and/or endocrinological deficits, and cognitive impair- nal, and FLAIR axial. Sagittal T1 should preferably be obtained using
ment. These may evolve over a long or a short period of time, depending 3D-GE sequences (MPRAGE/TFE/SPGR) because of their high spatial
on the location and aggressivity of the tumor. As always in intracranial definition and excellent gray-white contrast, in addition to the ability
hypertension, acute decompensation may occur at any time, even in to reformat the data in any plane. FSE T2 and FLAIR also should use
the case of a benign tumor. 3D volumetric acquisition or, if 2D acquisition is preferred, thin slices
The symptoms of children with CNS tumors depend upon the (3–4 mm or less if possible) with minimal gap to better demonstrate
age at the time of presentation. Infants present with increasing head the relationship of the tumor to surrounding structures. Generally,
circumference, nausea, vomiting, and lethargy (8). Children often midline (suprasellar, third ventricular—including pineal—and fourth
have the same presenting symptoms and signs; in addition, they may ventricular) masses are best imaged in the sagittal, coronal, and axial
complain of headache or decreased visual acuity and develop partial planes; for suprasellar masses, thin sections should be obtained to help
onset seizures or focal neurological signs such as cranial nerve palsies, determine the precise relationship of the mass to the optic chiasm, optic
truncal and limb ataxia, and hemiparesis. Deterioration in school per- nerves, and hypothalamus. Brainstem tumors are best seen in sagittal
formance is often noticed in older children. Tumors developing in the and axial planes. Cerebral and cerebellar hemispheric masses are best
region of the hypothalamus frequently produce endocrine dysfunc- imaged in the axial and coronal planes. High-definition submillimetric
tion such as diabetes insipidus, growth arrest, hyperphagia or preco- T2 imaging (constructive interference in steady state [CISS], FIESTA)
cious puberty. A rapid deterioration points to aggressive, typically is useful for studies of the cerebral aqueduct and extracerebral masses
malignant tumor, while a slow progression may go unnoticed until such as schwannomas. Diffusion-weighted imaging (DWI) is now
a major deficit becomes apparent. For example, in benign tumors used routinely and has proved to be extremely useful in distinguishing
such as craniopharyngiomas, rapid loss of vision may lead to diagno- hypercellular from hypocellular tumors. Susceptibility-weighted imag-
sis after years of diminished growth were not noticed. Headaches in ing (SWI) is sensitive to the presence of blood and calcification. MR
themselves are not an indication for neuroimaging (34). Indications proton spectroscopy is useful to evaluate the impairment of the neu-
for neuroimaging in pediatric patients with headaches have been pub- ronal function (NAA, 2.0 ppm), membrane construction and destruc-
lished (Table 7-2). tion (Cho, 3.2 ppm), energy metabolism (Cr, 3.0 ppm), and the degree
of necrosis (Lac, 1.33 ppm; Lip, broad peak at 0.9 ppm).
Gadolinium compounds (paramagnetic contrast) are always used
TECHNIQUES OF IMAGING PEDIATRIC to appreciate the degree of permeability of the blood–tumor barrier
BRAIN TUMORS and to improve the detection of CSF spread of tumor (“drop metas-
Imaging is critical to the diagnosis of brain tumor, as no other diag- tases”). It is administered in a dose of 0.1 mg/kg body weight, given as
nostic test demonstrates the lesion with as much reliability. Although an intravenous bolus immediately prior to the postcontrast sequences.
CT may be the imaging method most commonly used for the ini- It is best to always administer the contrast agent at the same time rela-
tial diagnosis of intracranial neoplasms, MRI is the study of choice tive to scanning, as delays allow the contrast to diffuse farther in the
because of its sensitivity and because the multiplanar imaging capa- extracellular space, resulting in larger regions of enhancement and may
bility is extremely useful in determining the exact extent of the tumor result in a false interpretation of change in tumor size based on extent
and its relationship to surrounding normal structures. MRI can be of enhancement. Either flow compensation techniques (peripheral or
used in conjunction with computerized navigation techniques that cardiac gating, or gradient moment nulling) or very short echo times
greatly facilitate tumor resection. MRI has a particular advantage in (<10 milliseconds) should be used when the tumor is in the posterior
the analysis of tumors of the posterior fossa, in which artifacts from fossa, to reduce obscuration of vital areas by spatial misregistration of
the surrounding bone hinder CT analysis. Moreover, MRI is more the contrast-enhanced flowing blood in the transverse and sigmoid

Barkovich_Chap07.indd 639 5/6/2011 9:15:29 PM


640 Pediatric Neuroimaging

sinuses. When a tumor prone to disseminate in the CSF spaces is sus- locally and globally, and prepare the surgical approach. Subsequent
pected, postcontrast T1 imaging of the entire spinal cord and cauda imaging will be used to assess the effects of therapy.
equina should be performed in the sagittal plane, complemented with Detecting a mass lesion is usually not a problem with modern
localized axial images when drop metastases are suspected. Fat sup- imaging, especially with MRI. Masses are typically recognized on CT
pression techniques are valuable in differentiating enhancing tumor or MRI on the basis of either density or signal intensity differences
from epidural fat on T1-weighted images (37). Finally, presurgical from normal brain, mass effect causing distortion of normal brain
sequences may include axial millimetric 3D-GE sequences compatible structures, or the presence of abnormal enhancement after the intra-
with the neuronavigation tools, functional MR imaging (fMRI) to venous administration of contrast. (In the normal central nervous sys-
locate the main regions for motor and cognitive functions, especially tem [CNS], contrast enhancement is only seen in the pituitary gland,
the language areas; sensorimotor white matter fascicles can also be pituitary stalk, tuber cinereum of the hypothalamus, pineal gland,
determined by diffusion tensor imaging (DTI). blood vessels, and the choroid plexuses.) Almost all brain tumors
When a CT scan is performed, the most important images are pre- are either hypodense or hyperdense compared to normal brain on
contrast images. These allow a reliable assessment of tumor cellularity: noncontrast CT images, hypointense to myelinated white matter on
high attenuation indicating high cellularity and low attenuation indi- T1-weighted MR images, and hyperintense to myelinated white mat-
cating low cellularity. It also shows presence of blood and calcification. ter on T2-weighted MR images. Hemorrhage, necrosis, calcifications,
Multiplanar reformats are useful, but, even though modern CT scans and patterns and intensity of contrast enhancement may vary among
have a much better resolution and fewer artifacts than older ones, the tumors of similar histologic class, and even more so among tumors
anatomical quality is still inferior to that of MRI. Iodinated contrast of different cell type, but they may also overlap; these characteristics
media are usually administered, but this is not really crucial if MRI is to are discussed more thoroughly later in this chapter. All tumors exhibit
be performed shortly afterwards. The recommended contrast dose in mass effect. In the case of relatively slowly growing, peripherally
these patients is 3 mg/kg of contrast (concentration 300 mg I2/mL) up located tumors, the mass effect results in expansion or erosion of the
to a maximum dose of 120 mL, usually given in a single bolus imme- cranial vault. In more rapidly growing, more centrally located tumors,
diately prior to the scan. the effect is a displacement of structures. Displacement may result in
herniation of brain tissue through or around rigid structures, such as
IMAGING CHARACTERISTICS USED the falx cerebri, tentorium cerebelli, or foramen magnum. Supratento-
rial masses typically cause herniation medially under the falx cerebri
TO IDENTIFY BRAIN TUMORS or downward through the tentorial incisura, whereas infratentorial
Imaging analysis must proceed by steps. In successive order, one must masses cause upward transtentorial herniation and downward her-
find the lesion and recognize that it is a mass, locate it precisely (as niation through the foramen magnum. Transtentorial herniation clas-
location, together with appearance, helps in recognizing the tumor sically results in pressure on the midbrain with secondary effects on
type and is basic to the therapeutic strategy), describe its character- eye movement and pupillary constriction (the “blown pupil”). Tran-
istic features (including vascularity, vascular permeability, and meta- stentorial herniation may also result in stretching and compression
bolic spectrum), identify it as a tumor, assess the margins to determine of the posterior cerebral arteries against the tentorium, and conse-
how much it infiltrates the surrounding tissue, evaluate the mass effect quent unilateral or bilateral infarction of the posterior cerebral artery

FIG. 7-1. Extraparenchymal tumor characteristics. A. Axial noncontrast T1-weighted image shows a sharply defined mass (meningioma, black arrows) in
the posterior fossa on the left. The ipsilateral cerebellopontine angle cistern (white arrows) is enlarged compared to the contralateral cerebellopontine angle
cistern (white arrowheads). The dilated right temporal horn indicates the presence of hydrocephalus due to compression/distortion of the fourth ventricle
and aqueduct. B. Perfusion curve (line 4, relaxivity in the vertical axis, time in the horizontal axis) shows characteristics typical of an extraparenchymal
tumor. The signal intensity decreases as the contrast bolus enters the tumor vessels but does not recover because contrast leaks into the interstitium of the
tumor in the absence of a blood–brain barrier. Line 5 shows the perfusion curve of normal cerebellar parenchyma.

Barkovich_Chap07.indd 640 5/6/2011 9:15:30 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 641

FIG. 7-2. Intraparenchymal tumor characteristics. A. Axial postcontrast T1-weighted image shows a poorly defined mass (arrows) in the left frontal lobe.
Surrounding sulci are compressed. B. Perfusion curve (line 5) shows characteristics of a typical intraparenchymal glial tumor. Signal intensity decreases as the
bolus of contrast courses through the tumor vessels, and then recovers nearly to baseline as the contrast leaves the tumor. The area over the perfusion curve of
the tumor (line 5) is greater than that of normal white matter (line 4), indicating increased blood volume in the tumor and, probably, high tumor grade.

distribution. Herniation of the cerebellar tonsils through the foramen A good anatomical analysis is central to the diagnosis of brain
magnum can occur both with supratentorial and infratentorial masses; tumor for three main reasons: it defines the resectability (e.g., cerebel-
tonsillar herniation, which is easily detected by MRI, can lead to respi- lar versus hypothalamic pilocytic astrocytoma), it allows the neuro-
ratory compromise. surgeon to choose the better approach, it can be a major indicator of
If a mass is identified, it must be determined whether the lesion the nature of a tumor (e.g., ventral diffuse intrinsic pontine glioma
arises from brain parenchyma or from extraparenchymal struc- versus dorsal tegmental pontine or medullary pilocytic astrocytoma).
tures, such as meninges or the subarachnoid space. Extraparenchymal T1, especially high-definition 3D-GE T1 imaging, thin (3.0 mm)
lesions typically displace, rather than invade, normal brain structures T2 and FLAIR in different planes, submillimetric T2 CISS-FIESTA if
(Fig. 7-1); the cortex is displaced medially (away from bone) by the mass, needed, all combine in providing the optimal information regarding
and this may result in an enlarged cistern. Intraparenchymal masses the precise location of the tumor (Table 7-3).
tend to compress cisterns and sulci. A well-defined margin separates The structural characteristics of the tumor include the cellularity;
the extraparenchymal mass from the brain; usually, little or no edema the presence of cysts, necrosis, blood or calcification; and the vascular-
surrounds the mass. Intraparenchymal masses often merge impercepti- ity and vascular permeability (blood–tumor barrier). (Obviously, any
bly into the normal tissue surrounding brain (Fig. 7-2) and may generate abnormality of signal, diffusivity, of MR spectrum, etc., should be analyzed
considerable edema. Other characteristics that are suggestive of an extra- only after consideration of the age and degree of maturation of the child;
parenchymal origin are a relative paucity of mass effect for the size of the serious mistakes can otherwise be made.) These characteristics alone often
lesion, extension of the mass across the midline without involving a cere- permit the identification of the tumor type; even if they don’t, they ori-
bral commissure, involvement of bone, and relatively mild presenting ent toward its benign or malignant nature. Although CT cannot replace
symptoms. The reader should be aware, however, that the differentiation MRI in this regard, it may be helpful. Low attenuation indicates high
of intraparenchymal and extraparenchymal masses is not always easy. water content/low cellularity (e.g., low grade glioma, tumor edema,
Intraparenchymal masses may be exophytic, thereby enlarging the adja- or necrosis); high attenuation close to that of gray matter suggests
cent cistern, whereas extraparenchymal masses may invade the brain high cellularity (e.g., medulloblastoma/PNET, germinoma); and avid
and elicit edema. Intraparenchymal masses may grow slowly and gener- enhancement suggests high vascular permeability. Cystic components,
ate relatively little mass effect. However, the general rules outlined above hemorrhage, and calcification are easily recognized and can help to
will allow determination of the location of tumor origin in most cases. specify a diagnosis in some locations. However, MRI is more specific.
A useful adjunct for differentiating intraparenchymal from extraparen- Reduced water diffusivity on diffusion imaging studies indicates the
chymal tumors is dynamic susceptibility-weighted contrast-enhanced high cellularity and high nuclear-cytoplasmic ratio (“little blue cells”)
perfusion imaging. The perfusion characteristics of intraparenchymal found in embryonal tumors (medulloblastoma, PNET, pineoblastoma,
and extraparenchymal tumors are different because of the absence of ependymoblastoma, retinoblastoma, and often atypical teratoid/rhab-
the blood–brain barrier in extraparenchymal tumors. This can be best doid tumors [ATRT]) and germinomas rather than a less cellular (typi-
appreciated by looking at the DR2* curve, which will show a return of cally more benign) tumor type. Uncommonly, reduced diffusion may
relaxivity toward baseline level after the passage of the bolus in an intra- result from recent tissue infarction (38). Other times, an area of reduced
parenchymal tumor (Fig. 7-2B), but persistence of abnormal relaxivity diffusivity (38) or increased fractional anisotropy (FA) (39) in a low-
(due to rapid extravasation of contrast into the interstitial spaces of the grade (high-diffusivity) tumor, such as a WHO grade II astrocytoma,
tumor) in an extraparenchymal tumor (Fig. 7-1B). results from a focus of malignant transformation. Tumoral cysts always

Barkovich_Chap07.indd 641 5/6/2011 9:15:30 PM


642 Pediatric Neuroimaging

TABLE 7-3 Main Tumor Types per Location


Compartment Segment Nature Development Specificities
Posterior fossa
Brainstem Cervicomedullary PA Intrinsic/exophytic Cystic/solid
Ventral pons DIPG BA encased ↑ myo-inositol
Dorsal pons PA Dorsal/exophytic Often cystic
Middle peduncle PA Well demarcated Often cystic
Midbrain PA Intrinsic ↓ Aqueduct
4th ventricle Roof, vermian MB Ventral/ventricular Reduced diffusion
Floor, medullary Ependymoma Exophytic cisternal CM or CPA
Cerebellum Any PA Intrinsic Cystic/solid
3rd ventricle
Anterior Optic pathways PA NF1, intrinsic Solid mostly
Hypothalamus PA Infiltrative Solid mostly
Hypothalamus Cranio Postchiasmatic Solid/cystic/Ca++
Sellar/suprasellar Cranio Prechiamatic Solid/cystic/Ca++
Sella/hypothalamus GCT Regional Reduced diffusion
Sellar/suprasellar Adenoma Prechiasmatic Solid
Posterior Pineal PB Cisternal Reduced diffusion
Posterior V3 GCT Ventricular Reduced diffusion
Pineal PA Cisternal Solid/cystic
Lateral ventricles Tela choroidea CPP Choroid plexus ↑ myo-inositol
Monro CPC Invasive ++ ↑ Cho, ↓ Cr/PCr
SEGA TSC Solid, Ca++
Hemispheres
Central gray Any Gliomas BG + thalamus Any grade
White matter Any Gliomas Transcerebral Any grade
Any Ependymoma Not ventricular Solid/cystic/Ca++
Cortex Temporal GGG GM and WM Solid/cystic
Any DNET GM and WM Segment-like
Any PXA Brain and meninges Solid/cystic

BG, basal ganglia; Cranio, craniopharyngioma; CM, cisterna magna; CPA, cerebello-pontine angle; CPC, choroid plexus carcinoma; CPP, choroid plexus papil-
loma; DIPG, diffuse infiltrating pontine glioma; DNET, dysembryoplastic neuroepithelial tumor; GCT, germ cell tumors; GGG, ganglioglioma; GM, gray matter;
MB, medulloblastoma;. PA, pilocytic astrocytoma; PB, pineoblastoma; PXA, pleomorphic xanthoastrocytoma; SEGA, subependymal giant cell astrocytoma; TSC,
tuberous sclerosis complex; WM, white matter.

differ in signal intensity from CSF of arachnoid cysts or loculations; and permeability of the tumoral blood vessels. Enhancement of the
contrast layering may be observed after intravenous contrast admin- lesion characteristically results from a combination of high vascularity
istration. Necrosis appears as rather heterogeneous loculations, typi- and of egress of contrast across the walls of abnormal vessels into the
cally showing nonenhancing, low T1, bright T2/FLAIR signal; it usually interstitium of the tumor. This enhancement may be diffuse, patchy,
reflects malignancy although it may be observed in large nonmalignant or ring-like; it may be intense or weak. A tumor with a same histology
tumors or in radiation injury (see Chapter 3). Vasogenic edema is well may enhance differently in different patients, and in the same patient
depicted in the surrounding white matter but difficult to differentiate on different studies. It is important to remember that, unlike in adults,
from infiltrative tumors using T2, FLAIR, or even DWI (38). Recent enhancement of a brain tumor in children is not evidence of malignancy;
blood or blood residue are easily recognized with SWI. Invasion and indeed, some of the most benign tumors (such as pilocytic astrocy-
destruction of the white matter tracts is best differentiated from dis- tomas) enhance the most. Although not commonly used in a clini-
placement by use of FA maps or DTI tractography (38) but, in the set- cal setting, perfusion studies, especially dynamic contrast-enhanced
ting of severe vasogenic edema, anisotropy of the water diffusion can be MRI can provide a relative quantification of the perfusion parame-
reduced and the tracts may not be identified. ters (cerebral blood volume [CBV], TT, cerebral blood flow [CBF]),
The administration of contrast media allows assessment of the and from these inform about the tumor type and grade (Figs. 7-1 to
integrity of the blood–brain barrier and, if perfusion imaging is used, 7-4) (38,40); it may also help in ruling out nonneoplastic tumor-like
can determine blood flow, mean transit time (TT), blood volume, lesions (38). Given the role of angiogenesis in tumor development,

Barkovich_Chap07.indd 642 5/6/2011 9:15:31 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 643

TABLE 7-4 Main MRS Markers of Brain Tumors


Compound Location (ppm) Metabolic Marker of… Change in Tumors
NAA 2.0 Neuronal activity mostly ↓ (all)
Cr/PCr 3.0 Energy cascade =/↓ (PA)
Cho 3.2 Cellular membranes, density, turnover ↑ (all)
Lac 1.33 Necrosis, malignancy ↑ (PA, malignant)
Lip (short TE) 0.9/1.3 Necrosis ↑ (malignant)
mI (short TE) 3.6 Cellularity, astrocytes ↑↑ (CPP)
Tau (short TE) 3.36 Cerebellar development ↑ (MB)
Cho, choline; CPP, choroid plexus papilloma; Cr/PCr, creatine/phosphocreatine; Lac, lactate; Lip, lipid; MB,
medulloblastoma/PNET; mI, myo-inositol; NAA, N-acetylaspartate; PA, pilocytic astrocytoma; Tau, taurine.

and the fact that new therapeutic agents target the vascular prolifera- thin and regular, and the central cavity demonstrates strongly restricted
tion, it may also provide an indicator of treatment efficacy (41,42). diffusion (49) (Fig. 7-5). In contrast, the cystic/necrotic components
Most reported studies, however, concern gliomas only (41), and adult of most tumors generally (but not always!) show increased diffusion
rather than pediatric brain tumors (38,43). Indeed, the pilocytic due to cellular necrosis and increasing size of the intercellular space
astrocytomas, very common in children, are highly vascularized and (49). The reduction of water motion may be less apparent with small
yet very benign. Permeability measures of the capillary bed may help abscesses, possibly secondary to averaging of diffusion information
to reveal the most malignant part of a tumor and direct the surgeon from normal surrounding brain tissue (50). Proton MR spectroscopy
or the irradiation, but those studies are complex and still at a develop- can also be a useful adjunct in differentiating tumor from abscess,
mental stage (38,43). as the elevated choline peak that is characteristic of most tumors is
Proton MR spectroscopy has proved to be a useful marker of not present in abscess. Aliphatic peaks of amino acid peaks (alanine
the tumor types and of their aggressiveness in children (Table 7-4). [at 1.5 ppm], acetate [at 1.9 ppm], succinate [at 2.4 ppm], and leu-
Several studies have demonstrated that most tumors have decreased cine, isoleucine, and valine [at 0.9 ppm] [51,52]) are seen instead (see
NAA (neuronal/oligodendrocytic marker), stable creatine except in Chapter 11). A lactate peak is also seen in most pyogenic abscesses (53),
energy failure (tumor necrosis), and increased choline (membrane but lactate is commonly seen in tumors as well and therefore is not
turnover, cellularity) is increased. Choline, lactate, and lipid are com- helpful in making this differentiation. The peaks of alanine, lactate,
monly increased in malignant tumors (44–48). In addition, specific leucine, isoleucine, and valine can be confirmed by the fact that they
tumors may have specific patterns on MR spectroscopy, such as high are inverted on point-resolved spectroscopy sequences using an echo
taurine and creatine in medulloblastomas, high lactate in pilocytic time of 135 to 144 milliseconds (51). Other types of infections, such as
astrocytomas, and high myo-inositol in choroid plexus papillomas cerebritis, are more difficult to differentiate from tumor, as they have
(46). Malignant degeneration is signaled by increasing choline, while variable anatomic, metabolic, diffusion, and perfusion characteristics
effective therapy usually results in decrease of all peaks except lactate that reflect the type and acuity of the infection (44,54). Infections are
(which increases). Recurrent tumor is signified by increasing choline further discussed in Chapter 11.
(Figs. 7-3 and 7-4) (46). Large mass-like, isolated inflammatory lesions may be seen in acute
The differential diagnosis of a brain mass is broad, as any lesion episodes of demyelination such as acute disseminated encephalomy-
with abnormal signal or attenuation and some mass effect may be a elitis or multiple sclerosis (both discussed in Chapter 3). The clinical
tumor. Dysplastic masses, such as gray matter heterotopia or focal context, a careful search for other associated lesions in the brain and
cortical dysplasia, are not a problem anymore with modern imag- spinal cord, MR spectroscopy or advanced imaging such as DTI may
ing (reviewed in Chapter 5); they have characteristic locations, lit- help. If not, the evolution and the response to anti-inflammatory treat-
tle mass effect, low blood volume, do not elicit edema, and do not ment give the final answer. Demyelinating plaques are hypodense on
enhance. Although not truly neoplastic, dysembryoplastic lesions CT images, hypointense on T1, and hyperintense on T2/FLAIR images.
such as a dermoid or epidermoid cyst are included among tumors They are often large and, in the subacute phase, manifest peripheral
and are described in this chapter, as well as other developmental cysts enhancement. They are most easily differentiated from tumors by
such as the sellar/suprasellar Rathke cleft cyst or the posterior fossa their relative lack of mass effect, their incomplete rim of enhancement
dermal cyst. High-definition MRI demonstrates that many cases of (see Fig. 3-42 and discussion in Chapter 3), and by the presence of
aqueductal stenosis actually are due to benign tumors of the mid- deep medullary veins running radially through them, undistorted
brain tegmentum or tectum; their diagnosis are discussed later in this (Fig. 7-6); typically, they will contact the superolateral wall of the lat-
chapter. eral ventricle but displace it minimally or not at all. Long echo MR
Some lesions, however, are difficult to differentiate from tumor. spectroscopy is not useful in this differentiation, as some demyeli-
Abscesses typically develop in a context of infection (see dedicated nating plaques masses may have a large choline peak (Fig. 7-6), and
section in Chapter 11). Although they may appear morphologically increased lactate and lipid due to the inflammatory infiltrates that
similar to malignant necrotic tumors, the enhancing rim typically is accompany them in the acute phase (55,56). Short echo proton spec-

Barkovich_Chap07.indd 643 5/6/2011 9:15:31 PM


644 Pediatric Neuroimaging

FIG. 7-3. Low grade intraparenchymal glial tumor. A. Axial postcontrast


T1-weighted image shows a homogeneous, sharply marginated, nonenhanc-
ing mass (T) deep in the left cerebral hemisphere. B. Proton MR spectra (TE
= 288 milliseconds) in a two dimensional array show that the spectra within
the mass have low NAA and creatine but only slightly elevated choline com-
pared to surrounding voxels containing normal brain. C. Perfusion curves
(relaxivity in the vertical axis, time in the horizontal axis) show similar blood
volume (which is proportional to the loss of signal intensity and decrease in
MR units) in the tumor (T, line 5) and normal brain tissue (N, line 6) in a
similarly located voxel in the contralateral hemisphere. This is characteristic
of most low-grade glial tumors (other than pilocytic astrocytomas).

troscopy may show increased glutamate and glutamine in demyelinat- Infarctions are most easily distinguished from tumors by clini-
ing plaques (57). Dynamic susceptibility-weighted imaging may allow cal presentation; neurologic deficits are characterized in infarcts by
differentiation of these lesions, as demyelinating lesions have low CBV an abrupt onset and in tumors by a slowly progressive or stuttering
and are characterized by radially oriented linear streaks of increased course. In young infants, however, and in older children with infarcts
blood volume coursing through the lesion, representing blood in and that occur in regions that do not cause obvious clinical deficits, the
around the deep medullary veins (58). It has been reported that DTI history is less useful. On imaging, infarcts are distinguished by their
might help in this differentiation, as corticospinal axons in patients location (in a vascular distribution) and involvement of both gray
with tumefactive plaque are few and truncated but in normal posi- and white matter; tumors and infections typically originate in white
tion, while those in patients with tumors show dislocation and/or matter or at the gray-white junction. However, some tumors are pri-
dispersion (59). marily cortical and may have an imaging appearance that is suggestive

Barkovich_Chap07.indd 644 5/6/2011 9:15:31 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 645

FIG. 7-4. Comparison of high-grade intraparenchymal glial tumor (A–C) and pilocytic astrocytoma (D–F). A. Axial postcontrast T1-weighted image
shows a right parietal mass with a central enhancing component (T). B. Proton MR spectrum (TE = 288 milliseconds) shows low NAA, low creatine, but
very high choline in the tumor compared to surrounding normal voxels. Note that the abnormal spectra extend far beyond the enhancing portion of the
mass. C. Perfusion curves (relaxivity in the vertical axis, time in the horizontal axis) show markedly increased blood volume (resulting in decreased signal
and decreased MR units) in the tumor (T, line 2) compared to normal brain tissue (N, line 1) in a similarly located voxel in the contralateral hemisphere.
This is characteristic of high-grade glial neoplasms. D. Axial postcontrast T1-weighted image shows a markedly enhancing mass (white arrows) deep within
the right cerebral hemisphere.

Barkovich_Chap07.indd 645 5/6/2011 9:15:32 PM


646 Pediatric Neuroimaging

FIG. 7-4. (Continued) E. Single voxel proton MR spectrum (TE = 288 milliseconds) from the center of a different pilocytic astrocytoma shows low NAA
and creatine (Cr), but very high choline (Ch) peak. In addition, lactate (L) is present at 1.33 ppm. These MRS finding are identical to those seen in high-
grade gliomas and emphasize the importance of correlating MRS findings with imaging characteristics that, in pediatric tumors, appear to be more specific.
F. Perfusion curves show substantially increased blood volume in the tumor (T, line 3) compared to contralateral normal brain tissue (N, line 2). Thus,
low-grade pilocytic astrocytomas have blood volume characteristics similar to those of high-grade tumors. Such studies should never be interpreted without
imaging correlation.

of acute infarction. DWI is the most useful technique if the infarct perfusion imaging can also be of use in differentiating tumor from
is recent (<8–10 days) by showing significantly reduced diffusion in infarct, as tumors are typically hypervascular, while acute infarcts
an arterial territory, an unusual appearance for tumors, even those are hypovascular (54). On MRS, lactate is high in infarcts during the
characterized by low diffusivity (medulloblastoma, PNET, pineoblas- acute/subacute phase, and the choline peak, characteristic of most
toma, retinoblastoma, ependymoblastoma or germinoma). If differ- tumors, is absent.
entiation is difficult, a follow-up study should be obtained before a Vascular malformations such as a cavernoma are usually not a diag-
biopsy. In an infarction, diffusivity will increase after 8 to 10 days nostic problem due to the characteristic “blooming” appearance on SWI
(less in infants, see Chapter 4), and enhancement will appear; mass (refer to Chapter 12). However, the diagnosis of the cause of an appar-
effect will progressively diminish afterward. Susceptibility-weighted ently isolated spontaneous hemorrhage in a child may be difficult. If no

FIG. 7-5. Use of DWI to separate tumor from abscess. A. Axial FLAIR image demonstrates right frontal heterogeneous cortical-subcortical hyperintensity
(arrows) in a child presenting with seizures. Tumor is a strong possibility. B. Trace image from DWI demonstrates a sharply marginated focus of very high
signal (white arrows) in the medial aspect of the lesion, suggestive of a pus collection; the surrounding white matter shows low signal suggestive of interstitial
edema. C. T1-weighted image postcontrast shows ring enhancement of the nodule; the enhancement extends anteriorly along the falx (arrows) to the vicin-
ity of the foramen cecum. Findings suspicious for nasal dermal tract were also observed; surgery confirmed both the dermal fistula and the abscess.

Barkovich_Chap07.indd 646 5/6/2011 9:15:34 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 647

FIG. 7-6. Imaging characteristics that help to differentiate demyelinating plaques from neoplasia. A. Axial T2-weighted image shows linear venous
structures coursing undistorted through the mass. The presence of undistorted veins running through a mass strongly suggests demyelination, which
occurs in a perivenular location. B. Postcontrast axial T1-weighted image shows enhancement only in the anterior wall (white arrows) of the mass.
Enhancing veins (black arrowheads) are seen coursing through the lesion. C. Gradient echo, echo planar perfusion image shows susceptibility effects
from increased blood volume (hypointensity, arrowheads) in linear bands running through the lesion that correspond to the veins seen in A and B. This
finding is characteristic of demyelinating plaques. D. Proton MR spectrum (TE = 144 milliseconds) shows a large choline peak (Ch) and a lactate peak
(Lac, inverted at TE = 144 milliseconds), but no visible creatine or NAA peak. This spectrum is identical to that of a high-grade tumor and is not a useful
test for differentiation.

Barkovich_Chap07.indd 647 5/6/2011 9:15:35 PM


648 Pediatric Neuroimaging

arteriovenous malformation is demonstrated, and if the characteristic of daughter cells that have migrated from their site of origin and are
appearance of a cavernoma is obscured by the abundance of fresh unable to proliferate (see discussion on the brain tumor biology above).
blood, a hemorrhagic tumor, typically a highly malignant one cannot Therefore, infiltration may be considered a mode of tumor expansion
be easily proved or disproved. Perfusion and spectroscopy are rendered that is independent from grade. In contrast, invasion is a property of
useless by the presence of blood. Careful examination of the adjacent malignant cells; it is the ability of cancer cells to break through normal
parenchyma and even a faint enhancement may reveal the tumoral ori- anatomic barriers. For example, a pilocytic astrocytoma of the pineal
gin of the hemorrhage. If not, follow-up imaging after resolution of the gland will compress but not invade the adjacent callosal splenium, but
blood may be necessary. a pineoblastoma will invade it easily. Any invasive tumor is biologically
Finally, although uncommon, radionecrosis may be a real chal- malignant. The invasion may be local, in the surrounding structures, or
lenge in the follow-up of brain tumors; perfusion imaging may help distant; brain tumor cells can migrate along axons or blood vessels and
in demonstrating that the blood volume is reduced in the radiation travel to distant parts of the brain where reproduction and invasion or
necrosis, while it is typically normal or increased in a tumor (see the infiltration can begin again. In addition, if brain tumors gain access to
more detailed discussion in Chapter 3). the CSF pathways (medulloblastoma or germinomas mostly, but also
Pretherapeutic assessment is the next step, and it is the role of imag- pilomyxoid astrocytomas [PMAs], ependymomas, and many other
ing to provide all the elements of the therapeutic discussion. Therapeu- histologies), they can disseminate throughout the neuroaxis. Assess-
tic options are surgical removal (as complete as possible), radiotherapy ment for CSF spread of tumor should be a part of every pretherapeutic
(except in the first 3 years of life), chemotherapy, or a combination. assessment.
Typically, children with brain tumors are part of a therapeutic protocol If the decision is made to remove the tumor, presurgical assess-
that has specific imaging requirements prior to treatment, during and ment typically entails the acquisition of sequences that are compat-
after completion of treatment. The first feature to look for is whether ible with neuronavigation surgical tools (typically axial, enhanced
the tumor is well demarcated from the surrounding structures. This 3D-GE volumetric T1 sequence such as MPRAGE/SPGR/TFE). The
is the main condition of complete resectability: a poorly demarcated so-called eloquent cortices, such as the sensory-motor area, and major
tumor may be infiltrative or invasive and will not be completely resect- neurocognitive functions such as the language or the memory, may
able. Infiltration does not necessarily relate to the tumor grade. For be located using DTI, fMRI, and the appropriate paradigm (Fig. 7-7A
example, anterior optic pathway gliomas, although classified as grade I, and B) (60). Along the same line, “eloquent” white matter tracts such
may infiltrate the hypothalamus or the basal ganglia, whereas a dorsal as the corticospinal tract or the optic radiations (especially the Meyer
medullary anaplastic ependymoma may not infiltrate any surround- loop along the medial temporal lobe) may be identified with DTI (Fig.
ing structure other than its site of origin at the obex. Infiltration is not 7-7A and C) (61). DTI may also show whether the fibers are simply
always apparent by morphologic imaging alone; additional techniques dislocated by the tumor, or infiltrated and even disrupted (Fig. 7-7C)
such as perfusion imaging, MR spectroscopy, or DTI may be needed (62–65).
to differentiate peritumoral edema from infiltration. In addition, the In general, arteriography is not used for the evaluation of neo-
new understanding of the biology of the brain tumors suggests that the plasms unless a highly vascular lesion is suspected based on vascular
original BTSC, which is responsible for the growth of the tumor, may flow voids seen on MRI or in the unlikely situation that a vascular mal-
be located in the SVZ, while the bulk of the tumor is composed only formation cannot be ruled out by noninvasive studies. For example,

FIG. 7-7. Presurgical functional assessment A. Anatomic imaging (FLAIR) shows large solid and cystic left frontal lobe tumor (arrows) in a child with
refractory epilepsy. The appearance of the tumor is consistent with a pilocytic astrocytoma or a ganglioglioma, of which both can be surgically removed.
However, the proximity to the tumor of the language area (Broca) in this left hemisphere makes the procedure perilous. B. Using a specific paradigm (verb
generation and letter fluency), fMRI showed a strong activation of the ipsilateral Broca area (large black arrow), as well as on the contralateral operculum.
The visual cortex (small black arrows) and surrounding association areas also were activated. C. DTI with color-coded (by convention: red transverse, blue
craniocaudal, green ventrodorsal) FA map shows that the main fiber tracts adjacent to the tumor are displaced rather than infiltrated.

Barkovich_Chap07.indd 648 5/6/2011 9:15:36 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 649

in suspected hemangioblastomas, arteriograms are often obtained to earlier in this chapter, in the section on tumor biology, it appears likely
identify the location of the vascular pedicle supplying the mass, thus that medulloblastoma derives from BTSC, mutated precursor cells
potentially reducing blood loss at the time of surgery. Arteriography that compose the small but actively proliferating component of the
may also be useful if noninvasive studies cannot clearly differentiate a tumor. Given the complex development of the cerebellum with two
dural-based lesion from a parenchymal lesion, a situation that usually different proliferative zones (in the ventricular zone and the external
arises in very large tumors. However, because large tumors sometimes granular layer), different variants of medulloblastoma likely emerge
parasitize blood flow, arteriography can be misleading, demonstrating from distinct cell lineages, each with different cytogenetic abnor-
dural vessels supplying intra-axial lesions or intracerebral vessels feed- malities and a different prognosis (see Gilbertson and Ellison [74] for
ing extra-axial tumors. review).
Follow-up neuroimaging is essential to evaluate the efficacy of In children, medulloblastoma is the second most common brain
the treatment. Typically, an MRI is requested shortly after surgery to tumor (after astrocytoma) and the most common malignant brain
appreciate the extent or the resection, the volume of residual tumor, tumor (15%–20% of intracranial neoplasms in children); it is the most
and any brain injury that might enhance and resemble recurrent common posterior fossa tumor in most series (30%–40% of posterior
tumor on the subsequent follow-up study (66). It also detects eventual fossa neoplasms), being slightly more common than the cerebellar
postsurgical complications such as a significant ischemia or hemor- astrocytoma (8,75,76). They are the most common tumor in the 6 to
rhage in the surgical field, and the ventricular size. Later studies are 11 year age group, comprising about 25% of brain tumors affecting
usually scheduled according to therapeutic protocols that take into children in that age range (77). Males are affected two to four times
account tumor histology as well as the chemotherapeutic and radia- as frequently as females. Medulloblastomas may also occur in adults,
tion protocols. The MR assessment typically uses conventional imag- accounting for 0.4% to 1.0% of adult brain tumors; overall, 14% to
ing. It should be noted that enhancement of the meninges and surgical 30% of medulloblastomas occur in adults (78–80). Medulloblastomas
cavity is almost always seen on contrast-enhanced postoperative MR are seen with increased frequency in patients with basal cell nevus
studies. Enhancement along the surgical cavity reflects damage to the (Gorlin) syndrome (see Chapter 6).
blood–brain barrier and typically slowly resolves over 3 to 4 months; it The duration of symptoms for patients with medulloblastoma
often correlates well with regions of reduced diffusion detected on the is usually short; approximately half will have symptoms for less than
immediate postoperative MRI (66). Dural enhancement can be thin or 1 month prior to diagnosis. The most common symptoms are head-
thick but is usually smooth. It is present over the cerebral convexities aches, vomiting, and nausea. The high incidence of vomiting may be
and along the tentorium and may persist for 20 years or more (67,68). related to growth of the tumor in proximity to the area postrema, the
Nodular meningeal enhancement is more ominous and enhancement emetic center of the brain, which is located near the inferior aspect
of the pia or arachnoid should raise suspicion for subarachnoid tumor of the fourth ventricle. In children less than 1 year of age, increasing
spread. Besides the diagnosis of persistence, recurrence, or dissemina- head size and lethargy are frequent presenting symptoms. In older
tion, MR imaging and spectroscopy can also detect unwanted effects children and adults, ataxia is the symptom that usually initiates medi-
of the therapy such as a radiation necrosis (54,69,70), atrophy, and cal attention. Rarely, paraparesis or symptoms referable to the cauda
the development of vascular malformations such as telangiectasias equina will result from tumor dissemination and necessitate medical
and cavernomas, of vasculitis, and of late secondary induced tumors evaluation (8,81,82). Spinal metastases are present at initial diagnosis
(25,71). in approximately 30% of patients and are a very poor prognostic sign
(83,84). Indeed, extent of disease at diagnosis is the feature most pre-
dictive of prognosis in affected children (85).
POSTERIOR FOSSA TUMORS
By far, the most common posterior fossa tumors of childhood are Pathology
medulloblastomas, astrocytomas (of the cerebellum and brainstem), Approximately two thirds of medulloblastomas in children are located
ATRT, and ependymomas (Table 7-5). Although medulloblastomas, in the vermis, in contrast to adolescents and adults, in whom medullo-
ependymomas, and ATRT may appear to be intraventricular tumors, blastomas are most often found in the cerebellar hemispheres. The
they originate from the parenchyma and grow into the fourth ventri- tumor usually forms a well-defined vermian mass widening the space
cle. They are, therefore, classified as intraparenchymal tumors. Note between the cerebellar tonsils. Anteriorly, it impinges upon the roof
that tumors originating from the brainstem are divided into several of the fourth ventricle and causes partial or complete obstruction
different groups. This division results from the fact that tumors to CSF flow. Posteriorly, it may project into the cisterna magna and
originating from different sites within the brainstem have vastly dif- rarely extend downward to the level of the upper cervical spinal cord
ferent prognoses. Therefore, the diagnosis of “brainstem tumor” is (8,75). Occasionally, the tumor will extend laterally through the lat-
no longer sufficient and a more specific diagnosis should always be eral recesses into the cerebellopontine angle cisterns. Invasion of the
rendered. brainstem is seen in approximately one third of patients, usually result-
ing from direct extension across the fourth ventricle (86). Invasion of
Intraparenchymal Tumors the leptomeninges via dissemination along CSF pathways is frequent,
the literature reporting incidences of up to 100% (87–90). Intracra-
Medulloblastoma nially subarachnoid tumor spread is seen more prominently within
Medulloblastomas are highly malignant tumors composed of very the Sylvian fissure and cisterns of the posterior fossa. Pathologically,
primitive, undifferentiated small, round cells. These undifferentiated subarachnoid tumor appears as small grayish patches of tumor or as a
cells were given the name “medulloblast” by Bailey and Cushing in the continuous “frosting” of tumor on the pia. Retrograde spread through
early 1900s, hence the name medulloblastoma (72). Together with the the aqueduct into the lateral and third ventricles can occur, as well.
primitive neuroectodermal tumors (PNETs) and the ATRT, they form From any of these sites, tumor may reinvade the brain parenchyma.
the group that the World Health Organization calls embryonal tumors Drop metastases to the spinal subarachnoid space and cauda equina
(73); all are highly malignant (WHO grade IV). As was mentioned are seen in approximately 40% of patients, most commonly at the tho-

Barkovich_Chap07.indd 649 5/6/2011 9:15:39 PM


650 Pediatric Neuroimaging

TABLE 7-5 Posterior Fossa Tumors in Children


Intraparenchymal Tumors
Medulloblastoma Usually in 4th ventricle
T2 intensity of GM, restricted diffusion
Astrocytoma Usually hemispheric
T2 hyperintense
Ependymoma Exophytic toward cisterns
Atypical teratoid/rhabdoid tumor Large heterogeneous mass with necrosis, cysts
Solid areas have T2 intensity of GM
Brainstem tumors Ventral: diffuse infiltrating pontine glioma
Medulla, midbrain, peduncles: pilocytic
Teratoma Most common in young infants, heterogeneous, contains fat
Hemangioblastoma (uncommon) Cyst with intensely vascular mural nodule
Extraparenchymal tumors (uncommon)
Dermal cyst Usually enhances (infection)
Dermal tract to skin possible
Dermoid Short T1, saturates with FatSat
Epidermoid Isointense with CSF on T1/T2, not FLAIR
Internal heterogeneity
Restricted diffusion
Enterogenous (enteric) cysts Cyst of variable intensity
Ventral to brainstem
Teratoma Heterogeneous, contains fat
Schwannoma Along cranial nerves, consider NF2
Meningioma Dural based; consider NF2
Skull base tumors Bone involvement

racic and lumbosacral levels (87–90). Rarely, intramedullary metasta- cases, while glial differentiation is rare, and ependymal differentiation
ses have been reported, presumably as a result of tumor dissemination is so exceptional that it raises suspicion regarding the diagnosis (74).
into the central canal of the spinal cord. Systemic metastases are rare
and most commonly develop after tumor recurrence. The skeleton is Imaging Studies
the most common site of involvement followed by lymph nodes and On CT, a typical medulloblastoma classically appears as a well-defined,
lung (87). hyperdense tumor of the cerebellar vermis (Fig. 7-8) or vermis and
The latest WHO classification of the tumors of the CNS describes hemisphere. Isolated involvement of the hemisphere is much less com-
several subtypes, all grade IV (73,91): the classic medulloblastoma mon and is suggestive of the desmoplastic variant in adolescents (78–
(65%–75%), desmoplastic medulloblastoma (7%), medulloblastoma 80,97) and the extensive nodularity variant in infants (92,93); these
with extensive nodularity (3%), anaplastic medulloblastoma (10%– laterally situated tumors may be exophytic and mimic tumors of the
22%), and large cell medulloblastoma (2%–4%) (73,74,91). Histologi- cerebellopontine angle (98). As a result of their composition of small,
cally, more than half of the medulloblastomas are of the classic type, round cells with a high nuclear to cytoplasmic ratio, medulloblastomas
composed entirely of completely undifferentiated cells, and typically are almost always hyperdense or isodense compared to surrounding
located in the inferior vermis. Desmoplastic medulloblastomas are white matter prior to the administration of intravenous contrast. The
detected more commonly in the second decade, are more commonly lesion usually appears well demarcated from the surrounding paren-
located in the cerebellar hemispheres; they exhibit a striking nodu- chyma. Mild to moderate edema surrounds the tumor in approxi-
larity with meningeal hyperplasia, and have a more benign course mately 90% of patients. Hydrocephalus is present in approximately
than the classic type. Medulloblastomas with extensive nodularity 95% of patients at the time of presentation. Because the tumor usually
are also located in the cerebellar hemispheres of infants younger than develops in the inferior portion of the vermis, it is often capped by the
3 years; their prognosis appears favorable (92,93). Anaplastic and large deformed lumen of the upper part of the fourth ventricle; the aqueduct
cell medulloblastomas have the poorest prognosis (94–96). Any variant may be dilated. Enhancement is seen in greater than 90% of medullo-
of medulloblastomas may develop myogenic or melanotic histological blastomas, most commonly diffuse, but sometimes irregular or even
features (73,74,91); neuronal differentiation may occur in 7% of the absent (10,99).

Barkovich_Chap07.indd 650 5/6/2011 9:15:39 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 651

FIG. 7-8. Typical medulloblastoma. A. Noncontrast CT image shows a well demarcated, rather homo-
geneous rounded mass (arrows) in the center of the posterior fossa, with increased attenuation compared
with cerebellar gray matter. This appearance and location are characteristic of classic medulloblastoma.
A few small necrotic cysts are common; punctate calcification may be seen as well. B. Sagittal T1 image
demonstrates the tumor (white arrows) filling the fourth ventricle. Its inferior margins (black arrow) are
somewhat blurred. Signal intensity of the tumor is approximately that of the supratentorial gray matter.
C. Coronal T2 image shows the signal of the tumor (T) to be close to that of the gray matter. Ventricular
CSF surrounds most of the tumor. Note the tonsillar herniation, and the tilt of the head (torticollis).
D. Axial FLAIR image best shows the hypointense small foci of necrosis (black arrows). E. Diffusion-
weighted image shows tumor hyperintensity, indicating reduced free water movement. This, like the
high attenuation on CT, reflects the high cellularity typical of medulloblastoma. F. Hypointensity of
the tumor (white arrows) on the calculated Dav map confirms the reduced diffusivity. The small areas of
hyperintensity within the tumor are necrotic or cystic foci. G. Postcontrast sagittal T1WI image shows
incomplete, irregular enhancement of the tumor.

Cysts or calcifications are found in 60% of CT studies in medullo- isodense to hyperdense on precontrast studies (Fig. 7-8A) (10). In the
blastomas, including calcification in up to 20% and cystic or necrotic, authors’ experience, the density of the tumor on the precontrast CT
nonenhancing regions in close to 50% (99,100). The presence of scan is a very reliable means of differentiating these tumors. It is impor-
intratumoral hemorrhage is very uncommon. Although the presence tant to look for falcine calcification if CT is performed. If calcification
of cystic, nonenhancing regions within the tumor may sway the neu- of the falx cerebri is seen prior to shunting or the initiation of radiation
roradiologist toward the diagnosis of pilocytic astrocytoma, the solid therapy, the possibility of medulloblastoma occurring in the setting of
portions of pilocytic astrocytomas are usually hypodense prior to the basal cell nevus syndrome or Gorlin syndrome, described in Chapter 6,
administration of IV contrast, whereas medulloblastomas are mostly should be considered and other manifestations of the syndrome should

Barkovich_Chap07.indd 651 5/6/2011 9:15:39 PM


652 Pediatric Neuroimaging

FIG. 7-9. Patchy enhancement in medulloblastomas. Postcontrast axial T1-weighted images (A and B) show varying degrees of enhancement. The arrow
in (B) points to a small tumor cyst.

be sought (101). As patients with basal cell nevus syndrome have a pro- values in medulloblastoma, reflecting its malignant nature (45,47,107–
pensity to develop basal cell carcinomas in irradiated fields, the pres- 109). In addition spectra with short TE show a significant taurine peak,
ence of the syndrome may lead to changes in therapy. which is characteristic of medulloblastomas (45,110).
On MRI, the appearance of medulloblastomas is variable. Tumor CSF spread of tumor is very poorly evaluated by noncontrast MRI.
location and patient age are the most important factors in prospec- Therefore, paramagnetic contrast should be used in the evaluation of
tively making the correct diagnosis. The tumors are most commonly these patients both prior to and after resection of the tumor. The most
situated within the inferior vermis (Fig. 7-8B) and can sometimes be common locations for intracranial metastases are the vermian and
seen originating from the inferior medullary velum: a midline infe- basilar cisterns, subependymal region of the lateral ventricles, and the
rior vermian tumor bulging ventrally in the fourth ventricle in a child subfrontal region (Figs. 7-11 and 7-12). Intraventricular (subependy-
from 5 to 10 year old is likely to be a medulloblastoma. The most mal) metastases frequently show limited enhancement (Fig. 7-12D);
common appearance is that of a round, slightly lobulated mass, isoin- this should not cause confusion in the proper clinical setting, as
tense to normal gray matter on T1-weighted images. T2-weighted periventricular nodular heterotopia (see Chapter 5) are rarely seen as
sequences typically reveal a heterogeneous mass in which the solid incidental findings. MRI is the primary imaging method to evaluate for
portions are hypointense or isointense compared to gray matter (Fig. the presence of drop metastases to the spinal cord or the cauda equina
7-8C). The small round cells also result in the solid regions of the and has, properly, replaced plain-film and CT myelography (35,36).
tumor being hyperintense compared with normal cerebellar tissue on MR techniques for imaging the spine are discussed in Chapter 1. MRI
DWI and hypointense on diffusivity maps (average diffusivity [Dav] of also seems to be more sensitive than CSF cytology in the detection of
solid portions of medulloblastoma is between 0.5 and 0.9 × 10−3 mm2/s subarachnoid spread of tumor in primary brain tumors, whereas CSF
[102]) (Figs. 7-8E, F, and 7-9 D, E) (103–105). The signal intensities cytology is more sensitive in the detection of hematologic tumors such
presumably relate to reduced amount of free water within the tumor. as leukemia and lymphoma (111). On contrast-enhanced MRI, drop
Less free water results in a shortened T2 relaxation time and reduced metastases may appear as a smooth enhancing coating of the spinal
diffusivity and, hence, lower signal intensity on T2-weighted and Dav cord or as brightly enhancing foci in the extramedullary, intradural
images. Heterogeneity probably results from the aforementioned and, occasionally, intramedullary space (Figs. 7-12, 10–11 and see dis-
cysts and calcification (106). The enhancement pattern of the tumors cussion in Chapter 10). The most common location is along the poste-
after intravenous infusion of paramagnetic contrast is variable; rior surface of the spinal cord in the thoracolumbar regions (along the
enhancement may be patchy or uniform (Figs. 7-8 to 7-10). Signal arachnoid septum that attaches the dorsal cord to the posterior dura,
and enhancement characteristics of hemispheric medulloblastomas, known as the septum posticum) (Fig. 7-12F); the caudalmost aspect
seen in adolescents and adults, are similar to those of fourth ventricu- of the thecal sac is also frequently involved. In the first few weeks after
lar medulloblastomas seen in children. The cerebellar hemispheric posterior fossa craniectomy, however, artifacts (presumably from
tumors often arise in the periphery of the hemisphere. Desmoplastic dependent subarachnoid blood along the septum posticum, spinal
tumors may incite desmoplastic reaction in the overlying meninges, subdural collections, or even from “leakage” of contrast into the suba-
resulting in leptomeningeal enhancement that mimics a meningioma rachnoid and subdural space) are common (112,113). These artifacts
(96) (Fig 7-10). It is important to be cautious in interpreting tumors can be impossible to differentiate from CSF spread of tumor. These prob-
with this appearance in older children (second decade) and young lems are best avoided by staging the tumor preoperatively with precontrast
adults. and postcontrast scans of the brain and spine. Alternatively, tumor spread
Proton spectroscopy, discussed in the introduction to this chapter, to the spine can be assessed after waiting for two weeks after surgery before
typically shows high choline, low creatine, low NAA, and high lactate imaging (114).

Barkovich_Chap07.indd 652 5/6/2011 9:15:40 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 653

FIG. 7-10. Desmoplastic medulloblastoma in a 15-year-old adolescent. A. Sagittal T1-weighted image shows a large rounded, well demarcated tumor
(arrows) in the superior aspect of the cerebellar hemisphere. B. Axial T2-weighted image demonstrates the tumor (arrows) to be slightly hyperintense
compared with cortex. Note the anterolateral superficial, almost exophytic location of the tumor in the left hemisphere; the pons and fourth ventricle are
displaced to the right. C. FLAIR image best depicts the associated hyperintense vasogenic edema. D. Diffusion-weighted image shows tumor hyperintensity,
indicating reduced free water movement. E. Hypointensity of the tumor (arrows) on a calculated diffusivity map confirms the reduced diffusion, which is
highly suggestive of medulloblastoma. F. Postcontrast image shows dense, homogeneous enhancement of the mass.

Medulloblastomas rarely disseminate hematogeneously since sys- techniques have dramatically different biological features (117,118).
temic chemotherapy has been added to the therapeutic regimen (115). Children with these tumors, called “atypical teratoid/rhabdoid
When extra-CNS spread occurs, it may be seen years after initial treat- tumors of infancy and childhood,” present at a younger age than true
ment, with a median interval of 12 to 32 months (116). As bone is medulloblastomas, with the median age at diagnosis being 2 to 4 years
the most common site for extraneural metastases, neuroimaging (118–121) (median age for diagnosis of medulloblastoma is about
follow-up studies will usually show metastases in the diploic space of 6 years). They belong to the same group of WHO grade IV embryonal
the calvarium or in the marrow cavities of the vertebrae (114). These CNS tumors as the medulloblastoma (73). The importance of distin-
skeletal metastases are typically very radiodense on plain films and CT; guishing ATRT from medulloblastoma lies in the lack of response of
they are hypointense on T1-weighted images and show enhancement ATRT to standard therapy for medulloblastomas. As a result of this
after administration of paramagnetic contrast (114). Signal intensity is lack of responsiveness, the majority of patients with ATRT succumb
variable on T2-weighted images. Lymph nodes are another common within 1 year of diagnosis, in contrast to those with medulloblastoma,
site of extraneural metastases in children, and may be detected on stud- who frequently respond to chemotherapy (118,119,121,122). ATRT
ies of the spine (96). can be distinguished from medulloblastomas by routine and special
microscopic techniques (118). However, many are nonetheless ini-
Atypical Teratoid/Rhabdoid Tumor of Infancy tially diagnosed as medulloblastomas or PNETs because rhabdoid
and Childhood cells, the histologic hallmarks of ATRT, compose only a minority of
Over the past 10 to 15 years, it has become apparent that some tumors the neoplastic cells; thus, the specimens examined by the patholo-
diagnosed as medulloblastomas by classical pathologic and radiologic gist may be indistinguishable from PNETs. From the reports avail-

Barkovich_Chap07.indd 653 5/6/2011 9:15:41 PM


654 Pediatric Neuroimaging

FIG. 7-11. Typical medulloblastoma with subarachnoid tumor spread. A. Sagittal T1-weighted image shows hydrocephalus and a hypointense mass
(arrows) obstructing the fourth ventricle. B. Axial T2-weighted image shows that the mass is nearly isointense to gray matter. C and D. Axial postcontrast
images show the main tumor enhancing homogeneously. Multiple foci of tumor (arrows) that has spread through the subarachnoid pathways are seen both
infratentorially and supratentorially.

Barkovich_Chap07.indd 654 5/6/2011 9:15:43 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 655

FIG. 7-12. Metastatic medulloblastoma. A. Postcontrast T1-weighted image shows enhancement around the periphery of the pons (small arrowheads), in
the cerebellar fissures (small arrows), and coating the cisternal portion of the right fifth cranial nerve (large arrow). B. Postcontrast T1-weighted image shows
a large tumor mass (arrows) in the region of the optic chiasm and a smaller deposit (arrowhead) in the right sylvian fissure. The chiasmal region is a com-
mon location for metastatic deposits because radiation ports try to miss the optic nerves and chiasm. C. Postcontrast T1-weighted image shows metastatic
deposits (arrows) in both sylvian fissures. D. Postcontrast T1-weighted image shows multiple intraventricular metastatic deposits (arrows) and a region of
subarachnoid tumor (arrowhead) in the cingulate gyrus. Intraventricular metastases frequently do not enhance.

Barkovich_Chap07.indd 655 5/6/2011 9:15:44 PM


656 Pediatric Neuroimaging

FIG. 7-12. (Continued) E and F. Postcontrast sagittal T1-weighted images show enhancing tumor coating the spinal cord. A focal lesion (arrows) at the C-3
level appears to be invading the spinal cord. Note the hyperintensity of the vertebral bodies secondary to prior radiation therapy.

able (120,121,123–125), it would appear that these tumors are about heterogeneous. Proton spectroscopy shows elevated choline and
equally distributed between the supratentorial space (44%) and the reduced NAA, although quantification of these changes has not yet
posterior fossa (44%) including the cerebellum (especially the vermis), been reported (121); no data are available yet regarding the pres-
midbrain, and cerebellopontine angle cisterns. Both supratentorial and ence or size of taurine peaks. In contrast to medulloblastomas and
infratentorial structures are involved in approximately 10%. The sep- ependymomas, ATRT seem to originate as commonly from the cer-
tum pellucidum, pineal gland, and spinal cord (2%) may be involved ebellar hemispheres as from the midline/walls of the fourth ventricle
as well. However, infratentorial ATRT occur mostly in patients less (Fig. 7-13) (130). The diagnosis of atypical teratoid/rhabdoid tumor
than 3 years old, and supratentorial sites are most common in chil- should be included in the differential diagnosis of tumors with the
dren older than 3 years (118,120–122,125–127). As with medulloblas- neuroimaging appearance of small, round-cell tumors in both the
tomas, the tumor may spread through the subarachnoid space when supratentorial and infratentorial compartments, particularly when
it arises in the ventricles or cisterns (119,128); indeed, Meyers et al. the tumor is very heterogeneous and the child is below the age of
(121) reported subarachnoid spread in 24% of their series at the time 4 years.
of diagnosis.
Neuroimaging characteristics (Fig. 7-13) can be similar to Cerebellar Astrocytomas
those of medulloblastomas in the posterior fossa and to germ-cell Astrocytomas are the most common brain tumor in children, account-
tumors and PNETs when arising in the supratentorial compart- ing for 40% to 50% of primary pediatric intracranial neoplasms
ment (129). On noncontrast CT, the solid portions of the tumor (8,10,11,75). Approximately 60% of astrocytomas are located in the
demonstrate attenuation similar to slightly higher than that of gray posterior fossa with 40% in the cerebellum and 20% in the brainstem.
matter, with calcification (130), hemorrhage, cysts, and necrosis Depending on their histology and grading, various types of astrocy-
(120,121,124,125). On MRI, solid portions of the tumor are isoin- tomas are identified in the WHO classification (Table 7-6). Most cere-
tense to gray matter on T1-weighted images and isointense to slightly bellar astrocytomas in children are of a specific histological type, called
hyperintense compared to gray matter on T2-weighted MR images juvenile pilocytic astrocytoma (JPA), which is considered a unique
(120,125,126,130). DWI shows reduced diffusivity in the solid por- tumor and is classified as grade I by the World Health Organization.
tions of the tumor (121,125) with Dav values that overlap with those JPAs are the most benign astroglial tumors of the CNS (76). Malignant
of medulloblastoma (102). As on CT, necrosis, cysts, and hemorrhage astrocytomas of the cerebellum, even glioblastomas (grade IV astroglial
are identified in up to 50% (120,121,130,131), giving an appearance neoplasms), can and do occur in children, however (133), and have a
of striking heterogeneity (132); medulloblastomas are generally less very poor prognosis (134).

Barkovich_Chap07.indd 656 5/6/2011 9:15:46 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 657

FIG. 7-13. Atypical teratoid/rhabdoid


tumor of infancy. A. Axial CT shows
a heterogeneous, partly necrotic high
attenuation mass (arrows) in the supe-
rior vermis. B. Sagittal T2-weighted
image demonstrates a huge mass in
the superior vermis, fourth ventricle,
and supravermian/quadrigeminal cis-
tern. The solid portions of the mass
are slightly hyperintense to gray mat-
ter; it has large necrotic portions (n)
and infiltrates the tectal plate. C. DWI
shows hyperintensity (reduced diffusiv-
ity) in the solid portions of the tumor.
D. Postcontrast sagittal T1-weighted
image shows partial enhancement
(arrows) of the mass.

JPAs are found with equal frequency in males and females and,
TABLE 7-6 WHO Grades I is Benign, overall, compose about 70% of pediatric cerebellar astrocytomas (135).
Grades II to IV Reflect The peak incidence of these tumors is from birth to 9 years of age (136),
but they remain common until about age 15 (77). Anaplastic astro-
Increasing Degrees of cytomas (∼ 5%–15% of pediatric cerebellar astrocytomas) are more
Malignancy common in older children, usually in the second decade (76,77,136).
Glioblastomas of the cerebellum are very uncommon and can present
Pilocytic astrocytoma WHO grade I at any age (18,133). In general, patients with cerebellar astrocytomas,
regardless of the histologic characteristics, present with early morning
Pilomyxoid astrocytoma WHO grade II
headache and vomiting, waxing and waning over a period of months
Pleomorphic xanthoastrocytoma WHO grade II and finally becoming persistent and acute (8). Cerebellar signs, such as
Diffuse astrocytoma (any type) WHO grade II truncal ataxia or dysdiadochokinesia, are frequently present. Prognosis
is related to extent of resection (137); therefore, best prognosis is asso-
Anaplastic astrocytoma WHO grade III ciated with single hemispheric location, small size, and cystic tumors
Glioblastoma (any type) WHO grade IV with posterior mural nodules (137). Patients with cystic JPAs have an
excellent prognosis, with the 25-year survival rate being close to 90%
After Louis et al. (73). (11). The 25% of patients with the solid cerebellar astrocytoma have a
more guarded prognosis, with a 25-year survival rate of approximately

Barkovich_Chap07.indd 657 5/6/2011 9:15:46 PM


658 Pediatric Neuroimaging

40% (11). With either variety, malignant transformation of benign cer- other locations (144), but it may occur and be massive (145). Pilocytic
ebellar astrocytomas is exceedingly rare, but does occur (138). Gross astrocytomas differ from most low-grade astrocytomas histologically
total resection of pilocytic astrocytomas by surgery is usually curative. in that they are very vascular (143) and the endothelial cells within the
Extension into the brainstem, seen in up to 25% of cerebellar astrocy- tumor have open tight junctions and fenestrations (146). The imaging
tomas, renders complete resection difficult and reduces long-term sur- implication of these “holes” in the vascular walls is a very profound
vival (139). Tumors that are not surgically accessible may be treatable enhancement of the tumors (see below).
by stereotactic radiosurgery (140). A pilomyxoid variant (PMA) with monomorphous features, lack
of Rosenthal fibers, and myxoid background has been recently reported
Pathology (143); it is classified as a WHO grade II astrocytoma (73), less benign
Cerebellar astrocytomas of childhood may originate in the midline or that the JPA. PMA are predominantly located in the forebrain, espe-
in the cerebellar hemispheres; the frequency of these locations has been cially in the suprasellar region (57%) and only rarely in the cerebellum
debated (75,76,138,139,141). The tumor is usually large at the time of (10%) or the fourth ventricle (5%) (147).
presentation, with an average diameter of greater than 5 cm (75,76).
Cerebellar astrocytomas can be cystic, solid, or solid with a necrotic Imaging Studies
center (8,75,142). Most frequently, the cystic lesions have a tumor nod- The typical appearance of cerebellar astrocytoma is that of a large
ule (“mural” nodule) found within the cyst wall; the remainder of the vermian or hemispheric tumor that is predominantly cystic. The solid
cyst wall consists of nonneoplastic, compressed cerebellar tissue. Cystic portion of the tumor is usually isodense to hypodense to normal
astrocytomas with mural nodules account for approximately half of all white matter on noncontrast CT, with pilocytic tumors nearly always
pediatric cerebellar astrocytomas (8,10,75,142). Another 40% to 45% hypodense; high-grade tumors may be hyperdense. Contrast enhance-
of cerebellar astrocytomas are composed of a rim of solid tumor with ment is usually irregular, half of the lesions showing mixed attenua-
a cyst-like, necrotic center. Necrosis of the tumor center is sometimes tion (10). In addition to the heterogeneities caused by cysts and tumor
incomplete and, on occasion, results in a polycystic appearance (142). necrosis, heterogeneous enhancement is seen in the solid portion of
Non-necrotic solid tumors are seen in less than 10%. Some authors the tumor; some degree of contrast enhancement is virtually always
describe a fourth type, with a mural nodule and an enhancing cyst wall present (10). Tumors of pilocytic cytology show intense enhancement
(139). The gross appearance does not correlate with histologic type of their solid portions (148).
(136). When the tumor is cystic with a mural nodule, the cyst is round
Histologic exam of pilocytic astrocytomas is characterized by a or oval, whereas the mural nodule may be round, oval, or plaque-like.
combination of piloid tissue (dense sheets of elongated bipolar cells After infusion of IV contrast, the nodule shows an intense, homoge-
and an abundance of Rosenthal fibers) and loose glial tissue (com- neous tumor blush (Fig. 7-14). The wall of the cyst may appear slightly
posed of multipolar protoplasmic astrocytes and eosinophilic granular hyperdense on CT because of the compressed cerebellar tissue; if
bodies) (143). Calcification is seen on histologic examination in 20% of it does not enhance, pathology of the cyst wall almost never reveals
cerebellar astrocytomas, most commonly in the solid variety. Tumoral tumor (11). If the wall of the cyst enhances, tumor is usually found.
hemorrhage is very rare and probably more so in the cerebellum than in When the cyst results from necrosis of a solid astrocytoma, it may be

FIG. 7-14. CT of JPA of the cerebellum. A. Images from a noncontrast CT through the cerebellum. There is a cystic lesion involving the left cerebellar
hemisphere (open arrows). A solid nodule of tumor, which is of lower attenuation than surrounding cerebellum, is located within the medial aspect of the
cyst (closed arrow). B. After infusion of iodinated contrast, the nodule in the medial wall of the cyst is seen to uniformly enhance (closed arrows). The cyst
wall does not enhance. The slight increased intensity seen in the ventral wall of the cyst (open arrows) results from compression of adjacent brain tissue.

Barkovich_Chap07.indd 658 5/6/2011 9:15:47 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 659

unilocular or multilocular. A well-defined mural nodule is not seen; size from small (Fig. 7-15) to very large (Fig. 7-16). Solid and cystic
instead, tumor enhancement surrounds the cyst and extends into the components are identified, just as on CT. In general, solid portions
cerebellum (10,11,149). Solid tumors are usually round to oval, lobu- appear as low-signal intensity masses (although not as low as CSF)
lated, and fairly well-defined masses that are isointense to hypointense on T1-weighted sequences and as high-intensity masses (although
precontrast and show heterogeneous to homogeneous enhancement. not as high as CSF) on T2-weighted and FLAIR sequences (Figs. 7-15
Rarely, no enhancement is discernible; nonenhancing tumors are to 7-17). Higher grade tumors may manifest lower signal intensity
essentially never of pilocytic histology. on T2-weighted images, thereby mimicking medulloblastoma (133).
The MR appearance of cerebellar astrocytomas is variable, Note that the signal intensity of pilocytic tumors is not as dramatically
depending upon the gross pathological appearance. Tumors vary in high on FLAIR images (Fig. 7-17) and therefore FLAIR images are less

FIG. 7-15. Small hemispheric astrocytoma; pathology revealed JPA. A. Sagittal postcontrast T1-weighted image shows a small hemispheric tumor (arrows)
with a small amount of central enhancement. B. Axial T2-weighted image shows the tumor (arrows) sitting in the cerebellar white matter abutting the fourth
ventricle. The central hyperintensity represents different histology of tumor but, because that region enhances, was not felt to represent necrosis. C. Axial
FLAIR image also reveals several areas of different signal intensity in the center of the tumor. D. Axial postcontrast T1-weighted image shows that the areas
of hyperintensity on the T2 and FLAIR images are the areas that enhance. These were felt to be areas of pilocytic tumor.

Barkovich_Chap07.indd 659 5/6/2011 9:15:48 PM


660 Pediatric Neuroimaging

FIG. 7-16. Large cystic JPA. A. Sagittal T1-weighted image


shows a large cystic mass involving the cerebellar hemisphere.
The more anterior, homogeneous hypointense component
(white arrowheads) was cystic while the posterior heteroge-
neous component (white arrows) was solid. B. Postcontrast
T2-weighted image through the posterior fossa shows the large
heterogeneous mass with cystic (c) and solid (s) components.
Note that the solid component of this pilocytic astrocytoma is
very hyperintense compared with brain parenchyma. This is typ-
ical of pilocytic astrocytomas. C. Postcontrast axial T1-weighted
image shows that the solid component (s) brightly enhances.
The walls of the cysts (c) do not enhance, indicating the absence
of tumor in the cyst walls.

useful than conventional T2-weighted images for differentiating pilo- medulloblastomas and ependymomas, the inexperienced physician may
cytic astrocytomas from higher grade tumors. Solid portions of tumor wish to use calculated average diffusivity (Dav or ADC) values to help
will enhance with paramagnetic contrast (Fig. 7-16) in an identical in this differentiation. The solid components of pilocytic astrocytomas
fashion to their enhancement with iodinated contrast on CT. As with will usually have higher Dav values (typically 1.25–1.95 × 10−3 mm2/s)
CT, it is possible to distinguish predominantly solid tumors with cen- than the more cellular ependymomas and especially medulloblastomas
tral necrosis (Fig. 7-17) from cystic tumors (Fig. 7-16) as necrosis tends (102). However, there may be more overlap with other types of gliomas
to have irregular (Fig. 7-17), rather than smooth (Fig. 7-16), margins. and other less typical tumor pathologies so, as always, it is important to
Also as with CT, enhancement of the cyst wall indicates the presence take into account all imaging characteristics as well as patient age.
of tumor. It is sometimes difficult to differentiate a solid from a cys- As discussed in the introduction to this chapter, the proton MR
tic mass with noncontrast MRI when the solid portion of the tumor is spectra of cerebellar astrocytomas vary considerably depending upon
homogeneous. A tumor should not be assumed cystic merely because it the histologic characteristics of the tumor (45,47,107–109). Interest-
demonstrates homogeneous low-signal intensity on T1-weighted and ingly, however, significant differences have been reported in the spectral
homogeneous high-signal intensity on T2-weighted images, even if it composition between the cerebellar and the suprasellar pilocytic astrocy-
does not enhance. Further evidence, such as diffusivity equal to CSF, tomas, with significantly higher myo-inositol and glutamate/glutamine
wave forms from fluid pulsations or fluid-fluid levels should be sought peaks in the suprasellar tumors, and a trend toward lower creatine in the
in order to reliably make this differentiation. Although the T2 charac- cerebellar tumors (107,150). Such differences could be related to the fact
teristics will usually allow differentiation of pilocytic astrocytomas from that, although JPAs are characterized by a similar histological appear-

Barkovich_Chap07.indd 660 5/6/2011 9:15:50 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 661

FIG. 7-17. Solid JPA with central necrosis. A. Sagittal


T2-weighted image shows large hyperintense cerebellar mass
with a hyperintense solid periphery (s) and necrotic central foci
of hyperintensity (n). Obstructive hydrocephalus is present,
causing stretching of the corpus callosum around the dilated
lateral ventricles and ballooning of the anterior recesses of the
third ventricle. B. Coronal FLAIR image shows that the solid
periphery of the mass is hyperintense compared to gray matter
but not as hyperintense as is seen on T2-weighted images such
as A. The necrotic center (n) is relatively hypointense. C. Post-
contrast axial T1-weighted image shows that the solid periph-
eral portion of the tumor (white arrows) enhances uniformly, as
characteristic of pilocytic astrocytomas. The central nonenhanc-
ing region (black arrowheads) is necrotic tumor.

ance regardless of their location, the proliferating tumor stem cells that of the fourth decade. Patients with ependymomas of the posterior fossa
produce the bulk of the tumor are location-specific and, therefore, are frequently present with a long clinical history. The delay in diagno-
cytogenetically different in different regions (see section on “Changing sis probably results from the insidious onset of symptoms. Nearly all
Concepts of Brain Tumor Biology” earlier in this chapter) (32). affected children have nausea and vomiting resulting from the hydro-
cephalus and increased intracranial pressure (from tumor obstructing
Ependymomas the fourth ventricle) develops in 90% of patients (8,81). Other signs and
Ependymomas constitute 8% to 12% of primary CNS neoplasms in chil- symptoms include torticollis and ataxia (when the tumor arises near
dren and 10% to 20% of posterior fossa tumors in children; their rela- the obex) and lower cranial neuropathies (when the tumor arises in the
tive frequency diminishes as children grow older, but affected children lateral recesses and grows into the cerebellopontine angle) (151).
can present at any age (9,77). In childhood, intracranial ependymomas
are more commonly infratentorial (70%) than supratentorial (30%) Pathology
and more commonly intracranial than intraspinal (see Chapter 10) Ependymomas were at one time assumed to derive from differenti-
(9,151). There is a slightly increased preponderance in males (152– ated ependymal cells that line the floor and roof of the fourth ventricle
154). Ependymomas of the posterior fossa have two age peaks, the first or from ependymal cell rests found posterior to the occipital horns,
occurring between the ages of 1 and 5 years and the second in the middle and along the tela choroidea (76). However, new evidence indicates

Barkovich_Chap07.indd 661 5/6/2011 9:15:50 PM


662 Pediatric Neuroimaging

that, like other CNS tumors, ependymomas result from mutated stem posterior fossa mass (Figs. 7-18 to 7-20), with punctate calcifications
cells, and those that produce ependymomas have been shown to be small cysts, and moderate, heterogeneous enhancement after intrave-
the radial glia cells (33). In the cerebral hemispheres, radial glia can be nous contrast administration (10,11). Extension of the tumor from the
found throughout the thickness of the brain mantle, explaining why lateral recess into the cerebellopontine angle or through the foramen
supratentorial ependymomas are characteristically extraventricular magnum into the cervical spinal canal further supports the diagnosis
and may even be strictly intracortical (155). of ependymoma. Small lucencies are seen within the tumor in approxi-
Most ependymomas are solid in nature, with calcification seen mately 15% of cases and nearly 50% will exhibit multifocal, small
in up to 50% of posterior fossa ependymomas and cysts in approxi- calcifications (Fig. 7-18); large, clump-like calcifications are occasion-
mately 20% (152). Approximately 20% of ependymomas are very soft ally seen. Intratumoral hemorrhage is found in approximately 10% of
and deformable. Therefore, in contradistinction to most brain tumors patients (Fig. 7-20) (10,154).
(which grow as steadily enlarging masses), ependymomas can insinu- On MRI, ependymomas may be homogeneous or heterogeneous.
ate themselves through the subarachnoid spaces and around blood ves- T1-weighted images commonly reveal a slightly hypointense mass with
sels and nerves, often surrounding and engulfing structures. As a result respect to brain parenchyma, sometimes with foci of marked hypoin-
of their adherence to surrounding brain and their invasive growth pat- tensity. T2-weighted images reveal a mass that is usually isointense
tern, ependymomas are quite difficult to cure; removal of the entire with gray matter (Fig. 7-19); foci of high intensity (necrotic areas or
tumor is uncommon and the recurrence rate is high (8,81). cysts) and low intensity (calcifications or hemorrhage) may be present
Posterior fossa ependymomas originate from the sides of the within the tumor mass (Fig. 7-20). Fluid-fluid levels can sometimes
medullary portion of the fourth ventricle. They are classified as WHO be seen within the cysts. Because of the marked variation in appear-
grade II (cellular type with its variants) or III (anaplastic) (156), but ance of the solid portion of the tumor, the diagnosis of ependymoma
whether this correlates with outcome is not clear (157). (The term is difficult to make on the basis of signal characteristics alone. The
ependymoblastoma does not describe a variety of ependymoma, but a most important imaging finding to identify ependymomas is extension
PNET [an embryonal tumor classified as WHO grade IV] with a “dif- of the tumor through the fourth ventricular outflow foramina. Exten-
ferentiation potential solely along ependymal lines” [156]). Posterior sion of tumor to the cisterna magna and through the foramen mag-
fossa ependymomas do not invade the medulla, instead form exophytic num into the dorsal cervical subarachnoid space, behind the cervical
masses that grow out of the ventricle into the surrounding cisterns. spinal cord (Fig. 7-18), is quite characteristic and very nearly specific
Occasionally, ependymomas arise in the lateral recess without evi- for ependymoma. Similarly, extension of tumor from the lateral recess
dence of tumor in the fourth ventricle (8,75,76,158). These laterally (Fig. 7-19) into the cerebellopontine angle cistern with insinuation
occurring posterior fossa ependymomas have a worse prognosis than around blood vessels and cranial nerves supports the diagnosis of
those in the midline (158–160), probably because complete resection is ependymoma. Midfloor-type ependymomas extend posteriorly; the
nearly impossible. For this reason, microanatomical localization of the brainstem therefore is displaced ventrally, not laterally, and can be
posterior fossa ependymomas correlates with the postoperative sur- seen wholly on a midline sagittal plane (Fig. 7-18B). Also, because the
vival (159). Those in the lower midfloor (Fig. 7-18) originate from the tumor originates about the obex, the lowest part of the fourth ven-
caudal end of the ventricle near the obex, extend dorsally into the ven- tricle appears blurred on the midline sagittal plane, and cannot be
tricle, toward the cisterna magna and into upper cervical canal, with recognized on axial cuts (Fig. 7-18B and C). Axial cuts at this level
only minimal lateral extension toward nerves and arteries; they can be show that the tumor is essentially dorsal to the brain, quite symmetri-
resected almost completely and the reported mean survival time is 170 cal, and usually without significant extension lateral to the medulla
months. Those originating from the lateral recesses/vestibular areas (Fig. 7-18C); in these cases, cranial nerves and arteries are usually not
(Fig. 7-19) extend laterally toward the cerebellomedullary and cer- invaded, resection may be fairly complete and prognosis is quite good.
ebellopontine angle cisterns, displace and encase the crossing cranial Lateral-type ependymomas originate in one of the lateral recesses and
nerves and arteries, and often involve the inferior cerebellar peduncle; extend toward the CP angle cistern; the brainstem is displaced con-
resection is always incomplete and the reported mean survival time is tralaterally and is only partially seen in the midline sagittal plane (Fig.
40 months. A few ependymomas originate from the ventricular roof 7-19B). On thin-section axial T2 images, the inferior extremity of
(inferior vermis, Fig. 7-20), extend caudally into and through the fora- the fourth ventricle is free of tumor (Fig. 7-19C); tumor growth into
men of Magendie, and can be totally resected; these have a reported the CP angle cistern and prepontine cistern may be recognized (Fig.
mean survival time of 116 months (159). Thus, imaging should strive 7-19C), raising suspicion for involvement of cranial nerves, and local
not only to identify the tumor but also to categorize it according to arterial encasement (Fig. 7-19B). In such cases, satisfactory resection
these criteria (158). is impossible. These lateral ependymomas may be completely extra-
Subarachnoid metastatic spread of benign ependymomas via cere- ventricular (Fig. 7-19). Roof-type ependymomas originate from the
brospinal fluid is uncommon (∼ 10%–12%) and the propensity for inferior vermis and extend into the ventricular lumen, through the
subarachnoid spread correlates with histology (higher grade tumors midline foramen of Magendie and into the cisterna magna and dorsal
more commonly disseminate [161,162]) and with age of the patient to the upper cord (Fig. 7-20); they do not come in contact with cranial
(younger patients have a higher incidence of dissemination [161]). nerves and do not encase arteries.
Therefore, when subarachnoid seeding is found at presentation, the Although medulloblastomas can extend into the fourth ventricular
presence of an anaplastic ependymoma should be suspected, especially outflow foramina, they do not send narrow tongues of tissue through
in an older child or adult. In contrast to medulloblastomas, ependymo- the foramina in the same way as ependymomas. Astrocytomas involv-
mas rarely have CSF dissemination at presentation; mean time from ing the medulla frequently extend into the cervical spinal canal, but
diagnosis to dissemination is 6.8 years (161). are intramedullary (i.e., within the cord, not dorsal to it). Dorsally
exophytic medullary astrocytomas may extend through the foramen
Imaging Studies magnum and lie posterior to the cervical spinal cord; however, upon
The most characteristic appearance of a posterior fossa ependymoma close examination, medullary tumors are seen to originate inferior to
on CT is that of an iso- to hyperdense (compared to gray matter) the fourth ventricle and to push it upward.

Barkovich_Chap07.indd 662 5/6/2011 9:15:51 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 663

FIG. 7-18. Midfloor-type ependymoma (from the caudal ven-


tricular floor, close to the obex). A. Noncontrast CT discloses
a rounded high attenuation mass in the low posterior midline
of the posterior fossa (arrows). Note that the typical medullo-
blastoma is located more centrally (see Fig. 7-8A). B. Sagittal
T1-weighted image shows that the tumor (T) is attached to the
dorsal medulla, but is separate from the vermis. It extends dor-
sally into the cisterna magna and inferiorly toward the upper
C-spine. The brainstem is pushed ventrally but is in the mid-
line. C. On this axial FLAIR image, the mass (T) fills the cisterna
magna. Although it originates from the floor of the lower fourth
ventricle it does not invade the brain stem but rather extends
dorsally, displacing the vermis. There is no or only little exten-
sion toward the lateral medullary cisterns. Ependymomas in
this location typically don’t invade the more laterally-located
cranial nerves and vessels; complete resection therefore is pos-
sible and their prognosis is better than it is for the lateral-type
ependymomas. D. Axial DWI shows reduced diffusion of the
tumor compared with cerebellum. E. Postcontrast T1-weighted
image shows diffuse, homogeneous enhancement.

Barkovich_Chap07.indd 663 5/6/2011 9:15:52 PM


664 Pediatric Neuroimaging

FIG. 7-19. Lateral-type ependymoma (origin in the lateral recess). A. Unenhanced CT shows a mass (T) located in the right side of the posterior fossa,
of similar attenuation to that of the gray matter. Note the fourth ventricle (arrow) displaced left of midline by the mass. B. Sagittal T2-weighted image
demonstrates a huge mass (T) that is nearly isointense to gray matter. The brainstem is displaced out of the plane of the image, suggesting that the mass is
pushing it from the side. Note the artery (arrow) encased within the tumor. C. Axial T2-weighted image demonstrates that the mass (T) is clearly mostly
extraparenchymal, and the laterally displaced fourth ventricle (arrow) is essentially tumor-free. Tumor location in the CP angle cistern commonly results in
the invasion of the cranial nerves and the encasement of the arteries, making complete resection nearly impossible; recurrences are common and prognosis
is worse than for the midfloor-type or roof-type ependymomas. D. Postcontrast T1-weighted image show no enhancement of the tumor (T); enhancement
is very variable in ependymomas.

DWI shows the solid portion of an ependymoma to be isoin- (TE = 30 milliseconds) show low to very high myo-inositol, although
tense to slightly hypointense compared with cerebellar white matter never as high as in choroid plexus papillomas (45,107).
(Dav values are intermediate between medulloblastoma and JPA, typi-
cally between 1.0 and 1.3 × 10−3 mm2/s [102]) (Figs. 7-18 and 7-20); Proton MR Spectroscopy of Medulloblastomas,
thus, they may be useful for differentiation from medulloblastoma or Cerebellar Astrocytomas, and Ependymomas
an ATRT, which have considerably lower Dav. Proton MR spectra show This subject has already been discussed briefly in the introductory
considerable heterogeneity (45). In general, ependymomas have low section of this chapter. Data have been published looking at metabo-
NAA and moderately elevated choline and creatine. Short echo spectra lite ratios of proton spectra of posterior fossa pediatric brain tumors

Barkovich_Chap07.indd 664 5/6/2011 9:15:53 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 665

FIG. 7-20. Roof-type ependymoma (from inferior vermis/


medullary velum). A. Axial noncontrast CT demonstrates a
dense posterior midline tumor (arrows) protruding into the
fourth ventricle, with a few punctate calcifications (arrowheads)
and a likely hemorrhage on its left anterior portion. B. Sagittal
T2-weighted image demonstrates the tumor (T) originating in
the inferior fourth ventricle and extending through the foramen
of Magendie into the dorsal cervical subarachnoid space. Note
the hemorrhagic/necrotic region (h) in the superior aspect of
the mass. This tumor originated from the caudal vermis; note
blurring of the tumor-vermis junction. The thin layer of CSF
separating tumor from the floor of the fourth ventricle shows it
is not a midfloor-type ependymoma. The dorsal, inferior exten-
sion of the tumor makes invasion of nerves and arteries less
likely; a total resection therefore is possible, which is the best
single factor of a good prognosis. C. Axial FLAIR image shows
blurring of the tumor-cerebellum junction dorsally (arrow) but
a sharp margin with the brainstem ventrally. D. DWI demon-
strates slight hyperintensity, suggesting moderate tumor cellu-
larity. E. Postcontrast T1-weighted image shows mild, patchy
enhancement of the tumor (T), again illustrating the variable
enhancement of ependymomas.

Barkovich_Chap07.indd 665 5/6/2011 9:15:55 PM


666 Pediatric Neuroimaging

(45,107,109,163). In general, proton spectra of brain tumors yield these are often at least partially surgically resectable, they should be
high choline (Cho) peaks and diminished peaks of creatine (Cr) and specifically identified. However, most astrocytic tumors of the pons
N-acetyl aspartate (NAA), but different types of tumors have differ- (the most common location of brainstem tumors) have fibrillary
ent levels of Cho elevation and NAA and Cr diminution (45). Proton histology (177); these have a typical neuroimaging appearance (T2
spectra of normal cerebellum in the pediatric patient (at 1.5 T using hyperintensity, slightly blurry margins, minimal or no enhancement)
echo time of 135 milliseconds) yield NAA/Cho ratio of 1.49 ± 0.36 and and, at many institutions, such tumors are treated without biopsy. It is
Cr/Cho ratio of 1.13 ± 0.23, whereas medulloblastomas yield NAA/ important to remember, however, that atypical features of a brainstem
Cho ratio of 0.17 ± 0.09, and Cr/Cho ratio of 0.32 ± 0.19 (163) and mass, particularly marked enhancement prior to therapy, the presence of
have elevated taurine when studied with short echo time (45). Low subarachnoid tumor spread at presentation, isointensity to gray matter
grade astrocytomas and ependymomas typically have NAA/Cho and on T2-weighted images (suggests a PNET), the presence of chronic blood
Cr/Cho values between those of medulloblastomas and those of nor- products (well marginated T1 shortening from methemoglobin or circular
mal cerebellum on using echo times of 135 milliseconds 1.5 T (163); areas of T2 shortening from hemosiderin), or the presence of extensive
both have elevated myo-inositol at 1.5 T with TE = 35 milliseconds, acute hemorrhage, may signal the presence of a nonglial mass, such as cav-
which may help to differentiate them from medulloblastomas (45). It ernoma, abscess, or nonastrocytic tumor. Such features should be brought
has been suggested, therefore, that proton MRS may be useful in pro- to the attention of the treating oncologist before therapy is initiated.
spectively differentiating medulloblastomas from astrocytomas and MRI is the study of choice when imaging neoplasms of the brain-
ependymomas (163). Harris et al. suggest that ependymomas have stem, because of the multiplanar capacity and the lack of artifact from
higher creatine than astrocytomas or medulloblastomas and that the the bony structures of the skull base (178). When using MR to image
presence of high myo-inositol levels strongly suggests a diagnosis of tumors of the brainstem, sagittal T2-weighted or FLAIR images should
ependymoma when short echo time (30 milliseconds) is used at 1.5 T always be obtained to help determine the full extent of the tumor and
(107). As these contradictory results indicate, the proton MRS findings thereby plan proper therapy. T2-weighted and FLAIR images are also
in tumors cannot be compared at different echo times; spectroscopy the most helpful for separating tumors into diffuse and focal groups
parameters for tumor assessment need to be standardized. They also (179,180). Diffuse neoplasms comprise 60% to 75% of all brainstem
suggest that the spectra of tumors with the same histologic type can tumors and usually correspond to histology of grade III or grade IV
vary. Indeed, the metabolite values for all tumors seem to vary with the fibrillary astrocytoma (177). In the great majority of cases, they affect
type of histologic subtype and the degree of histological malignancy the ventral pons and are labeled diffuse infiltrating pontine glioma
(47,109); thus there is considerable overlap among the different tumor (DIPG). They tend to smoothly enlarge the affected area (see Figs. 7-22,
types in individual cases. Moreover, pilocytic astrocytomas, considered 7-24, and 7-26) without focal areas of exophytic tumor, although they
the most benign of glial brain tumors, have high choline, high lactate, can bulge anteriorly and surround the basilar artery (Fig. 7-24A-C).
and low NAA (NAA/Cho ratio of 0.29, Cr/Cho ratio of 0.29 at TE = They are generally poorly marginated and involve more than 50%
20 milliseconds [164]), features that are usually associated with high (usually more than 75%) of the brainstem in the axial plane at the level
grade tumors. Finally, inflammatory lesions can have spectra that are of maximal involvement. Moreover, they tend to cross boundaries to
identical to those of malignant tumors (165). Therefore, even though extend into adjacent segments or structures (e.g., pontine tumors into
careful analyses of spectroscopy may, eventually, help to preopera- medulla or midbrain); this extension into an adjacent structure, cau-
tively differentiate among different histologies of brain tumors (45), it dally or (especially) cranially, is a poor prognostic sign (181). Minimal
still seems (even after 20 years of research on the subject) that proton or no contrast enhancement is seen on scans at presentation, although
spectroscopy is more useful for differentiating recurrent tumor from enhancement is common after therapy. Focal neoplasms are generally
granulation tissue and radiation injury than for differentiating among well marginated and involve less than 75% of the brainstem in the axial
different tumor histologies. plane (Figs. 7-23, 7-25, 7-27, and 7-28). Focal tumors often enhance,
but do not cross segmental boundaries to extend into adjacent seg-
ments or structures; as a result, when they enlarge they tend to become
Brainstem Tumors
focally exophytic. Histologically, most correspond to pilocytic astro-
General Concepts cytomas (WHO grade I), but gangliogliomas, though less common,
Brainstem gliomas constitute approximately 15% of all pediatric CNS are not unusual. Anaplastic or even PNETs may occasionally have this
tumors and account for 20% to 30% of infratentorial brain tumors. appearance (177). Overall, focal neoplasms of the brainstem have a
Males and females are affected with equal frequency. The peak inci- much better prognosis than diffuse neoplasms (179,180,182–185), due
dence of these tumors is between 3 and 10 years of age, although they to their very slow evolution and in spite of the fact that complete resec-
can be seen at any age from neonates to adults (8,75,166,167). MRI tion is usually impossible.
has simplified the diagnosis of brainstem tumors; it has also given new
information that has allowed the identification of subgroups with dif- Medullary Tumors
ferent therapeutic options and prognoses (166,168). In fact, brainstem The medulla is the least common site of origin for tumors of the brain-
tumors should not be considered as a single entity but should be sepa- stem. Medullary tumors typically present in young children with signs
rated into at least four separate major categories: medullary tumors, and symptoms of increased intracranial pressure (182,186). Dysfunc-
pontine tumors, mesencephalic tumors, and tumors associated with tion of lower cranial nerves may be found, resulting in difficulty in
NF1. Within each of these broad categories, further separations can be swallowing and nasal speech (185). Eventually, affected patients may
made, primarily into focal and diffuse tumors (see Table 7-3). Although develop hemiparesis or quadriparesis (185).
tumors of many different histologies (astrocytomas, gangliogliomas, On neuroimaging, medullary tumors can be differentiated into two
hemangioblastomas, PNETs, Langerhans cell histiocytosis [LCH], ger- major categories, focal dorsally exophytic tumors and diffuse tumors.
minomas, lymphomas, and glioblastomas, among others [169–175]) A third group of small, focal, intrinsic medullary tumors is usually seen
can develop in the brainstem, the vast majority are astrocytic tumors in the setting of NF1 (see Chapter 6).
(76,176). In general, dorsally exophytic tumors and other localized, Focal dorsally exophytic medullary tumors are generally sharply
markedly enhancing tumors are pilocytic astrocytomas (176); as marginated masses that originate in the dorsal aspect of the medulla

Barkovich_Chap07.indd 666 5/6/2011 9:15:56 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 667

FIG. 7-21. Dorsally exophytic medullary glioma extending into the cervical spinal cord. A. Sagittal T1-weighted image shows a large tumor originating
in the medulla and extending posteriorly between the cerebellar hemispheres. The cerebellum is pushed posteriorly and upward by the tumor mass (white
arrows). Caudal extension widens the cervical spinal cord (closed white arrowheads). B. Postcontrast sagittal T1-weighted image shows heterogeneous
enhancement of the mass.

and grow exophytically into the vallecula and cisterna magna, often exophytic medullary tumors inconsistently enhance after administra-
causing upward and posterior displacement of the inferior cerebel- tion of contrast; when enhancement is present, it may be homogeneous
lar vermis (Fig. 7-21). The dorsal extension is a result of the inability or heterogeneous (179).
of the low-grade tumor to infiltrate caudally below the decussations Diffuse medullary tumors (Fig. 7-22) are more infiltrative neo-
of the corticospinal tracts and medial lemnisci at the craniocervical plasms that typically extend rostrally into the pons or caudally into the
junction (177); uncommonly, they may extend into the tegmentum cervical spinal cord, but do not have a large exophytic component. Dif-
pontis as well. Therefore, dorsally exophytic tumors should be con- fuse medullary tumors have a significantly worse prognosis than their
sidered as focal (177). The dorsal extension makes it difficult to dif- dorsally exophytic counterparts (179), possibly because they are more
ferentiate an exophytic medullary glioma from a cerebellar vermian difficult for the neurosurgeon to adequately resect. On CT, diffuse med-
astrocytoma using axial images (10,11); therefore, acquisition of sag- ullary tumors may be difficult to detect because of the extensive arti-
ittal images is essential. The exophytic component may extend infe- fact that may be present around the skull base and in the spinal canal;
riorly through the foramen magnum, dorsal to the cervicomedullary the typical CT appearance is that of a mildly to moderately enlarged,
junction, superficially mimicking an ependymoma (Fig. 7-21). Histo- hypodense medulla. The full extent of these tumors is demonstrated
logically, these tumors are typically pilocytic astrocytomas (186) and, best by using sagittal T2-weighted or FLAIR MR sequences (Fig. 7-22).
therefore, they are of low attenuation on CT and are very bright on Their typical MR appearance is that of a diffusely enlarged medulla,
T2-weighted MR images. Thus, they can be differentiated from mid- exhibiting T1 hypointensity and T2/FLAIR hyperintensity, that infil-
floor type of ependymomas, which are isodense to gray matter on CT trates into the caudal pons and rostral cervical spinal cord. Enhance-
and T2-weighted MR images. Medullary pilocytic astrocytomas tend ment is inconsistent and is usually focal or multifocal (179).
to invade the medulla in depth in addition to being exophytic, while Focal medullary tumors (Fig. 7-23) are most commonly seen in the
midfloor ependymomas tend to be almost exclusively exophytic from setting of NF1. They are not usually identified on CT images. On MRI,
their site of attachment on the fourth ventricle. In addition, dorsally they appear as focal expansions of the dorsal medulla that exhibit T1
exophytic medullary tumors can be seen to push the fourth ventricle hypointensity and T2/FLAIR hyperintensity. Enhancement is uncom-
upward, in contrast to ependymomas, which originate within the wall mon. These tumors seem to exhibit very slow growth, showing little
of the fourth ventricle, grow into the ventricle, and expand it. Dorsally change on sequential imaging studies.

Barkovich_Chap07.indd 667 5/6/2011 9:15:56 PM


668 Pediatric Neuroimaging

FIG. 7-22. Diffuse medullary glioma. A. Sagittal T2-weighted image shows T2 prolongation (white arrows) associated with the tumor extending superiorly
(large white arrowheads) into the pons as well as inferiorly (small white arrowhead) into the spinal cord. B. Sagittal T1-weighted image shows expansion of
the medulla with poorly defined hypointensity (white arrows) centered in the medulla and extending inferiorly into the cervical spinal cord. The extension
into adjacent segments is not seen on this image. C. Axial T2-weighted image shows that the hyperintense tumor (arrows) involves more than 50% of the
transverse diameter of the medulla. D. Postcontrast T1-weighted image shows lack of enhancement of the mass (arrows), characteristic of diffuse tumors
of the brainstem.

Cervicomedullary tumors have much in common with cervical shows the tumor infiltrating into the spinal cord from the medulla or
spinal cord tumors and, indeed, usually originate in the cervical spinal vice versa, a high-grade tumor should be suspected.
cord and push up the decussations of the corticospinal tracts and medial
lemnisci at the craniocervical junction, thereby appearing to invade the Pontine Tumors
medulla. These tumors are discussed in Chapter 10. It will suffice to say The pons is the most common location of tumors originating in the
in this section that most of these tumors are of low histologic grade, brainstem. The classic presentation of a pontine tumor is cranial nerve
with no or minimal infiltrative capacity (177). If the sagittal MR image palsies, usually multiple, with pyramidal tract signs and cerebellar

Barkovich_Chap07.indd 668 5/6/2011 9:15:57 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 669

FIG. 7-23. Focal medullary glioma in patient with NF1.


A. Sagittal T1-weighted image shows focal dorsal con-
tour abnormality (black arrows) in the dorsal medulla.
A hypothalamic mass (m) is also present. B and C. Axial
T1-weighted and T2-weighted images show the focal
mass (black arrows), expanding the left dorsal medulla.

dysfunction (ataxia and nystagmus) (8,166). Smaller, more focal T2-weighted and FLAIR MR images. Expansion by the tumor will
tumors may present with isolated cranial neuropathies (184). Hydro- result in an increased sagittal dimension of the pons and posterior dis-
cephalus and signs of increased intracranial pressure are uncommon. placement of the fourth ventricle. In addition, the floor of the fourth
The prognosis for most patients is poor with an overall 5 year sur- ventricle may become flattened. When the tumor extends into the
vival rate of approximately 10% despite optimal therapy (166,179,180). cerebellar peduncles, the lateral aspects of the fourth ventricle can be
Prognosis depends upon the location and aggressiveness of the tumor. flattened, causing an apparent rotation of the ventricle. The pontine
DIPGs, which are poorly circumscribed, large, and often extend into surface is irregular, and contains exophytic nodules that can grow into
the medulla and midbrain, are usually high grade fibrillary gliomas and the cerebellopontine angle or prepontine cistern or can grow peripher-
have extremely poor prognoses, with a two year survival rate of only ally along the cranial nerves. Anterior growth frequently engulfs the
7% (176,187). However, more focal, smaller tumors, particularly those basilar artery (particularly in fibrillary astrocytomas [176]), which
that originate in the dorsal pons and are exophytic into the fourth ven- eventually lies within a deep groove bounded by tumor anteriorly and
tricle, have much better prognoses, with 5-year survival rates as high as laterally (Fig. 7-24B). Histologically, the tumor cells infiltrate widely
73% (176,179,180,182–184,188). along the fiber tracts of the brainstem rather than destroying them so
DIPGs (Fig. 7-24) are infiltrative masses that are hypodense on that one sees tumor cells intermingling intimately with neurons and
CT, hypointense on T1-weighted MR images, and hyperintense on nerve fibers (8,189). As a result of this characteristic, the appearance

Barkovich_Chap07.indd 669 5/6/2011 9:15:59 PM


670 Pediatric Neuroimaging

FIG. 7-24. Diffuse pontine glioma. A. Sagittal T1-weighted image shows an expanded pons with T1 prolongation involving more than 50% of the transverse
diameter of the pons. The fourth ventricle is compressed by the enlarged pons. Tumor mass is seen to extend anterior to the basilar artery (arrows). Hydro-
cephalus is not present. B. Axial FLAIR image shows the large pontine mass (white arrows) expanding the pons and engulfing the basilar artery (black arrow).
C. Postcontrast axial T1-weighted image shows the basilar artery (white arrow) engulfed by the mass. D. Proton MR spectrum (TE = 288 milliseconds)
shows very large choline peaks and small NAA peaks in the tumor mass. E. Perfusion curves (relaxivity in the vertical axis, time in the horizontal axis) show
markedly increased blood volume (resulting in decreased signal and decreased MR units) in the tumor (T) compared to normal brain tissue (N) in a simi-
larly located voxel in the cerebellum. This is characteristic of high-grade glial neoplasms.

Barkovich_Chap07.indd 670 5/6/2011 9:15:59 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 671

FIG. 7-25. Focal pontine glioma. A. Sagittal T1-weighted image shows a


small hypointense mass (black arrows) in the dorsal pons. B. Axial FLAIR
image shows the hyperintense mass (white arrows) slightly protruding into
the fourth ventricle. C. Proton MR spectra (TE = 288 milliseconds) shows
that the choline peaks are not much elevated compared with unaffected
pontine and cerebellar voxels. NAA is decreased. These findings are more
characteristic of a lower grade neoplasm. D. Perfusion curves (relaxivity
in the vertical axis, time in the horizontal axis) show similar blood volume
(resulting in decreased signal and decreased MR units) in the tumor (T)
compared to normal brain tissue (N) in a similarly located voxel in the
cerebellum. This is characteristic of low-grade glial neoplasms.

of the pons can return to normal after treatment of these tumors. brainstem tumors. By definition, these tumors occupy less than 50%
However, significant tumor recurrence is usually seen within 18 to of the transverse area of the pons at the involved levels. They may be
24 months following therapy (166,190). In spite of the marked enlarge- located anywhere within the pons and may be poorly or sharply mar-
ment of the brainstem and its frequent bulging into the fourth ventri- ginated. When they originate in the periphery of the pons, they may
cle, hydrocephalus is uncommon in pontine gliomas. Diffuse pontine grow exophytically into the fourth ventricle or cerebellopontine angle.
tumors rarely enhance after administration of contrast (179). After administration of contrast, the solid portions of these tumors
Focal pontine tumors (Fig. 7-25) are very uncommon, compris- typically enhance heterogeneously (179,182,184,188). They are usually
ing only about 10% of pontine tumors and, therefore, less than 5% of low-grade tumors, often pilocytic astrocytomas.

Barkovich_Chap07.indd 671 5/6/2011 9:16:01 PM


672 Pediatric Neuroimaging

FIG. 7-26. Diffuse midbrain glioma.


A. Axial T1-weighted image shows a
poorly marginated mass with foci of
hyperintensity (arrows), likely rep-
resenting blood, originating in the
midbrain and extending inferiorly
into the pons and superiorly into the
thalamus. B. Axial FLAIR image shows
the hyperintense mass extending from
the right cerebral peduncle anteriorly
into the temporal lobe (arrows). The
heterogeneity within the mass likely
represents necrosis. C. Postcontrast
T1-weighted image shows heteroge-
neous enhancement in the cerebral
peduncle and extending into the
temporal lobe (arrows). D. Postcon-
trast T1-weighted image shows het-
erogeneous enhancement in the pons
(arrows). The thalamus was similarly
involved (not shown). This rostral and
caudal extension is strongly suggestive
of a high grade tumor.

Proton MR spectroscopy and perfusion imaging may be useful in being dependent upon the location of the mass (188,192,194); upward
differentiating low grade (Fig. 7-25), which have lower choline peaks gaze paresis (Parinaud sign) may be present (192). Tectal tumors cause
and smaller blood volumes, from high-grade tumors (Fig. 7-24). As signs and symptoms of hydrocephalus (see Chapter 8); upward gaze
discussed with cerebellar tumors, however, pilocytic astrocytomas paresis is conspicuously absent (188,191,192,195).
(WHO grade I) have relatively high choline peaks and lactate peaks and Precise diagnosis of the different midbrain tumors by neuroimag-
relatively high blood volumes, thus mimicking high-grade tumors. ing is very important in patient management; focal midbrain tumors
are treated surgically, whereas diffuse tumors are treated with chemo-
Midbrain Tumors therapy/radiation, and tectal tumors are typically treated only by CSF
The midbrain is the second most common site of tumors originating in diversion unless tumor growth is documented on sequential imaging
the brainstem. As with pontine and medullary tumors, midbrain tumors studies (growth of tectal tumors is unusual [191,195]).
should be separated by imaging into focal and diffuse tumors. In addi- Diffuse midbrain tumors (Fig. 7-26) are uncommon tumors that
tion, many authors consider tectal tumors as a distinct third group of are centered in the midbrain, but extend rostrally into the cerebral
midbrain tumors (191–193). In contrast to the predominance of dif- hemispheres and caudally into the pons and, sometimes, medulla.
fuse tumors in the pons, focal tumors are much more common in the These tumors have relatively little local mass effect and show mini-
midbrain. The manner of patient presentation varies with the type of mal enhancement (Fig. 7-26) after administration of paramagnetic
tumor. Diffuse midbrain gliomas typically cause blurred vision or dou- contrast.
ble vision, often of acute onset and, sometimes, accompanied by motor Focal midbrain tumors (Fig. 7-27) are typically sharply circum-
weakness. In contrast, patients with focal tumors typically present with scribed masses that focally enlarge the affected area. The tumor may be
headache, vomiting, diplopia, and hemiparesis, with the exact symptoms midline or eccentric within the cerebral peduncle. They are typically of

Barkovich_Chap07.indd 672 5/6/2011 9:16:02 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 673

FIG. 7-27. Focal midbrain glioma. A. Sagittal T1-weighted


image shows a sharply marginated hypointense mass (arrows)
within the tegmentum of the midbrain. B. Sagittal T2-weighted
image shows that the hyperintense mass (arrows) involves the
right cerebral peduncle and midbrain tegmentum. No surround-
ing edema is identified. C. Axial postcontrast T1-weighted image
shows nearly homogeneous enhancement of the solid portion
of the tumor. A small cyst (white arrowheads) is present at the
posterior tumor margin.

low attenuation on CT, of low intensity on T1-weighted MR images, and mass. Most will show no growth and can be followed without treat-
of high intensity on T2-weighted images. Hemorrhage or cysts are pres- ment (188,191,193,195,197).
ent in approximately 25% of focal midbrain tumors (76,189,192). The
tumors occasionally extend superiorly into the thalamus and, less com- Brainstem Tumors Associated with Neurofibromatosis Type 1
monly, inferiorly into the pons (179,196). Administration of contrast Patients with NF1 have a high incidence of brain tumors, with the
may show ring enhancement when the tumor is small; larger tumors brainstem being a common site of involvement (see Chapter 6). In one
show homogeneous (Fig. 7-27) or heterogeneous enhancement (179). series, brainstem tumors were detected in 9% of patients with NF1 who
Tumors of the quadrigeminal plate (Fig. 7-28), also called tectal were studied by MR (198). The relative frequency of sites of tumor
gliomas, are discussed in the section on “Pineal Region Masses.” These origin differs, with the medulla (Fig. 7-23) being the most common site
tumors are, in general, extremely benign and are initially treated only (68%–82%) in NF1, in contrast to the marked pontine predominance
by CSF diversion of the resultant hydrocephalus. Sequential neuroim- in patients without NF1 (198–202). Although brainstem tumors of NF1
aging studies (preferably MR) are obtained to look for growth of the may have an identical imaging appearance to those that occur outside

Barkovich_Chap07.indd 673 5/6/2011 9:16:04 PM


674 Pediatric Neuroimaging

FIG. 7-28. Glioma of the quadrigeminal plate. A. Sagittal TWI


shows an isointense to hypointense mass (m) expanding the tec-
tal plate with effacement of the intercollicular sulcus. The lateral
and third ventricles are markedly expanded due to hydrocepha-
lus. B. Sagittal T2 also demonstrates the enlarged tectal plate and
shows diffusely increased signal that extends around the aque-
duct and over the cap of the midbrain. C. Axial FLAIR image
shows the abnormal morphology and thickening of the tectal
plate (arrows) and the heterogeneous hyperintensity.

the spectrum of NF1, their clinical behavior is often strikingly differ- from pontine gliomas with more guarded prognoses. Broniscer et al.
ent (198,200–202). Many of the patients with NF1 are asymptomatic have reported that patients with NF1 who have pontine enlargement
from their tumors, which are detected incidentally (198,201). The most with benign clinical courses have significantly higher NAA and choline
striking difference is that of tumor growth. In follow-up periods aver- levels, as detected by proton MRS, than patients with more clinically
aging 3.75 to 4.3 years, only 32% to 42% of patients with NF1 showed aggressive pontine gliomas (203). Unfortunately, because metabolite
radiologic progression and only 14% to 18% showed clinical progres- levels, as detected by MRS, vary from scanner to scanner, this is a rela-
sion of these tumors (198,199,201). Even patients with tumors that tive measurement and no absolute values are available to differentiate
have imaging appearances identical to diffuse pontine tumors often between these two entities.
have relatively indolent clinical courses (198). Spontaneous remissions
have been reported (199). Therefore, the recommendation at present Differential Diagnosis of Brainstem Tumors
is that only those tumors that exhibit rapid or unrelenting growth on The major differential diagnoses of brainstem tumors are encephali-
serial images or that produce significant clinical deterioration should tis (viral, autoimmune, or protozoan [204,205]), abscess (see Chapter
have intervention (198,201). 11), demyelination (see Chapter 3) (206), dysmyelination secondary to
It is possible that localized proton MRS may help in the differ- NF1 (see Chapter 6), LCH (175), hamartomas (206), resolving hema-
entiation of benign pontine enlargement (low grade tumor?) of NF1 toma, cavernous malformation (see Chapter 12), and tuberculoma

Barkovich_Chap07.indd 674 5/6/2011 9:16:05 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 675

(207). Because of its exquisite demonstration of blood and blood brain, a portion of the tumor is always connected to a pial surface. The
breakdown products, MR can differentiate brainstem astrocytomas hemangioblastoma is classically described as a well-circumscribed,
from cavernous malformations and subacute hemorrhage very easily. soft, cystic tumor with a mural nodule. However, 30% to 40% of symp-
Abscesses are now differentiated by the use of DWI (reduced diffusion tomatic hemangioblastomas are solid; small, asymptomatic tumors are
in abscesses, increased diffusion in cystic/necrotic tumors) and proton much more commonly solid (210). On gross inspection, the solid por-
spectroscopy (amino acid peaks are seen in abscesses but not tumors) tion of the tumor may be intensely hemorrhagic. Calcification is not
(49–52,208,209). Demyelination, Langerhans cell histiocytosis, and found in these lesions.
dysmyelination due to NF1 can typically be differentiated by the pres-
ence of other lesions, a past medical history, or history of current dis- Imaging Studies
ease. Tuberculoma can typically be identified by the short T2 relaxation The CT appearance of hemangioblastomas is usually that of a cystic or
time of the caseated central portion (see Chapter 11). However, at this solid posterior fossa mass. When large enough, the solid portion of the
time, it is still not possible to definitively differentiate acute encephali- tumor always enhances homogeneously and intensely after infusion of
tis from a brainstem tumor on a single imaging study. History, labora- intravenous contrast. However, when the solid portion of the tumor
tory studies, and, sometimes, sequential imaging studies are crucial to is extremely small, it will not be seen on CT, particularly if it is in the
making the correct diagnosis. lower cerebellum or brainstem where artifact is most severe. Angiogra-
phy will confirm the vascular component of the tumor and will detect
Hemangioblastoma small hemangioblastomas not seen on contrast CT. These very small
Hemangioblastomas are relatively rare benign tumors of vascular ori- lesions can be reliably detected on contrast-enhanced MRI.
gin, with an incidence ranging from 1% to 2.5% of intracranial neo- The most common MR appearance is that of a cystic mass with
plasms. Less than 20% of hemangioblastomas occur in children, with a vascular mural nodule (Fig. 7-29) (211). The cyst may be hyperin-
most being seen in young and middle-age adults. The incidence in tense on T1-weighted images if hemorrhage has recently occurred.
males exceeds that in females (8,75,76). Approximately 10% of heman- More commonly, cysts are hypointense on T1-weighted images and
gioblastomas occur in association with retinal angiomas, a condition hyperintense on T2-weighted images (211,212) (Fig. 7-29). Moreover,
known as von Hippel-Lindau disease (see Chapter 6). Patients with von on T2-weighted and FLAIR images, a peripheral rim of hyperinten-
Hippel-Lindau disease tend to have multiple hemangioblastomas of sity may be seen as the result of surrounding edema. Solid portions of
the CNS, multiple cysts, or tumors involving the pancreas, liver, kidney, tumor enhance dramatically after intravenous infusion of paramagnetic
and lung. Patients with hemangioblastomas may present with eryth- contrast (213) (Fig. 7-29E and F). Other examples of MR images of
rocythemia, thought to result from erythropoietin production by the hemangioblastomas are provided in Chapter 6, in the section on “von
tumor (8,75,76). Hippel-Lindau Disease.” When the mural nodule is small, it may be
difficult to differentiate a hemangioblastoma from an arachnoid or
Pathology neuroepithelial cyst. In such cases, contrast-enhanced MRI should be
Hemangioblastomas occur most commonly in the cerebellar hemi- obtained in at least two planes, as volume averaging may obscure a
sphere, especially the paramedian hemispheric area. Tumor origin small region of enhancement in a single plane. When performed in at
within the spinal cord is common and is often accompanied by syrin- least two planes, contrast-enhanced MRI allows identification of very
gomyelia. Rarely, hemangioblastomas may be located in the brainstem small tumor nodules, because they enhance intensely. Because of the
or the cerebral hemispheres. As they originate from the surface of the very dense vascularity of the tumor, MR angiography often shows the

FIG. 7-29. Hemangioblastoma of the dorsal medulla. A. Sagittal T2-weighted image shows a large loculated, heterogeneous mass (m) located in the dorsal
medulla, protruding into the inferior fourth ventricular. Signal is higher than that of the cerebellum, and multiple vascular signal voids (arrowheads) suggest
a rich vascularity. Although mostly solid, the tumor also contains two cysts (c) in its inferior aspect. There is massive associated edema in the medulla and
the upper spinal cord. B. Axial FLAIR imaging demonstrates hyperintensity of the tumor (m), multiple vascular signal voids and edema of the medulla.

Barkovich_Chap07.indd 675 5/6/2011 9:16:06 PM


676 Pediatric Neuroimaging

FIG. 7-29. (Continued) C. DWI shows low signal of the mass (m) with respect to the cerebellar hemispheres, reflecting intratumoral edema, and relatively
low cellularity. D. Maximum intensity projection from time-of-flight MR angiography shows the rich arterial network feeding the mass. E. Postcontrast
T1-weighted image reveals intense homogeneous enhancement of the solid part of the tumor (m), but not of the walls of the associated cysts (c). F. Conven-
tional angiogram, late arterial phase. This intense angiographic blush (b) is characteristic of hemangioblastomas.

main arterial feeder, and even some branching (Fig. 7-29D). In addi- system. Schwannomas account for about 8% of primary tumors in
tion, conventional arteriography may be indicated sometimes to visual- the intracranial cavity; however, they are more frequent in adults and
ize the hypervascular nidus (Fig. 7-29F), and many neurosurgeons will compose only 2% of posterior fossa tumors in children (8,75,214).
order preoperative angiography on all suspected hemangioblastomas When schwannomas are diagnosed in childhood, the possibility of
to determine the location of the vascular pedicle prospectively. Pre- type 2 neurofibromatosis (NF2) should be considered (see Chapter
operative embolization may serve to reduce blood loss and operative 6). An evaluation for other schwannomas and meningiomas should
morbidity. In all patients with suspected or known von Hippel-Lindau be carried out (214). Schwannomas tend to occur at the sites of gan-
disease, the spine should also be imaged, as affected patients have a glia, i.e., at the transition from oligodendroglial cells to Schwann cells
surprisingly high incidence of spinal hemangioblastomas (210). (215). For example, vestibular schwannomas tend to occur at Scarpa
ganglion, which lies within the internal acoustic canal or in the cer-
Extraparenchymal Tumors ebellopontine angle. The location of the tumor often determines the
symptoms with which the patient presents. Because schwannomas are
Schwannomas tumors of the nerve sheath and not of the nerves themselves, patients
Schwannomas are tumors derived from Schwann cells, which form the present with neuropathy only if the schwannoma occurs within a bony
myelin sheaths around the axons of nerves in the peripheral nervous canal. In such cases, outward growth of the tumor is restricted by bone;

Barkovich_Chap07.indd 676 5/6/2011 9:16:06 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 677

FIG. 7-30. Schwannoma of the ninth cranial nerve. A. Axial T1-weighted


image shows a large mass (arrows) filling the cerebellomedullary angle
cistern, displacing the medulla (arrowheads) to the left, and entering the
jugular foramen anteriorly. B. Axial T2-weighted image shows the tumor
(arrows) to be heterogeneous, with areas of probably necrosis. C. Postcon-
trast T1-weighted image shows nearly homogeneous enhancement of the
mass.

the neoplasm therefore grows centripetally into the nerve causing com- intratumoral hemorrhage, whereas the areas of T2 hyperintensity are
pression of the nerve and, hence, a neuropathy (216). However, if the usually regions of cystic necrosis. Presentation with acute intratumoral
schwannoma occurs within the cranial cavity, symptoms will not occur hemorrhage is extremely rare. The enlarged cranial nerves coursing
until growth of the tumor compresses adjacent neural structures; the through enlarged neural foramina (i.e., the foramen ovale and fora-
presenting signs and symptoms are those of brainstem compression or men rotundum in trigeminal schwannomas and the internal auditory
hydrocephalus from compression of the aqueduct or fourth ventricle. canal in acoustic schwannomas) and enlargement of adjacent CSF cis-
The most common nerve to be involved by a schwannoma is the eighth terns are easily demonstrated by MRI. Administration of paramagnetic
cranial nerve, followed by the fifth, ninth, and tenth cranial nerves. contrast results in enhancement of solid portions of the tumor (Figs.
Other intracranial schwannomas are extremely rare, other than in the 7-30 and 7-31) (219). An example of schwannomas in the setting of
setting of NF2 (215). NF2 is illustrated in Figure 7-31 and several examples are in Chapter 6.
Small schwannomas may also be detected on high definition T2 imag-
Imaging Studies ing (CISS/FIESTA) as focal enlargements of the nerve.
On noncontrast CT, schwannomas appear as hypodense to isodense
Dysembryoplastic Tumors (Epidermoids, Dermoids,
masses that occasionally calcify. Central necrosis can occur in large
and Enteric Cysts)
lesions. After the infusion of intravenous contrast, the solid portion
of the tumor typically shows homogeneous enhancement (215). When Dermoids and Epidermoids: Presentation and Pathology
they occur within bony canals, schwannomas almost always enlarge Dermoid and epidermoid tumors will be discussed in this section
the canals by pressure erosion. For example, acoustic schwannomas because they occur somewhat more frequently in the posterior fossa
normally enlarge the internal auditory canal and facial schwannomas than in the supratentorial space or spine. Epidermoids develop from
frequently enlarge the facial canal within the petrous bone. the ectoderm-derived epidermis, whereas dermoids arise not only
On MRI, schwannomas have prolonged T1 and T2 relaxation times from the epidermis but also from the subjacent dense connective tis-
(217–219). Large schwannomas often show heterogeneity with areas of sue, the mesoderm-derived dermis. Both of these tumors are believed
high and low signal intensity on T2-weighted sequences (Fig. 7-30); the to arise from congenital rests of tissue that remain in the intracranial
areas of T2 hypointensity are most likely caused by the by-products of cavity as a result of incomplete separation of the neuroectoderm from

Barkovich_Chap07.indd 677 5/6/2011 9:16:07 PM


678 Pediatric Neuroimaging

are diagnosed in middle age with a peak incidence during the fifth
decade. Epidermoids in the cerebellopontine angle tend to present
with cranial neuropathies. In contrast, suprasellar and pineal region
epidermoids present with hydrocephalus, while middle cranial fossa
epidermoids most frequently present with chemical meningitis sec-
ondary to leakage of tumor contents into the subarachnoid space
(8,75,76,220).
Dermoids are less common in the intracranial cavity than epider-
moids, but are more common in the spinal canal. In the intracranial
cavity they are most frequently located in the posterior fossa, either
within the vermis or in the fourth ventricle. In the spinal canal, most
dermoids occur in the lumbosacral region, either in an extramedul-
lary or an intramedullary location. Approximately 20% of dermoids
are associated with dermal sinuses; those dermoids associated with
dermal sinus tracts are most common in the spine, cerebellar vermis,
and subfrontal regions. Symptoms from intraspinal dermoids usually
arise in the first two decades of life, whereas those from intracranial
dermoids are more typically manifested in the third decade. Symp-
toms may be the result of obstruction of the CSF pathways, chemical
meningitis from leakage of the contents of the cyst into the CSF, or
(when associated with dermal sinus tracts) from infection and abscess
formation within the tract or the dermoid itself (8,75,76,221,222).
Extracranial dermoids, which typically present as masses on the head,
are discussed in Chapter 5. Spinal dermoids developing in associa-
FIG. 7-31. Multiple schwannomas in a patient with neurofibromatosis
tion with dermal sinuses are discussed in Chapter 9, whereas those
type 2. Postcontrast T1-weighted image shows bilateral enhancing acoustic
schwannomas (closed arrows) and an enhancing facial nerve schwannoma developing in the spine in the absence of dermal sinuses are discussed
(open arrow). in Chapter 10.

Imaging Studies
the cutaneous ectoderm at the time of closure of the neural tube. Epidermoids CT scans show epidermoids as low density, lobulated
Epidermoids are more common than dermoids (8,75,76). masses that occur in characteristic locations (Fig. 7-32). The attenuation
The location of epidermoids shows a much greater variation than is often identical to that of CSF, making identification of these extra-
that of dermoids, as well as a greater tendency to deviate from the axial lesions difficult. If necessary, the introduction of water-soluble
midline. The most frequent site is the cerebellopontine angle, fol- contrast into the subarachnoid space will demonstrate insinuation of
lowed by the pineal region, the suprasellar region, and the middle the contrast into the interstices of the epidermoid, revealing its charac-
cranial fossa. Although epidermoids may present at any age, most teristic lobulated appearance (Fig. 7-32B). However, particularly with

FIG. 7-32. Epidermoid tumor: use of CT


cisternography. A. Contrast-enhanced CT
scan shows a low attenuation, nonenhanc-
ing suprasellar mass (arrows). Note that
the margins of the mass are somewhat
irregular and lobulated. B. After introduc-
tion of intrathecal contrast, the contrast
material can be seen to diffuse into the
interstices of the mass, showing a char-
acteristic lobulated appearance (arrows).
This lobulated appearance is very charac-
teristic of epidermoid tumors.

Barkovich_Chap07.indd 678 5/6/2011 9:16:08 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 679

FIG. 7-33. Infravermian epidermoid tumor. A. Sagittal T1-weighted image shows a hypointense, slightly heterogeneous, lobulated mass (arrows) with
lobular indentations on the dorsal medulla (arrowheads). Epidermoids are characteristically isointense with CSF. B. Axial T2-weighted image shows a lobu-
lated mass displacing the cerebellar hemispheres and medulla. C. Postcontrast axial T1-weighted image shows that the tumor does not enhance, instead
remaining isointense with CSF. D. Diffusion-weighted image (b = 1000 s/mm2) shows that the mass is heterogeneously hyperintense, indicated reduced
diffusion compared with CSF and differentiating it definitively from an arachnoid cyst, which would remain hypointense (see Fig. 7-34). The heterogeneity
is likely due to fibrous tissue and perhaps fluid in the interstices of the tumor.

FLAIR and diffusion-weighted sequences, the diagnosis of epidermoid pied cisterns and the characteristic lobulated appearance of the mass
by MRI is so accurate that CT should only be used if MRI is not available. (Fig. 7-33) (223). Differentiation from a smooth-walled arachnoid
On MRI, epidermoids appear as somewhat heterogeneous, mildly cyst is not difficult when the mass is large; a lobulated appearance and
lobulated extra-axial masses with prolonged T1 and T2 relaxation the presence of linear heterogeneities (Fig. 7-33) within the mass sug-
times; enhancement is not seen after infusion of intravenous contrast gest that it is an epidermoid. Diagnosis is confirmed by DWI, which
(Fig. 7-33). Thus, as with CT, most epidermoids have a signal inten- shows diffusivity of a solid mass (signal intensity similar to or higher
sity that is similar to CSF on standard T1- and T2-weighted images. than brain tissue, Fig. 7-33D), whereas cysts have characteristics of
Difficulty may arise in identifying small lesions and, once identified, fluid (similar to CSF, Fig. 7-34D) (224,225). FLAIR (Fig. 7-35) and
in differentiating an epidermoid from an arachnoid cyst (Fig. 7-34). CISS sequences improve the conspicuity of epidermoids (225,226)
Cisternal epidermoids are identified by enlargement of the occu- and also are useful in differentiating them from cysts. Alternatively,

Barkovich_Chap07.indd 679 5/6/2011 9:16:09 PM


680 Pediatric Neuroimaging

FIG. 7-34. Infravermian arachnoid cyst for contrast with epidermoid. A. Sagittal T1-weighted image shows a hypointense infravermian mass. The margins
of the displaced brain are completely smooth other than the fastigium of the fourth ventricle (arrow). No lobulations are seen, in contrast to the contour
around the epidermoid tumor in Figure 7-33. B. Axial T2-weighted image shows the homogeneous cyst displacing the medulla and cerebellar hemispheres.
No lobulations are seen. C. Postcontrast T1-weighted image shows absence of enhancement of the homogeneously hypointense mass. D. Diffusion-weighted
image (b = 1000 s/mm2) shows the cyst to be homogeneously hypointense, identical to CSF, indicating water diffusion equal to that of CSF. Contrast this
with the heterogeneous hyperintensity in the epidermoid tumor illustrated in Figure 7-33.

magnetization transfer techniques may be useful, as epidermoids will dermoids, enhancement is not seen after infusion of IV contrast unless
show significant transfer of magnetization from the solid matrix of the tumor is or has been infected (223). Whenever a midline dermoid
the tumor to adjacent free water whereas cysts show no magnetiza- or epidermoid tumor is encountered, the occipital and naso-frontal
tion transfer (227). Occasionally, epidermoids show T1 hyperintensity regions of the skull (when intracranial) or the posterior elements of the
(228); in this circumstance, they are more difficult to differentiate from vertebra (when intraspinal) should be examined for defects that might
dermoids or lipomas. Differentiation can be made by application of fat indicate the presence of a dermal sinus tract. CT is more sensitive than
suppression; the high signal from dermoids or lipomas will be satu- MRI for detection of the calvarial defect, although MRI is more sensi-
rated (disappear), but the high signal from epidermoids will remain tive in detecting the tumor (229). Dermal sinuses result from a focal
unaffected. nondisjunction of neural ectoderm from cutaneous ectoderm and
may be lined by fat (see Chapter 9). If infected, dermoid cysts have the
Dermoids The CT appearance of a dermoid is that of a fat-attenua- typical appearance of an abscess with a low diffusivity and a uniform,
tion, extra-axial mass that is usually located in the midline. As with epi- contrast-enhancing rim surrounding them.

Barkovich_Chap07.indd 680 5/6/2011 9:16:11 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 681

FIG. 7-35. Utility of FLAIR and diffusion imaging in epidermoid tumors. A. Postcontrast axial T1-weighted image shows a hypointense mass (arrows) in
the left cerebellomedullary angle. B. Axial T2-weighted image shows the hyperintense mass (arrows) to have slightly lobulated margins. C. Axial diffusion-
weighted image (b = 1000 s/mm2) shows the mass (arrows) to be hyperintense, indicated reduced diffusion compared with cerebral parenchyma. D. Axial
FLAIR image shows the mass (arrows) is hyperintense compared with CSF, differentiating it from an arachnoid cyst.

On MRI, dermoids have hyperintense T1 and hypointense T2 signal the split notochord syndrome. They are most commonly found in the
with associated chemical shift artifact, similar to lipomas (Fig. 7-36) (for spine and are discussed more completely in Chapter 9. Briefly, they are
discussion of intracranial lipomas, see Chapter 5). The high signal inten- thought to result from a persistent endodermal-ectodermal adhesion.
sity will disappear after fat suppression is applied (Fig. 7-37). However, Affected children typically present with headaches, gait disturbance,
dermoids are typically less lobulated than lipomas and will displace blood cranial neuropathies, sensorimotor deficits, or recurrent aseptic men-
vessels and nerves; lipomas engulf vessels and nerves (see Chapter 5). ingitis (231–233); however, they may be found incidentally (234).
Because affected patients often present with headaches from chemical Imaging studies show an extraparenchymal posterior fossa mass.
meningitis, inspection of the subarachnoid spaces and cerebral ventri- The most common locations are the cerebellopontine angle and the
cles for fatty droplets should be performed. Occasionally, the dermoid prepontine cistern (231,232); however, they may be found in the fourth
contains a solid or cystic component (which will have T1 hypointensity ventricle or in the supratentorial subarachnoid spaces (233). Rarely, as
and T2 hyperintensity compared with normal brain) or calcification in the spine, intraparenchymal extension may be seen in intracranial
(which has a variable signal on MRI [230]). Rarely, dermoid contain enteric cysts. The cysts have variable imaging characteristics, depending
hair and other components that lack mobile protons; in such cases, the upon the protein concentration. CT most commonly reveals sharply
dermoid tumor will appear completely black on MR images. demarcated, hypodense masses; occasionally, they may be isodense
or hyperdense to brain parenchyma (235) or show ring enhancement
Enteric (enterogenous) Cysts (233). On MRI, T1-weighted images typically show the cysts to be
Enteric cysts (also known as enterogenous, neurenteric, foregut, epi- hyperintense with respect to CSF and hypointense or isointense with
thelial, bronchogenic, endodermal, and respiratory cysts) are a part of respect to brain parenchyma (Fig. 7-38); they may be heterogeneous

Barkovich_Chap07.indd 681 5/6/2011 9:16:12 PM


682 Pediatric Neuroimaging

FIG. 7-36. Dermoid. A. Coronal T1-


weighted image shows a small mass of
diminished T1 relaxation time within the
right temporal lobe extending into the
temporal horn of the right lateral ven-
tricle (arrows). Note the dilatation of the
left temporal horn (arrow heads) and the
frontal horns (open arrows), indicative
of hydrocephalus. B. Axial T2-weighted
image shows fat floating within the fron-
tal horns of lateral ventricles (arrows) as a
result of rupture of the dermoid into the
ventricular system. Note the chemical-shift
artifact manifested as the low signal inten-
sity along the dorsal aspect of the dermoid
in the left frontal horn.

FIG. 7-37. Use of fat suppression in a


dermoid. A and B. Axial noncontrast CT
images show a fat-density mass (arrows)
expanding the left ambient cistern. C. Cor-
onal T1-weighted image shows a lobulated
heterogeneous hyperintense mass (large
open black arrows) in left ambient cistern,
curved around the free edge of the tento-
rium cerebelli. Note droplets of fat (small
black arrows) in the interhemispheric fis-
sure and sulci secondary to rupture of the
tumor into the subarachnoid space. D.
After application of fat suppression, the
hyperintensity of the fat (open black arrows)
is suppressed and the subarachnoid foci of
dermoid tumor appear hypointense.

Barkovich_Chap07.indd 682 5/6/2011 9:16:13 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 683

FIG. 7-38. Enteric (enterogenous) cyst. A. Sagittal T1-weighted image shows a slightly lobulated mass with diminished T1 relaxation time situated within
the premedullary space and extending through the foramen magnum into the ventral spinal subarachnoid space (arrowheads). B. Axial T1-weighted image
at the C1 level shows the mass sitting ventral within the spinal canal, slightly compressing the spinal cord (arrowheads).

(234). T2-weighted images typically show hyperintensity compared to as a-fetoprotein and b-human chorionic gonadotropin, which can
both CSF and brainstem, although when protein concentration is high, be measured in the CSF and serum. Grossly, teratomas are lobulated
they may be uniformly hypointense. Mild peripheral enhancement and variegated in appearance. The tumors tend to have both solid and
may be seen possibly due to an inflammatory reaction (233). Rarely, cystic components and frequently contain calcification or bone. More
a fluid-fluid layer, or even air, may be identified (231,236). Associ- malignant areas of tumor show less differentiation (8,76).
ated anomalies of adjacent bone are rare (233). Differential diagno-
ses include arachnoid cyst, which has an identical appearance when Imaging Studies
the enteric cyst contains clear fluid, and epidermoid tumor, which can On both CT and MRI, teratomas generally appear as sharply
usually be differentiated by the use of diffusion imaging (epidermoids marginated, heterogeneous lesions that are located in the midline. On
have diffusion characteristics intermediate between brain parenchyma CT, they can be reliably diagnosed if both fat and calcium are noted
and CSF, while enteric cysts have diffusion characteristics very similar within the mass; when fat and calcium are seen in a midline lesion
to CSF). with soft tissue intensity, the diagnosis of teratoma should be sug-
gested (Fig. 7-39A). On MRI, the presence of fat and soft tissue inten-
Teratomas sity together with punctate foci of low signal intensity (suggestive of
Intracranial teratomas are rare, and account for only 0.5% of primary calcium) in a midline mass should raise the suspicion of teratoma (Fig.
intracranial neoplasms. However, in children younger than 15 years 7-39B and C). The author has seen several patients, however, in whom
old, they account for 2% of intracranial tumors. Moreover, teratomas teratomas were of homogeneous soft tissue intensity on both CT and
represent a significant fraction of brain tumors that are diagnosed in MRI. Teratomas with a homogeneous imaging appearance are com-
infancy (see section on “Tumors in First Year of Life” later in this chap- monly malignant and are indistinguishable from many other types
ter). Intracranial teratomas are more common in males than females, of intrinsic brain tumors by imaging methods. Malignant teratomas
as a result of the preponderance of pineal teratomas in males. The clini- more often elicit vasogenic edema and also have more irregular shapes,
cal presentation depends upon the location of the tumor. Hydrocepha- less sharply defined margins, fewer cysts, and smaller areas of calcifi-
lus is common, as it is in other midline tumors (8,76). cation than benign teratomas (237). The degree of contrast enhance-
ment is variable. A lack of contrast enhancement suggests a low-grade
Pathology tumor; however, the presence of enhancement does not assure that the
Teratomas are most commonly found in the pineal and parapineal tumor is malignant. Further examples of teratomas will be shown in
regions, followed by the third ventricular region (especially its floor), the section on “Germ Cell Tumors” and in the section on “Tumors in
and the posterior fossa. Other than sacrococcygeal teratomas (see the First Year of Life.”
Chapter 9), teratomas are less common in the spine than intracrani-
ally; they may occur at any spinal level and are associated with spina Miscellaneous Tumors of the Posterior Fossa
bifida occulta. Most teratomas are well circumscribed and benign, but and Skull Base
some contain primitive elements and are highly malignant neoplasms Other tumors that occur in the posterior fossa in children are
that carry a poor prognosis. Some secrete biochemical markers, such extremely rare.

Barkovich_Chap07.indd 683 5/6/2011 9:16:15 PM


684 Pediatric Neuroimaging

FIG. 7-39. Teratoma. A. Noncontrast axial CT shows a dorsal posterior fossa


midline mass, which has a heterogeneous appearance (arrowheads). A large focus
of calcification is seen within the mass (arrow). Hydrocephalus is present, as
manifested by the enlarged temporal horns. B. Sagittal T1-weighted image shows
the teratoma to be of mixed signal intensity. Fatty components are of very high
signal intensity (arrow heads), soft tissue components are of soft tissue intensity
(solid arrows) and the calcification is of very low intensity (open arrow). C. Axial
T2-weighted image shows that the solid portion of the tumor is now of high signal
intensity whereas the fatty and calcified portions are now of low signal intensity.

Meningiomas histiocytoma, histiocytic lymphoma, and xanthoma disseminatum


Although meningiomas do occur in children, they are unusual and are (238–240). The most common histiocytic disorder to involve the CNS
most frequently found in the supratentorial space (particularly arising is LCH, a clonal proliferative disorder of cells of the mononuclear
from the choroid plexus in the lateral ventricular trigone) in associa- phagocytic and dendritic cell system of unknown pathophysiology
tion with Type 2 neurofibromatosis (see Chapter 6 and the section later (241,242). LCH can involve the bone, the meninges, the choroid plexus,
in this chapter). or brain parenchyma.

Langerhans Cell Histiocytosis Bone Involvement in the Face and Head When an osteolytic mass
A number of histiocytic disorders may involve the CNS, including involves the skull base, orbit, or facial bones in a child, the diagnosis
Rosai-Dorfman disease, Erdheim-Chester disease, malignant fibrous of LCH should always be considered and a CT or MR scan should be

Barkovich_Chap07.indd 684 5/6/2011 9:16:16 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 685

FIG. 7-40. Langerhans cell histiocytosis of the skull base. A. Axial CT image photographed with bone windows shows sharply demarcated defects (arrows)
in the squamous portion of the right temporal bone and in the midline of the occiput. B. Coronal contrast-enhanced CT scan shows enhancement (arrows)
of both lesions.

obtained to characterize the lesion and look for other signs of LCH. that enhances uniformly after administration of contrast (Figs. 7-40
Patients may present with tender, palpable skull masses, with proptosis, and 7-41). Lesions may be single or multiple; thus, other lesions, in the
or with evidence of cerebral or cerebellar dysfunction (175,220,241– calvarium and extracranial bones, should be sought by skeletal sur-
243). The most common imaging finding is that of a firm mass vey. MRI shows the bone lesions as sharply defined soft tissue masses
involving the calvarium, orbit, or skull base. CT shows a sharply cir- that have signal intensity comparable to skeletal muscle and enhance
cumscribed skull lesion, most commonly involving the temporal bone markedly after IV administration of paramagnetic contrast (Figs. 7-41

FIG. 7-41. Langerhans cell histiocytosis of the temporal bones. A. Axial CT with bone windows shows large, sharply marginated lytic defects (arrows) in the
mastoid and squamous portions of both temporal bones. B. Postcontrast axial T1-weighted image shows uniform enhancement (arrows) of the lesions.

Barkovich_Chap07.indd 685 5/6/2011 9:16:17 PM


686 Pediatric Neuroimaging

FIG. 7-42. MRI of Langerhans cell histiocytosis of the skull base.


A. Axial T1-weighted image shows a mass (open arrows) eroding
through the anterolateral wall of the right middle cranial fossa
(closed arrow). B. Axial T2-weighted image better defines the mass
(arrows), which has a hypointense rim. C. After administration of
contrast, the mass (arrows) uniformly enhances.

and 7-42) (241,244). Involvement of the mastoid is common and can deficits of insidious onset. Early cerebellar and long-tract signs and
mimic mastoiditis (241). symptoms may progress to profound neurological dysfunction with or
without intellectual deficits (175,243,244,247). Findings in the brain
Involvement of the Brain The most common signs and symptoms on imaging studies are variable. Affected patients may show diffuse
of LCH involving the CNS are those of diabetes insipidus from involve- or focal regions of T1 hypointensity and T2/FLAIR hyperintensity
ment of the pituitary stalk. An enhancing mass is typically seen in the in the brainstem and corpus medullare of the cerebellum (Fig. 7-43)
tuber cinereum or infundibulum (see section on “Sellar and Supra- (175,246,248). These findings reflect the histologic changes of demy-
sellar Masses”). Rarely, Langerhans cell histiocytosis can affect regions elination, reactive astrogliosis, microglia activation, and loss of cells in
of the brain other than the hypothalamus (175,245,246). Patients may the Purkinje and granular cell layers of the cerebellar cortex (249,250);
present with acute neurologic deficit or with progressive neurologic however, the MR findings do not correlate with clinical deterioration

Barkovich_Chap07.indd 686 5/6/2011 9:16:18 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 687

FIG. 7-43. Langerhans cell histiocytosis involving the pons and cerebel-
lar white matter. Axial T2-weighted image shows T2-prolongation in the
central pons (open arrows) and in the white matter of the cerebellar hemi-
spheres (closed arrows). These are the most common sites of parenchymal
involvement.

(251). Calcification may be seen in the cerebellar dentate nuclei on


CT. Diffuse atrophy and dilated perivascular spaces may be present
(246). Proton MR spectroscopy shows increased choline and reduced
NAA in the affected cerebellum, while 18-FDG PET shows reduced
uptake of glucose (252). Some patients have punctate lesions scattered
throughout the cerebrum, brainstem, and cerebellum. These punc-
tate lesions are granulomas, composed of Langerhans cells, microglial
cells, fibrillary astrocytes, CD1a+ histiocytes, CD68+ macrophages
and T and B cells (250); they show prolonged T1 and T2 relaxation
times compared with myelinated white matter and they enhance
markedly after intravenous infusion of contrast (175,243,244,247).
Typically, patients with cerebral parenchymal involvement also have
multiple lesions of the bone and most commonly have multisystemic
disease (249).

Chordoma
Chordomas are benign, slowly growing, locally infiltrative tumors that
arise from notochordal remnants. They are exceedingly rare in children
and only a few dozen cases are described in the literature. In contrast
to chordomas in adults, which are most common in the sacral region,
childhood chordomas are most common in the skull base and upper
cervical vertebrae; the most common location is at the sphenoid-
occipital synchondrosis (253,254). The behavior of these tumors in
children is similar to that in adults (255). Clivus chordomas usually
produce marked destruction of the sphenoid bone; they commonly
extend into the sphenoid sinuses, nasopharynx, and occasionally the
FIG. 7-44. Clivus chordoma. A. Sagittal T1-weighted image shows a large
ethmoid region. Patients with chordomas most frequently present with
mass (large arrows) pushing the pons posteriorly and pushing the pituitary
cranial neuropathies (diplopia, palatal or tongue weakness) resulting gland (small arrows) anteriorly. The displacement of the pituitary implies an
from extension of tumor into the cranial neural foramina. Headache, origin within the clivus. B. Postcontrast T1-weighted image shows a nearly
pyramidal signs, and symptoms of increased intracranial pressure may complete lack of enhancement of the tumor (arrows). C. Axial T2-weighted
also be present (254). On imaging studies, destruction of the clivus by image shows lateral displacement of both temporal lobes (arrows) and pos-
the tumor mass is readily apparent (256) (Figs. 7-44 and 7-45). When terolateral displacement of the pons by the hyperintense mass.

Barkovich_Chap07.indd 687 5/6/2011 9:16:20 PM


688 Pediatric Neuroimaging

FIG. 7-45. Clivus chordoma. A. Postcontrast sagittal T1-weighted image shows a large, mostly hypointense mass that has arisen from and destroyed most
of the basiocciput (white arrows). The lack of enhancement is common in pediatric chordomas. The pons (p) is pushed upward and the medulla (m) is
pushed dorsally and compressed by the mass. The fourth ventricle (arrowhead) is pushed posteriorly and compressed. BT. Axial T2-weighted image shows
prolonged T2 relaxation time within the tumor. The fourth ventricle (white arrows) is makedly pushed back. C. Axial T1-weighted image shows the bulky
hypointense mass distorting the brainstem and cerebellum and invading the clivus (white arrows). D. Axial FLAIR image shows the tumor (arrows) to be
somewhat heterogeneous.

tumor extends into the nasopharynx, it is often difficult to differentiate prolonged T1 and T2 relaxation values whereas chondroid chordomas
from a primary nasopharyngeal tumor extending into the clivus. The (a histological variant that have a significantly better prognosis [257])
presence of spicules of bone within a minimally enhancing tumor on have less T2 prolongation. FLAIR images often show more heterogene-
CT should suggest the diagnosis of chordoma. The MR appearance of ity (Fig. 7-45D) than T2-weighted images (Fig. 7-45B). Enhancement
chordoma is that of a large intraosseous mass extending into the pre- is variable, ranging from minimal to marked; in the author’s experi-
pontine cistern, sphenoid sinuses, middle cranial fossa, or nasopharynx. ence, chordomas of childhood (Figs. 7-44 and 7-45) rarely enhance. No
Considerable posterior displacement of the brainstem may be present known correlation exists between degree of enhancement and tumor
when the tumor is large (Figs. 7-44 and 7-45). Typical chordomas have grade or prognosis.

Barkovich_Chap07.indd 688 5/6/2011 9:16:21 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 689

FIG. 7-46. Chondrosarcoma of the skull base in a child. A. Axial


T1-weighted image shows a mass (white arrows) arising from the
petroclival synchondrosis and extending medially into the clivus and
laterally into the petrous apex. B. Fat-suppressed T2-weighted image
shows that the tumor (arrows) has prolonged T2 relaxation time. C.
Fat-suppressed T1-weighted image shows uniform enhancement of
the mass (arrows).

Chondrosarcomas Extension into the cavernous sinus, sella turcica, sphenoid sinus, or
Very rarely, chondrosarcomas arise from the skull base in children. parapharyngeal space is common (261). On imaging studies, chon-
Chondrosarcomas are malignant cartilaginous tumors that may arise drosarcomas are heterogeneous (59% on MR, 44% on CT) second-
de novo or from sarcomatous changes in benign cartilaginous tumors, ary to mineralization and prominent fibrocartilagenous elements.
such as chondromas or chondroblastomas. Only about 10% of chon- The nonosseous elements have long T1 and T2 relaxation times and
drosarcomas present in the pediatric age group (258,259); the mean enhance heterogeneously after intravenous contrast administration.
age of presentation is in the third decade (220,260). A disproportion- As the tumors often lie adjacent to fat in the marrow, infratempo-
ate percentage (43%) of chondrosarcomas in children are high-grade ral fossa, and deep facial regions, the use of fat saturation pulses is
neoplasms with “unconventional” histology, most commonly the mes- helpful on postcontrast studies. These tumors are nearly impossible to
enchymal type (258,259). Patients typically present with headache, confidently differentiate from chordomas on imaging studies, as the
sinus symptoms, proptosis, or cranial neuropathies. The most com- appearance is essentially identical (Fig. 7-46). However, paramedian
mon locations are the petrooccipital, sphenooccipital, and sphenoeth- origin from the petroclival synchondrosis and uniform enhancement
moidal synchondroses, presumably arising from cartilaginous rests. favor the diagnosis of chondrosarcoma.

Barkovich_Chap07.indd 689 5/6/2011 9:16:22 PM


690 Pediatric Neuroimaging

FIG. 7-47. Metastatic neuroblastoma to skull base. A. Sagittal T1-weighted image shows elevation of the pituitary gland (arrows) by a mass that is isointense
to the basisphenoid bone. B. Postcontrast coronal T1-weighted image shows elevation of the periosteum (arrows) from the sphenoid bone by the tumor.

Neuroblastoma useful to reduce intraoperative blood loss. Radiographic examination


When neuroblastoma presents with signs or symptoms referable to shows an expansile calvarial lesion with classic “soap bubble” rim of
the skull and brain, it is most commonly with proptosis or a scalp calcium around the lesion. CT shows a multilobular mass confined
mass secondary to metastasis to the orbital wall or calvarium (dis- by a smooth, fine peripheral layer of bone; a fluid-fluid layer may be
cussed later in this chapter). The involvement of brain parenchyma seen within the soap bubble calcification. Direct coronal CT images
by neuroblastoma is very rare, occurring in only 1% of cases (262). may be useful to establish that the lesion is intraosseous. MRI shows
Sometimes, neuroblastoma can originate in the upper cervical or skull an expansile calvarial mass with a low intensity rim and smooth
base portions of the sympathetic chain. These primary neuroblasto- margins. T1-weighted MR images show intrinsic heterogeneity with
mas present as homogeneous, uniformly enhancing extraparenchy- intensity similar to that of gray matter. T2-weighted MR images show
mal masses in the cerebellomedullary or cerebellopontine angles. An heterogeneity and, often, fluid-fluid layers; the lower (dependent)
extracranial, as well as intracranial, component will often be present, layer is very hypointense and the upper (nondependent) layer vari-
giving a clue to the correct diagnosis. Neuroblastoma primary to other ably hyperintense (265,268). Other features include round to ovoid
parts of the body (abdomen, pelvis, or chest) may present with cra- regions of very high signal intensity, probably representing areas of
nial neuropathy as a result of metastasis to the skull base (Fig. 7-47). blood or areas of serum with a high protein concentration (269). This
The tumor appears as an enhancing, infiltrative mass that elevates the appearance is no different than that seen in adults. Sagittal and coro-
periosteum from the bone. Identification of a mass or of periosteal nal images are often useful to determine that the mass originates from
elevation is important in making the diagnosis of metastatic tumor to the bone.
the skull base, as the normal infant’s skull base contains hematopoi-
etic marrow that enhances (263). As a result, enhancement of marrow Ewing Sarcoma
in an infant or young child with neuroblastoma is not sufficient to Very rarely, Ewing sarcoma can arise primarily from the skull. The most
establish a diagnosis of metastatic disease. Ewing sarcoma (both pri- common location of origin is the temporal bone, followed by fron-
mary and metastatic [264]) and leukemia, which can also involve the tal, parietal, and occipital bones. Patients typically present with signs
skull base, all have identical neuroimaging appearances. Metastatic and symptoms of an intracranial mass: headache, vomiting, blurred
neuroblastoma to the skull base, therefore, cannot be differentiated vision, ataxia, or cranial neuropathy. Imaging shows an extraparenchy-
from them. mal mass that has typical characteristics of a small round cell tumor;
the mass is isodense to slightly hyperdense on noncontrast CT and
Aneurysmal Bone Cyst nearly isointense to cortical gray matter on noncontrast MRI. Diffuse
Only 2% to 6% of aneurysmal bone cysts involve the calvarium enhancement may be seen after intravenous administration of contrast
(265,266). When they do, the orbital and occipital bones are most material (270,271). Sometimes the periosteum may be lifted off of the
commonly involved. Affected patients may present with proptosis, a inner table of the calvarium, resulting in an “onion skin” appearance
tender, enlarging mass on the head, or with signs of increased intrac- or coarse calcification on CT. Cystic regions can sometimes be found
ranial pressure, the latter sometimes associated with cerebellar dys- within the mass. A key finding is the involvement of the overlying cal-
function or cranial neuropathies (265,267). It is important to identify varial bone, giving evidence that it is a primary tumor of bone. This
that these lesions originate in the calvarium and to suggest the diag- appears as permeative destruction on CT and as alteration of the nor-
nosis of aneurysmal bone cyst, as preoperative embolization may be mal absence of bone signal on MRI (271).

Barkovich_Chap07.indd 690 5/6/2011 9:16:23 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 691

SUPRATENTORIAL TUMORS Hemispheric Astrocytomas


Pilocytic Astrocytomas, Astrocytomas, Anaplastic
As previously stated, supratentorial tumors are more common than
Astrocytomas, Glioblastomas
infratentorial tumors in children less than 2 years and older than
10 years of age. Astrocytomas of the cerebral hemispheres constitute approximately
30% of supratentorial brain tumors in children. Most series show
a slight male predominance. All pediatric age groups are affected,
Tumors of the Cerebral Hemispheres
although there is a slight peak at 7 to 8 years of age. The presenting
Astrocytomas are clearly the most common type of cerebral hemi- signs and symptoms depend upon tumor location. Seizures, focal
spheric tumor of childhood. However, other diagnoses should be neurological deficits, and symptoms of increased intracranial pres-
strongly considered when hemispheric tumors are found in neonates sure (headache, vomiting, and altered sensorium) are the most com-
and infants (Table 7-7). In neonates, teratoma should always be con- mon presenting symptoms, but ocular/ophthalmological signs and
sidered, especially if the tumor is heterogeneous or has calcium or fat abnormal cognition may be present (8,76,272–275). Seizures are likely
within it. In infants, atypical teratoid/rhabdoid tumor, ependymoma, to be the presenting symptom when the tumor is located in the frontal
desmoplastic infantile ganglioglioma, and primitive neuroectodermal or temporal cortex (276).
tumor should be considered in the diagnosis if the solid portions of the
tumor have a high attenuation on CT or if reduced diffusion and signal Pathology Grossly, hemispheric astrocytomas are similar to cerebellar
intensities similar to gray matter are found on MRI. Finally, if a child astrocytomas; they can be solid, solid with a necrotic center, or cystic
with a predominantly cortical tumor presents with a longstanding with a mural nodule. Most pediatric hemispheric astrocytomas have a
seizure history, the diagnoses of ganglioglioma and dysembryoplastic low histologic grade, although GBM does occur in childhood and has a
neuroepithelial tumor should be considered. better prognosis than in adults (277,278). JPAs are less common in the

TABLE 7-7 Cerebral Hemispheric Tumors in Children


Tumor Key Findings
Astrocytoma Most common tumor
MR appearance varies with grade/histology
Giant cell tumor Develop from ventricular wall
Usually near foramen of Monro
Ependymoma Peritrigonal, heterogeneous
Primitive neuroectodermal tumor Young children, heterogeneous
Solid part: gray matter intensity
Astroblastoma Children and teens. Peripheral hemisphere.
Lobulated. Solid/cystic. Little edema.
Solid part: gray matter intensity on T2
Mixed neuronal-glial tumors Cortical location
Calcification, cysts common
Desmoplastic neuroepithelial tumors Young infants. Large cysts, solid part invades dura
Dysembryoplastic neuroepithelial tumor Cortical location,
Marked T2 hyperintensity
Atypical teratoid/rhabdoid tumor Young infants. Often large at presentation.
Cortical intensity on T2, cysts
Medulloepithelioma No enhancement
Plasma cell granuloma Hypointense on T2 images
Meningioangiomatosis Hypointense with hyperintense
periphery on T2 images
Germinoma Basal ganglia
Heterogeneous with solid and cystic areas; solid portions
isointense to gray matter on T2 and uniformly enhancing
Postransplant lymphoproliferative Multiple foci of gray matter intensity on T2 images, slightly
disorder Immunosuppressed children hyperintense to gray matter on FLAIR
Marked vasogenic edema
Markedly enhance

Barkovich_Chap07.indd 691 5/6/2011 9:16:24 PM


692 Pediatric Neuroimaging

FIG. 7-48. Cystic astrocytomas. A. Axial postcon-


trast CT shows a large cyst (open arrows) with an
enhancing mural nodule (closed arrow). When cysts
are this large, it is difficult to determine whether
they are intraparenchymal or extraparenchymal.
Therefore, contrast should be administered to all
patients with intracranial “cysts” to rule out the
presence of an enhancing mural nodule. B. Non-
contrast coronal T1-weighted image shows a small
cyst (arrows) in the temporal lobe. C. Postcontrast
T1-weighted image shows enhancement of a mural
nodule (arrows) within the cyst.

cerebral hemispheres than in the cerebellum and, therefore, the over- or not at all. The tumor can be completely solid, solid with a central
all prognosis of supratentorial astrocytomas is somewhat worse. The area of necrosis, or partially cystic with an enhancing solid component.
tumors have no characteristic location, although they tend to occur When cysts are large, it is often difficult to determine whether they
deep within the hemispheres, more frequently involve the basal gan- are intraparenchymal or extraparenchymal. Therefore, it is impor-
glia or thalamus than do adult tumors, and may involve more than tant to administer contrast when cysts are detected. The presence of
one lobe of the brain (8,76,272,274,279). They are often very large at an enhancing mural nodule (Fig. 7-48) makes the diagnosis of cystic
the time of presentation. Pediatric astrocytomas, even those of low his- astrocytoma likely. It is not possible to differentiate benign from malig-
tologic grade, may be multicentric at the time of presentation; this is nant astrocytomas of the cerebrum by CT criteria alone. In general,
most likely the result of subarachnoid or intraventricular tumor spread malignant tumors tend to be larger, their solid components have higher
(280,281). Multicentricity is most common when the primary tumor attenuation, and they occur in younger children but these differences
site is the hypothalamus (hypothalamic tumors are discussed in a later are not significant enough to help determine pathology in individual
section of this chapter). cases (282). Occasionally, hemorrhage is seen in high-grade supraten-
torial astrocytomas (10).
Imaging Studies CT of the brain reveals considerable variation in On MRI, astrocytomas typically appear as large, medially located
tumor appearance. The solid portion of the tumor tends to be isodense hemispheric masses that have prolonged T1 and T2 relaxation values
to hypodense prior to intravenous contrast administration. After infu- when compared with normal brain (Figs. 7-49 and 7-50). Although typ-
sion of contrast, the solid portion may enhance completely, partially, ically found deep within the hemisphere, they can occur in the cerebral

Barkovich_Chap07.indd 692 5/6/2011 9:16:24 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 693

FIG. 7-49. Pilocytic astrocytoma. A. Sagittal T1-weighted image shows a mass that is isointense with gray matter located in the left thalamic region
(arrows). Note that the anterior portion (arrowheads) is more hypointense, suggesting that it might be a cyst. B and C. Axial FLAIR (B) and T2-weighted
(C) images show the mass to have varying components with variable degrees of hyperintensity. D. After infusion of paramagnetic contrast, the varying
components of tumor are seen to include a tumor cyst (nonenhancing and the most hypointense, c), a proteinaceous cyst (mildly enhancing and slightly
hyperintense, p), a solid component (brightly enhancing, s), and a microcystic enhancing component (black arrows).

white matter or in the cortex. In general, pilocytic astrocytomas are be separated from homogeneous, solid portions of tumor by FLAIR and
very hyperintense on T2-weighted and FLAIR images, elicit little or DWI: cysts have similar appearance to CSF. Low grade astrocytomas of
no vasogenic edema in the surrounding white matter, have areas that other histologies than pilocytic are homogeneous, free of hemorrhage,
enhance quite markedly after administration of paramagnetic contrast, well circumscribed, nonenhancing masses associated with mild or
and are associated with cysts of all sizes (Figs. 7-49 and 7-50). Cysts can no surrounding edema (Fig. 7-51). In contrast, higher grade tumors

Barkovich_Chap07.indd 693 5/6/2011 9:16:25 PM


694 Pediatric Neuroimaging

FIG. 7-50. Pilocytic astrocytoma. A. Sagittal T1-weighted


image shows an ovoid thalamic mass with an anterior
(solid) component that is slightly hypointense compared
to surrounding brain and a more hypointense (cystic)
posterior component. B and C. Axial first and second
echo T2-weighted images show that the anterior solid
portion of the tumor is very hyperintense on both echoes.
D and E. Postcontrast T1-weighted images show uniform
enhancement in the anterior solid portion of the tumor.

FIG. 7-51. Grade II astrocytoma. T2-weighted (A), FLAIR (B), and postcontrast T1-weighted (C) images show that the tumor (arrows) is homogeneous
(no hemorrhage or necrosis), sharply marginated, elicits no vasogenic edema, and does not enhance.

Barkovich_Chap07.indd 694 5/6/2011 9:16:27 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 695

FIG. 7-52. Glioblastoma multiforme. A. Axial T2-weighted image shows a heterogeneous, partially cystic or necrotic mass in the right temporo-occipital
region with moderate peritumoral edema and significant mass effect upon adjacent structures. B. Postcontrast axial T1-weighted image shows moderate
rim enhancement and focal hypointensity in the right anterior portion (arrow) probably representing flow voids from feeding vessels.

show heterogeneity, resulting from areas of necrosis or hemorrhage, SEGAs of tuberous sclerosis is better than that for patients with giant
and generate more extensive vasogenic edema (283) (Fig. 7-52). These cell components of ordinary astrocytomas (290), even though both are
characteristics are not absolute; however, low-grade tumors can have considered grade I astrocytomas by the World Health Organization.
some high grade characteristics and vice versa. Enhancement patterns Indeed, there is some question whether the tumors associated with
after paramagnetic contrast infusion are similar to those seen on CT tuberous sclerosis may have a different histology and should be clas-
after infusion of iodinated contrast (Figs. 7-49 and 7-50). sified separately from other astrocytomas with giant cell components
DWI is of some use in that solid areas of lower grade tumors (291). Males and females are equally affected. The tumor may be diag-
tend to have less tightly packed cells solid than areas of higher grade nosed at any age, but peak occurrence is between ages 5 and 10 years.
tumors and, therefore, have higher diffusivity (284–287). Unfortu- When the affected child does not have tuberous sclerosis, the clinical
nately, higher-grade tumors more commonly undergo necrosis, which presentation is almost always that of hydrocephalus. Rarely, the tumors
increases diffusivity and diminishes the value of this technique. can undergo malignant degeneration and invade surrounding brain
As stated and illustrated in the first section of this chapter, some evi- (8,76,289,292).
dence suggests that lower NAA/Cho ratio on proton MR spectroscopy
of glial tumors correlates with higher tumor grade (45,109); however, Pathology SEGAs usually arise from the wall of the lateral ventricle
pilocytic astrocytomas also have low NAA/Cho ratios (164), making near the foramina of Monro and produce hydrocephalus because of
this ratio rather meaningless if used in isolation. In addition, inflam- obstruction at the foramen. They are believed to arise from the sub-
matory and dysplastic lesions may have spectroscopic characteristics ependymal hamartomas that are part of the tuberous sclerosis complex
identical to that of malignant neoplasms (165). Other evidence sug- (see Chapter 6); in fact, the tumors bear histological similarity to the
gests that increased blood volume within the tumor as demonstrated subependymal hamartomas. Giant cell astrocytomas tend to be sharply
by perfusion imaging (54,109) correlates with higher tumor grade and defined and homogeneous on pathological examination. They usually
worsened survival (288). All of these characteristics, when combined are mostly intraventricular, but they may also develop deep in the brain
with imaging, can help to characterize a mass as a neoplasm. However, tissue. Focal calcification is common. Although usually histologically
determination of the histology of the neoplasm still requires biopsy benign, some show evidence of anaplasia (76,289). They may need to
or resection. At the time of the writing of this book, no characteristic be removed when they result in hydrocephalus. Recently, the use of
features have been described that allow one to confidently differentiate rapamycin or rapamycin-analogs has been shown to reduce the volume
pediatric astrocytomas from ependymomas or oligodendrogliomas on of the tumor; however, the tumor expands again when the treatment is
the basis of any MR characteristics. discontinued (293).

Subependymal Giant Cell Astrocytoma Imaging Studies The CT and MR findings of tuberous sclerosis are
Subependymal giant cell astrocytoma (SGCA or SEGA) is the name described in Chapter 6. The characteristic CT appearance of the sub-
given to a mass that is characteristically associated with tuberous scle- ependymal giant cell astrocytoma is that of a hypodense to isodense,
rosis. As these tumors occur in 5% to 15% of patients with tuberous well demarcated, rounded lesion originating from the ventricular wall,
sclerosis (289), discovery of a giant-cell tumor should motivate a search most commonly in the region of the foramen of Monro, and usually
for other manifestations of this condition (see Chapter 6). SEGAs may producing hydrocephalus (Fig. 7-53). Other subependymal masses,
occasionally be found in other settings. The prognosis for patients with both calcified and noncalcified, are usually present in the setting of

Barkovich_Chap07.indd 695 5/6/2011 9:16:29 PM


696 Pediatric Neuroimaging

FIG. 7-53. Giant cell astrocytoma. A. Noncontrast CT scan through the level of the foramen of Monro shows a mass originating from the foramen of
Monro (arrows) that is essentially isodense with brain. There is moderate vasogenic edema surrounding the mass. B. After infusion of iodinated contrast,
the mass uniformly enhances (arrows).

tuberous sclerosis. After the infusion of IV contrast, giant cell tumors The temporal lobe is the most common site (∼50%). Involvement of
always uniformly enhance (10,294). Examination with MR also reveals the parietal lobe (15%–20%), frontal lobe (10%) and occipital lobe
a well circumscribed, ovoid or rounded mass, most commonly in the (5%–10%) is less common (300). Rarely, other gray matter structures
region of the foramen of Monro, and often extending into the adja- such as the basal ganglia, cerebellum, or spinal cord may be the site of
cent lateral ventricle. Typically, the mass is hypointense to isointense origin (299).
with brain tissue on T1-weighted sequences (Fig. 7-54) and hyperin-
tense on T2-weighted sequences (292,295–297). Multiple smaller sub- Pathology Pathologically, PXAs arise in the periphery of the cere-
ependymal nodules are almost always present if the child has tuberous brum and often extend into and involve the leptomeninges, although
sclerosis. Moreover, on MRI, one typically sees multiple cortical invasion of the dura is rare. Cystic components are seen in about half
hamartomas that are identified by enlargement of the involved gyrus, of cases; the cysts may be quite large. Calcification is rare. Histologic
variable signal intensity on T1-weighted sequences and high signal examination shows pleomorphic spindle cells containing deposits of
intensity on T2-weighted sequences (see Chapter 6 and Fig. 7-54). intracytoplasmic lipid within a dense fibrotic reticulin network. Mono-
Infusion of paramagnetic contrast results in uniform enhancement of nucleated and multinucleated giant cells and eosinophilic granular
the giant cell tumor (Fig. 7-54B) (292,296). As stated in Chapter 6, bodies are present within the fibrosis (298–300).
giant cell astrocytomas are characterized by slow, progressive growth.
If rapid growth or invasion of cerebral parenchyma is seen in a lesion Imaging The classic appearance of a PXA is that of a well-cir-
originating in the region of the foramen of Monro, it should raise sus- cumscribed peripheral temporal lobe mass. Solid components are
picion of anaplasia. isodense to gray matter on CT and isointense to gray matter on
T1- and T2-weighted MR images (Fig. 7-55A and B), but in the
Pleomorphic Xanthoastrocytoma authors’ experience, diffusivity is not reduced, differentiating PXAs
Pleomorphic xanthoastrocytomas (PXAs) are in some ways very simi- from small round cell tumors such as PNET. They are hyperintense
lar to desmoplastic infantile gangliogliomas (DIG) and desmoplastic to gray matter on FLAIR images (Fig. 7-55C). The isointensity or
astrocytomas of infancy (DACI) (see section on “Neuronal and Mixed near-isointensity to gray matter helps to differentiate these from
Neuronal-Glial Tumors” below). All are tumors that are dispropor- low grade astrocytomas, which typically have higher water content
tionately common in young patients, typically arise peripherally in the and are more hypodense on CT, more hypointense on T1-weighted
cerebral hemispheres and involve the meninges, and have good prog- images, and more hyperintense on T2-weighted images. Intratumoral
noses despite histologic features that suggest high-grade malignancy. hemorrhage has been reported (Fig. 7-55A and B). The solid portion
However, unlike the desmoplastic tumors, PXAs are rarely diagnosed enhances homogeneously (Fig. 7-55D) or heterogeneously, depend-
in infants, being discovered most commonly in adolescents and young ing on whether microcystic change is present. Surrounding vasogenic
adults (298–300). In addition, affected patients most commonly pres- edema is usually minimal or absent. Invasion of the leptomeninges is
ent with seizures, probably because the cortex is nearly always involved. common, seen in more than two thirds of studies (Fig. 7-55D). As with

Barkovich_Chap07.indd 696 5/6/2011 9:16:29 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 697

FIG. 7-54. Giant cell astrocytoma. A. Coro-


nal T1-weighted image shows a mass in the
region of the left foramen of Monro (arrows)
and extending into the frontal horn of the
left lateral ventricle. B. After infusion of
paramagnetic contrast, the mass is seen
to uniformly enhance (arrows). C and D.
Axial T2-weighted image images near the
vertex show multiple subcortical areas of
prolonged T2 relaxation, compatible with
the cortical tubers that are characteristic of
tuberous sclerosis.

all slow growing, peripherally located cerebral tumors, erosion of the Neuronal and Mixed Neuronal-Glial Tumors
inner table of the calvarium is occasionally seen. Large or small cysts Gangliogliomas and Gangliocytomas
(Fig. 7-55) are present in about half of cases; enhancement of the cyst
wall is variable (301,302). If angiography is performed, the solid por- Gangliogliomas and gangliocytomas are tumors in which nerve cells and
tion of the tumor is found to be avascular; if seen, a vascular blush glial cells, usually astrocytes, participate in the neoplastic process; in this
indicates focal necrosis (303). sense, they differ from most primary CNS neoplasms in which only the
Differential diagnosis depends on the age of the patient. In infants, glial cells show neoplasia. Gangliogliomas and gangliocytomas (also called
DIG and PNETs can have similar appearances. Pilocytic astrocytomas ganglioneuromas) are uncommon tumors, constituting approximately
can be differentiated by the high water content of solid portion of 3% of brain tumors in children and approximately 6% of supratentorial
the tumor and ependymomas can be differentiated by their relatively pediatric brain tumors; they are found in older children and young adults
deep location in the cerebral hemisphere, in contrast to the superfi- more frequently than younger children or infants (77). Males are affected
cial location of the pleomorphic xanthoastrocytoma. In older children slightly more commonly than females (304,305). Although presentation
and adolescents, the main differential diagnoses would be high-grade occurs most often during the late first decade or early second decade of
astrocytomas, oligodendroglioma, and ganglioglioma. life (304), it is not uncommon for patients with these slowly growing

Barkovich_Chap07.indd 697 5/6/2011 9:16:30 PM


698 Pediatric Neuroimaging

FIG. 7-55. Pleomorphic xanthoastrocytoma. A. Sagittal T1-weighted image shows an extensive transcerebral mass that involves the cortex. The deep
solid component is isointense to the cortex, and contains hemorrhagic (hyperintense) areas. The peripheral cystic component also is isointense, probably
due to the presence of blood. B. Axial T2-weighted image shows the deep portion of tumor (arrows) to be isointense to the parenchyma, the presence of
hypointense recent blood (arrowheads) and the bright signal of the cystic areas. C. Axial FLAIR image shows that the solid component (arrows) is slightly
hyperintense, while the cysts (c) are strongly hyperintense. No peritumoral edema is found. D. Postcontrast T1-weighted image shows that the solid portion
of the tumor enhances significantly. Note that the enhancement extends into the sulci and involves the falcine dura (arrows).

neoplasms to present for evaluation in adulthood. The clinical history Pathology Mixed neuronal-glial tumors occur mainly in the cerebral
of affected patients is usually one of a protracted history of focal find- hemispheres; the temporal lobes are most commonly affected, fol-
ings. When the neocortex is affected, the history tends to be one of long lowed by the parietal lobe, frontal lobe, occipital lobe, third ventricle
standing focal epilepsy; half of affected patients present with seizures, (including pineal region), and hypothalamus (304,305). The cerebel-
most commonly partial complex epilepsy (304). Indeed, complete tumor lum, brainstem, and spinal cord can also be affected (172,304). The
removal is generally curative whatever the presentation (306, 307). When tumors tend to be small, firm, and well-defined; foci of calcification
the region of the third ventricle and hypothalamus is affected, symptoms and cysts are frequent. The differentiation between ganglioglioma and
(obesity, diabetes insipidus, or bulimia) tend to reflect hypothalamic dys- gangliocytoma is histological. Dysplastic neurons are a consistent find-
function. These tumors should not be confused with dysplastic cerebellar ing in these tumors; if neoplastic glial elements are present, the lesion
gangliocytomas (Lhermitte-Duclos syndrome), a congenital malforma- is called a ganglioglioma and when dysplastic neuronal elements are
tion described in Chapter 5 (8,308–311). found in the absence of neoplastic glia, the term gangliocytoma (or

Barkovich_Chap07.indd 698 5/6/2011 9:16:32 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 699

FIG. 7-56. Gangliogliomas.


A. Axial contrast-enhanced CT
scan shows a lesion of mixed sig-
nal intensity in the left occipital
lobe near the visual cortex. Both
calcifications (open arrows) and a
low-intensity area (closed arrows)
are present. Areas of low attenu-
ation in gangliogliomas are not
necessarily cystic. B. Coronal
postcontrast T1-weighted image
shows a right frontal mass that
causes pressure erosion of the
inner table of the calvarium.
Although the mass was entirely
solid at surgery, part of it
enhanced (solid black arrow) and
most (open black arrows) did not.

ganglioneuroma) is used (312). Temporal lobe gangliogliomas can be absent. Occasionally, the tumor can elicit surrounding edema (low
seen in association with mesial temporal sclerosis (313). attenuation). When the tumor is peripherally located within the cere-
brum, erosion of the adjacent inner table of the calvarium may occur
Imaging Studies Imaging findings of gangliogliomas and ganglio- (11,311,314).
cytomas are identical. The CT appearance is that of a low density, On MRI, the ganglioglioma may have sharply or poorly defined
well circumscribed lesion with little mass effect or edema, typically margins. The tumor may be solid (Fig. 7-56B), cystic (Fig. 7-57), cys-
located within the cerebral cortex (Fig. 7-56). Solid portions of the tic with a mural nodule, or have the appearance of many small cysts
tumor can be isodense, mixed, or hypodense; when hypodense, care (Fig. 7-58). Signal intensity on T1-weighted sequences is variable
should be taken not to assume that the hypodense area is a cyst. Cal- (and often mixed); T2-weighted sequences generally reveal high sig-
cification is seen in approximately 35% of gangliogliomas on CT. nal intensity. Regions of slight T1 hyperintensity, probably repre-
Contrast enhancement is variable; part or all of the solid portion of senting calcification, may be helpful in identifying these neoplasms
the tumor may enhance or contrast enhancement may be completely (Fig. 7-58). Solid portions of the tumor enhance variably (Fig. 7-56B).

FIG. 7-57. Ganglioglioma.


A. Axial T2-weighted image
shows a largely cystic, lobulated
mass (arrows) in the anterior
right temporal lobe. B. Axial
postcontrast T1 weighted image
shows that the lesion does not
enhance (arrows).

Barkovich_Chap07.indd 699 5/6/2011 9:16:33 PM


700 Pediatric Neuroimaging

FIG. 7-58. Ganglioglioma. A. Coronal


T1-weighted image shows a small cyst (small
straight arrow) in the right temporal cortex
with hyperintensity (curved arrows) in the
adjacent temporal white matter. B. Axial
T2-weighted image shows the hyperintense
tumor (arrows) in the right temporal cortex.

As on CT, the peripheral location within the hemispheres and the ero- The suggestion of DIG or DACI as the possible diagnosis in infants
sion of the adjacent calvarium can be helpful in making the diagnosis of with large, cystic, cerebral tumors containing peripheral, plaque-like
ganglioglioma (315). When located in the hypothalamic area, ganglio- solid components is important to clinical management because the his-
gliomas are difficult to differentiate from hypothalamic astrocytomas tologic characteristics may be confusing. In addition to the characteris-
(316). When located in cortex or subcortical white matter, differen- tic desmoplasia with astroglial and neuronal tumor cells, these tumors
tiation from astrocytomas, oligodendrogliomas, and dysembryoplastic often have a component of small, mitotically active cells that can be
neuroepithelial tumors (DNETs) is difficult. misinterpreted as malignant. Indeed, a number of reported patients
were initially treated aggressively with chemotherapy for high-grade
Desmoplastic Neuroepithelial Tumors (Desmoplastic Infantile malignancies before the correct diagnosis was made (323,329). DIG
Ganglioglioma/Desmoplastic Astrocytoma of Infancy) are differentiated from cystic astrocytomas by the peripheral location
The DIG and the DACI are very similar tumors seen primarily in and the relatively high density (CT) and T2 hypointensity (MRI) of the
infants and young children (317–322). Both are characterized by the solid tumor component as compared to the low density and long T2,
presence of both astrocytic and ganglionic cells with a prominent respectively, of astrocytomas. Other differential considerations, based
desmoplastic stroma (predominantly fibroblasts). Despite areas that on the neuroimaging appearance, include high-grade astrocytoma,
appear to have a high number of mitoses mimicking high-grade ana- pleomorphic xanthoastrocytoma, PNET, and ependymoma. If the lep-
plasia, the tumors demonstrate a surprisingly benign clinical course tomeningeal component of tumor is large, meningioma and meningeal
(319,320,322,323). In view of their histologic and clinical similarities, sarcoma are differential considerations.
some authors have suggested that these tumors have a common ori-
gin with different histological differentiation (324). As a result, several Dysembryoplastic Neuroepithelial Tumor
authors have suggested grouping these tumors together as desmoplas-
DNETs are benign masses of the cerebral cortex that nearly always pres-
tic neuroepithelial tumors (321,324,325). That grouping will be fol-
ent as partial complex seizure disorders in children or young adults;
lowed in this book.
other neurologic signs or symptoms are rarely present (330–332).
Patients with DIGs and DACIs typically present in infancy with
These are common and important tumors; recent series indicate that
macrocephaly or partial complex seizures, the median reported age
DNETs are the cause of as many as 20% of cases of medically refractory
of presentation being about 5 months of age (318–321,326); rarely,
epilepsy in children and young adults (333). The diagnostic criteria for
affected patients may present later in childhood (327). Evaluation
these tumors have evolved since the initial description of this tumor.
with neuroimaging reveals a large tumor arising in the cerebral hemi-
At present, the best criteria are as follows: (a) partial seizure disorder
sphere; typically the cysts are quite large and dominate the image, but
beginning before age 20 years; (b) no neurologic deficit or a stable con-
occasionally the cysts may be relatively small at the time of the initial
genital deficit; (c) cortical tumor location (334). Rarely, the tumors
imaging study (321). Involvement of multiple lobes is common with a
may develop in the deep parts of the cerebral hemisphere, particularly
predilection for the frontal and parietal lobes. A cortical solid compo-
in area of the caudate nucleus (335).
nent can always be identified; this solid component often involves the
leptomeninges (318,319,322) and, therefore, may appear extraparen-
chymal. The solid portion is iso- to hyperdense compared with gray Pathology Pathologically, greater than 60% of DNETs are found
matter on CT and is isointense to cortex on T1- and T2-weighted MR in the temporal lobe, approximately 30% in the frontal lobe, and
sequences. The solid portion enhances markedly after the administra- the remainder scattered through the parietal and occipital lobes, the
tion of contrast (Fig. 7-59). Enhancement is not seen in the walls of deep cerebral nuclei, brainstem, and cerebellum (330–332,334–336).
the cysts (326,328). Complete resection of the tumor may be curative, The tumors are solid but most have cystic or microcystic compo-
obviating the need for chemotherapy or radiation (329). nents with “floating” neurons (331). Many are adjacent to regions of

Barkovich_Chap07.indd 700 5/6/2011 9:16:34 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 701

FIG. 7-59. Desmoplastic infantile ganglioglioma. A and B. Axial T2-weighted images show multiple large parenchymal cysts (white arrows) in the left
cerebral hemisphere, displacing midline structures to the right. Low intensity solid material (black arrows) is seen in the lateral aspect of the cysts. C and D.
Postcontrast coronal T1-weighted images show that the lateral solid component (open white arrows) enhances uniformly.

cortical dysplasia (FCD Type IIIb), but it is not clear whether both are lobulated or multilobulated appearance (Fig. 7-61) (332,338–340).
developmental or whether seizures resulting from the tumor cause the These areas have variable signal on FLAIR images, ranging from
FCD. Many tumors that, in the past, were histologically classified as hypointense (appearing cyst-like) to hyperintense (332). Diffusivity
gangliogliomas or mixed oligo-astrocytomas would probably be char- of the tumor is elevated compared with normal gray and white mat-
acterized as DNETs if reviewed today. ter, probably because of its multicystic nature (340). Tumoral calcifica-
tion may be present in about one third of affected patients (330,336);
Imaging Studies Imaging studies show a very variable appearance, hemorrhage is rare (332). Because the lesions are primarily cortical and
which probably reflects significant biologic heterogeneity in those therefore superficial, convexity DNETs may erode the inner table of
lesions classified as DNETs. Despite this heterogeneity, almost all DNETs the calvarium (337) (Fig. 7-60). Subcortical extension is seen in about
are well-demarcated, lobulated cortical masses that are hypodense com- 30% (334,337,338); in such cases, the greatest diameter is at the cortical
pared to white matter on noncontrast CT. They are hypointense on T1 level, leading to development of a triangular shape when viewed in the
images and hyperintense on T2 images on MRI (Figs. 7-60 and 7-61) coronal plane (339). Intracortical lesions tend to be round or oval. Con-
(334,336–338). Cystic or microcystic components are present in about trast enhancement is seen in 20% to 40% and is typically patchy (339)
30–40%; they appear as sharply defined areas of very hypointense T1 but may be diffuse (334,337,338); enhancement patterns may vary of
and hyperintense T2 signal and often give the tumor a characteristic sequential studies (332).

Barkovich_Chap07.indd 701 5/6/2011 9:16:35 PM


702 Pediatric Neuroimaging

FIG. 7-60. Dysem-


bryoplastic neuroepi-
thelial tumor. A. Sagittal
T2-weighted image shows
a multilobular hyper-
intense homogeneous
mass in the base of the
left frontal lobe. Note the
scalloping of the orbital
roof (arrows), indicating a
slow growing, superficial
mass. B. Axial T1-weighted
image shows the tumor as
a sharply marginated, non-
enhancing, homogeneous
mass (arrows) in the left
frontal lobe.

FIG. 7-61. Dysembryoplas-


tic neuroepithelial tumor.
A. Parasagittal T1-weighted
image shows a cortical/sub-
cortical hypointense mass
(arrows) involving the pos-
terior frontal lobe. B and C.
Axial T2-weighted and post-
contrast T1-weighted images
show the multiple microcys-
tic components (arrows) that
compose the nonenhancing,
lobulated mass. D. Coro-
nal FLAIR image shows the
microcystic components and
the subcortical extension of
the tumor (arrows), seen in
about 30% of DNETs.

Barkovich_Chap07.indd 702 5/6/2011 9:16:36 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 703

FIG. 7-62. Supratentorial


ependymoma. A. Axial non-
contrast CT scan shows a
hemispheric mass (arrows)
that is isodense with gray
matter. A small focus of cal-
cification (open arrow) is
seen within the tumor mass.
There is mild surrounding
vasogenic edema. B. After
infusion of iodinated con-
trast, there is heterogeneous
contrast enhancement.

Proton MR spectroscopy of DNETs shows no significant difference Imaging Studies


of metabolite ratios from normal cortex other than myo-inositol/ As with the infratentorial ependymomas, those located supratentorially
creatine ratio, which is elevated (340). Dynamic susceptibility-weighted have an extremely variable appearance on both CT and MRI. They
perfusion imaging shows lower CBV than normal cortex (340). tend to be sharply demarcated tumors that are isodense to hyperdense
Metabolically, these are inactive tumors with no significant glu- compared with normal brain on noncontrast CT scans; foci of calcifi-
cose uptake on 18F-fluorodeoxyglucose PET or on 11C-methionine PET cation and cystic areas are identified in about half of affected patients
(341). Because of the marked variability in imaging appearance, the (Fig. 7-62). The solid portion of the tumor enhances variably after
diagnosis of DNET should be suggested any time a primarily corti- contrast administration. The diagnosis of ependymoma should be
cal tumor with high water content is seen in a patient with (a) a long entertained when an isodense frontal or parietal juxtaventricular mass,
standing history of partial epilepsy and (b) a normal neurologic exam with foci of calcification and cysts, shows heterogeneous enhancement
or a stable, long standing neurologic deficit (334). (11,154,345).
MRI generally shows large, heterogeneous tumors in a peritrigo-
Supratentorial Ependymomas
nal location; however, the tumors may be intraventricular or subcor-
Supratentorial ependymomas comprise between 20% and 40% of tical. The heterogeneity results from the combination of solid tumor
childhood ependymomas (151). They occur in males more commonly (isointense to gray matter on T1-weighted, T2-weighted, and FLAIR
than females with a peak incidence between ages 1 and 5 years. The images, diffusivity similar to normal brain tissue), intratumoral calci-
presenting symptoms depend upon the location of the tumor. Signs of fication, cysts, and, occasionally, hemorrhage, which combine to give
increased intracranial pressure (headache, vomiting and altered sen- a mixture of signal intensities with all imaging parameters (Figs. 7-63
sorium), ataxia, and focal seizures are the most common reasons for and 7-64) (152,346). However, supratentorial ependymomas can
presentation (8,151,154,274). appear homogeneous on MRI (and thus be indistinguishable from
low-grade astrocytomas) or ring enhancing with extensive edema
Pathology
(indistinguishable from a high-grade astrocytoma or a primitive neu-
Supratentorial ependymomas are identical histologically to their roectodermal tumor). CSF spread of tumor is rare, as most tumors are
infratentorial counterpart. Small foci of calcification are seen in about completely intraparenchymal in location (346).
one-half of cases; cystic areas are common, especially in the larger
lesions. Although pathology texts state that they are most commonly
located in the frontal lobe, where they usually abut the wall of the fron- Differential Diagnosis
tal horn, our experience at UCSF and the radiology literature suggests When composing a differential diagnosis for a supratentorial brain
that they develop most commonly in the parietal and temporoparietal tumor in a child, one should remember that ependymomas are most
regions. In contrast to infratentorial ependymomas, the supratentorial often seen in young children. Therefore, when a heterogeneous mid-
tumors are uncommonly intraventricular; metastatic spread of tumor line tumor is seen on MRI in a child in the first year of life (especially
through the CSF is, therefore, uncommon (8,75,76). As was mentioned in the first 6 months), the first diagnosis considered should be tera-
before about the posterior fossa ependymomas, the radial glia give toma. When heterogeneity is seen in a supratentorial tumor that is off
rise to the tumor stem cells of ependymomas. Therefore, supratento- the midline and extraventricular, ependymomas, PNETs, and ATRT
rial ependymomas often are located away from the ventricles; some should be considered as diagnoses, especially if the child is between
are strictly intracortical (31,33,155,342,343). Moreover, in contrast the ages of 1 and 5 years; both PNET and ATRT tend to have reduced
to astrocytomas, ependymomas have fairly well defined margins at diffusivity when compared to normal cerebral parenchyma. When a
their periphery; as a result, complete tumor resection is more likely markedly heterogeneous intraventricular tumor with periventricular
at surgery (344). extension is seen, the diagnosis of a choroid plexus carcinoma should

Barkovich_Chap07.indd 703 5/6/2011 9:16:38 PM


704 Pediatric Neuroimaging

FIG. 7-63. Supratentorial ependymoma with small


cysts and necrosis. A. Axial FLAIR image shows a
heterogeneous mass adjacent to the left trigone. The
hypointense regions (white arrowheads) are tumor cysts,
the hyperintense regions (black arrowheads) are areas of
necrosis. The solid portion of the tumor has signal char-
acteristics similar to gray matter. B. Axial T2-weighted
image does not distinguish between cysts and necrosis.
C. Postcontrast axial T1-weighted image shows that the
necrotic areas of the tumor (white arrows) enhance but
the cysts (black arrows) do not.

be suggested, although ependymomas may uncommonly have this ganglioneuroblastoma, medulloepithelioma, and ependymoblastoma
appearance. are classified as subtypes of the PNETs (73). PNETs are uncommon
tumors, probably comprising less than 5% of supratentorial neo-
Tumors with Neuroblastic or Glioblastic Elements
plasms in children, although their reported incidence varies widely,
Primitive Neuroectodermal Tumor probably because of variable willingness of pathologists to make
PNETs were first conceptualized by Hart and Earle (347) who defined this diagnosis and a changing understanding of the tumor type (77).
them as highly cellular tumors composed of greater than 90% to 95% Although they have been described in patients up to the age of 24
undifferentiated cells. Although foci of differentiation along glial or years, they are more commonly seen in children under the age of 5
neuronal lines may be present within the tumor mass, the high percent- years. Seizures, focal neurological deficits, and symptoms of increased
age of undifferentiated cells theoretically sets these tumors apart from intracranial pressure (headache, vomiting and altered sensorium) are
other tumors. PNETs are histologically similar to medulloblastomas, the most common presenting symptoms. No male or female predomi-
pineoblastomas, ATRT, and peripheral neuroblastomas. Many nance has emerged. Patients most frequently present with macroceph-
neoplasms that were previously classified as primary cerebral neu- aly (if diagnosed in infancy) or signs of increased intracranial pressure
roblastomas, primary cerebral medulloblastomas, ependymoblasto- or seizures (349–351).
mas, and undifferentiated small cell neoplasms of the brain are now
considered by many to be PNETs, although some controversy exists Pathology Cerebral PNETs most frequently occur in the deep cere-
regarding the classification of these neoplasms (76,348). In the last bral white matter and are usually quite large at the time of presentation.
WHO Classification of Brain Tumors, CNS neuroblastoma, CNS Grossly, they have sharp margins, even though histological examination

Barkovich_Chap07.indd 704 5/6/2011 9:16:39 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 705

FIG. 7-64. Supratentorial ependymoma


with large cysts. A. Axial T2-weighted
image shows a heterogeneous cystic and
solid mass in the left temporal lobe. B.
Postcontrast axial T1-weighted image
shows the solid portion of the tumor
to enhance heterogeneously. The cysts
(arrows) are hyperintense compared to
ventricles and are therefore tumor cysts.

shows spread of the tumor cells peripherally beyond the apparent The most typical MR appearance of a PNET is that of a large,
tumor edge. Necrotic areas and foci of calcification are seen in about apparently sharply marginated mass that can be located either in the
one-half of tumors. Seeding of tumor through the CSF pathways and cerebral hemisphere or the lateral ventricle (Figs. 7-65 to 7-67). As with
metastases to the spinal cord, lungs, liver, and bone marrow has been ependymomas, the tumors are remarkable for their marked variability;
reported (8,76,351). appearance ranges from solid (Fig. 7-66) to cystic (Fig. 7-67) and from
homogeneous to markedly heterogeneous to a rim of solid tumor sur-
Imaging Studies On noncontrast CT, the solid portions of the PNET rounding central necrosis (Fig. 7-65). Solid portions are typically isoin-
tend to be hyperdense when compared to white matter, presumably tense to gray matter on T2-weighted and FLAIR images (354). Punctate
due to the high nuclear to cytoplasmic ratio and the subsequent high calcifications may be inapparent or appear as foci of low signal inten-
electron density of the tumors (Fig. 7-65). Cystic areas and punctate sity. Solid portions of the tumor show reduced diffusion on DWI (354)
calcifications are common, and hemorrhage is seen in approximately and increased blood volume on susceptibility-weighted perfusion
10%. After intravenous infusion of iodinated contrast, some enhance- studies (355) compared to normal surrounding brain. Cystic areas have
ment is always seen; it may be solid and homogeneous, heterogeneous, low intensity on T1-weighted sequences, have very high intensity on
or ring-like, depending upon the size and number of associated cysts T2-weighted sequences, and show increased diffusion. FLAIR images
and necrotic (350,352,353). help to differentiate necrosis (hyperintense) from cysts (hypointense)

FIG. 7-65. Primitive neuroectodermal tumor. A. Axial noncontrast CT scan shows a large heterogeneous mass principally in the left frontal lobe with low
attenuation cystic/necrotic areas (c) and higher attenuation (similar to cortex) solid areas (s). B. Axial FLAIR image shows necrotic areas (n) that are mark-
edly hyperintense compared to both the solid and cystic components. Hyperintensity is seen in the corpus callosum (arrows), indicating probably tumor
spread into the right hemisphere. C. Axial postcontrast T1-weighted image shows moderate enhancement (arrows) of the solid portion of the tumor.

Barkovich_Chap07.indd 705 5/6/2011 9:16:40 PM


706 Pediatric Neuroimaging

FIG. 7-66. Nearly homogeneous primitive neuroec-


todermal tumor. A. Axial T2-weighted image shows a
left parietal mass (arrows) that is largely isointense to
gray matter. The mass appears well demarcated and
elicits little vasogenic edema. Some central hypoin-
tensity may represent calcium or blood. B. Axial
T1-weighted image shows the central area as hyper-
intense (arrows), again suggesting the presence of
calcium or blood. C. Postcontrast axial T1-weighted
image shows enhancement of only the central portion
(arrows) of the tumor. The enhancement suggests that
this area was undergoing necrosis and that the abnor-
mal signal on the noncontrast exams was blood due
to the necrosis.

(Fig. 7-65B). When hemorrhage is present (Fig. 7-66), it is of high sig- vomiting secondary to increased intracranial pressure (76,357). These
nal intensity on the T1-weighted and FLAIR sequences and of variable tumors most commonly develop in the periventricular region of the
intensity on the T2-weighted sequences depending upon the chemical cerebral hemispheres, but may develop in the suprasellar region (358),
state of the iron. Infusion of paramagnetic contrast results in variable cerebellum, or brainstem. When the tumors develop in the posterior
enhancement, similar to that seen on CT (356). When a large, sharply fossa, presenting signs and symptoms may include cranial neuropathy,
marginated mass is seen in a young child, the diagnosis of PNET should hemiparesis, or even coma (357).
be suggested, particularly when the mass is markedly heterogeneous.
Differential diagnosis includes high grade glioma, ependymoma, and Pathology Medulloepitheliomas are characterized histologically by
ATRT. the papillary, tubular, or trabecular arrangement of cells with an exter-
nal and internal limiting membrane that mimics the structure of the
Medulloepithelioma primitive neural tube (76,357). These are highly malignant tumors
Medulloepithelioma, a subtype of PNET in the last WHO classification with a high incidence of intratumoral hemorrhage histologically and a
of tumors of the CNS (73) is the name given to a very rare tumor that high incidence of recurrence after resection. The prognosis is very poor
typically presents in the first 5 years of life with headache, nausea, and (76,357).

Barkovich_Chap07.indd 706 5/6/2011 9:16:41 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 707

FIG. 7-67. Intraventricular primi-


tive neuroectodermal tumor. A. Axial
T2-weighted image shows the tumor
(arrows) in the enlarged left occipital
horn. The tumor is isointense to gray
matter peripherally with a hyper-
intense center. B. Postcontrast axial
T1-weighted image shows heteroge-
neous enhancement of the mass. C.
Postcontrast coronal T1-weighted
image shows the marked enlargement
of the occipital horn more clearly. In
addition, spread of the tumor to the
superior wall of the ventricle (arrow)
is appreciated.

Imaging Studies Neuroimaging characteristics are similar to other included in the group of embryonal tumors of the CNS in the last
cerebral brain tumors, except that enhancement is uncommon after WHO classification, together with medulloblastomas and PNETs
administration of intravenous contrast material. Medulloepitheliomas (73). ATRTs of the brain are rare, accounting for 1.3% of primary
are typically well-circumscribed, homogeneous masses that are isodense CNS tumors in children and 6.7% of CNS tumors in children diag-
to slightly hypodense on noncontrast CT (10,357). MRI shows sharply nosed before the age of 2 years (77,118,129,359). Most reported cases
marginated masses that are hypointense on T1-weighted images and of cerebral ATRT have presented in the first decade of life (usually
hyperintense on T2-weighted images; slight internal heterogeneity may before the age of 4 years) with lethargy, vomiting, and visual distur-
be seen, particularly in large tumors. The lack of enhancement should bances. Prognosis continues to be dismal, with survival of less than
raise the possibility of a medulloepithelioma (357). 18 months in most cases (118–122,129,359). Some authors suggest
that gross total resection improves prognosis, but the tumors are typi-
Atypical Teratoid/Rhabdoid Tumor cally so large at the time of presentation and the locations so challeng-
of Infancy and Childhood ing that total removal is not possible (121). These tumors have been
ATRT are neoplasms of unknown histogenesis that were originally discussed in the section on “Tumors of the Posterior Fossa”; however,
described in the kidney and were named because their light micro- approximately half of ATRT of the CNS arise in the supratentorial
scopic appearance resembles that of rhabdomyosarcoma. They are compartment.

Barkovich_Chap07.indd 707 5/6/2011 9:16:42 PM


708 Pediatric Neuroimaging

FIG. 7-68. Atypical teratoid/


rhabdoid tumor. A. Axial non-
contrast CT image shows a
large heterogeneous mixed
hyperdense and hypodense
mass (arrows) in the left pos-
terior parietal lobe, with ante-
rior vasogenic edema. B. Axial
T1-weighted image shows the
heterogeneous mass (arrows)
appears grossly well margin-
ated. C. Axial T2-weighted
image shows that the solid por-
tions of the heterogeneous mass
are isointense to gray matter.
Hyperintense portions probably
represent necrosis and hypoin-
tense portions likely represent
hemorrhage. D. Postcontrast
T1-weighted image shows het-
erogeneous enhancement. The
imaging appearance of these
tumors is identical to those of
PNETs and ependymomas.

Pathology are typically solid with areas of necrosis, hemorrhage, or calcification;


Pathologically, ATRT are solid with regions of necrosis, hemorrhage, the solid regions are isodense to hyperdense to gray matter on CT
and cysts; margins are poorly defined. The characteristic histologic (Fig. 7-68) and isointense to gray matter on MRI (120,121,130,361).
feature is the rhabdoid cell, a round to ovoid cell with eccentric nuclei Areas described as cysts, but having the appearance of necrosis (irregu-
and prominent nucleoli, abundant eosinophilic cytoplasm, and cyto- lar walls, see Figs. 7-68 and 7-69), are present in nearly all at the time
plasmic hyaline inclusions. Rhabdoid cells are not abundant and can of presentation. Solid portions enhance heterogeneously after infusion
be overlooked unless sought. Other characteristic histologic features of intravenous contrast (121,129,359). Leptomeningeal tumor dissemi-
include abundant mitoses, malignant mesenchymal spindle-shaped nation is present in about 20% of patients at the time of presentation.
cells, cells with epithelial differentiation, and small round blue DWI reveals reduced diffusivity compared with surrounding white mat-
(PNET) cells. The presence of these varying cell types is responsible ter (121). Proton MR spectroscopy shows elevated choline and reduced
for the naming of the tumor as “teratoid/rhabdoid” (360). NAA, but these changes have not been quantified (121). The tumors can-
not be differentiated from ependymomas or PNETs on the basis of their
Imaging Studies imaging characteristics; as stated in the prior section, all three diagnoses
On imaging studies, ATRTs are usually large (averaging 5 cm in diame- should be included in the differential. ATRTs and ependymomas appear
ter) at the time of patient presentation (Figs. 7-68 and 7-69). The masses to be relatively more common in infants (below the age of 2 years).

Barkovich_Chap07.indd 708 5/6/2011 9:16:44 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 709

FIG. 7-69. Atypical teratoid/rhabdoid tumor. A. Axial


T2-weighted image shows an enormous bilobed right
frontal mass, eliciting vasogenic edema and subfalcine
herniation. The solid portion of the mass (arrows) is
isointense to gray matter, but shows some heterogene-
ity, probably due to necrosis. B. Axial T1-weighted image
reveals more heterogeneity in the anterior portion of the
solid tumor (arrows) suggesting more necrosis in that
region. C. Postcontrast axial T1-weighted image shows
enhancement of the solid portion of the tumor. Note that
the anterior part of the solid tumor (white arrows), which
has more necrosis, enhances more than the posterior por-
tion (black arrows).

Astroblastoma large cerebral tumors, most commonly headaches, vomiting, and sei-
Although the term astroblastoma dates back to the time of Bailey and zures. Histological grade varies greatly; prognosis is strongly correlated
Cushing, the current concept of this tumor derives from the work of with degree of malignancy (364,365).
Bonnin and Rubenstein, who described a tumor of children and young Imaging studies reveal large, well-circumscribed lobulated masses
adults that is generally solid or solid and cystic, well circumscribed, that are located in the periphery of the cerebral hemisphere. Typically,
and defined histologically by the presence of astroblastic pseudoro- both solid and cystic components are seen. The solid component is
settes and prominent perivascular hyalinization (362). It is classified typically hypointense on T1-weighted images, and heterogeneous on
as “other neuroepithelial tumor” in the last WHO classification (73). T2-weighted images, largely of gray matter intensity and contain-
Recognition of astroblastoma as a distinct entity is supported by the ing small, cystic-appearing heterogeneities. Relatively little vasogenic
work of Brat et al. (363), who found chromosomal alterations that are edema surrounds the mass, which appears sharply marginated by
not typical of other hemispheric tumors of children and young adults. imaging (but is not sharply marginated pathologically). Solid por-
Clinically, astroblastomas are tumors of children and young adults tions of the tumor enhance heterogeneously; ring enhancement is seen
(mean age of 14 years, range 3–46 years); a female predominance has around tumor cysts (364–366). Imaging features cannot differenti-
been noted in some studies (363). Presenting symptoms are typical of ate benign astroblastomas from malignant ones. No data concerning

Barkovich_Chap07.indd 709 5/6/2011 9:16:45 PM


710 Pediatric Neuroimaging

FIG. 7-70. Oligodendroglioma. A. Axial


noncontrast CT shows a heavily calci-
fied mass in the left temporal lobe with
a medial hypodense component (arrow).
B. Axial T1-weighted image shows the
mass to be heterogeneous, with a hypoin-
tense central component (large black
arrows) and a more hyperintense periph-
eral component (small black arrows) that
may be from the calcification. Artifact
is misregistration from ocular motion.
C. Axial postcontrast T1-weighted image
shows marked enhancement of part of
the tumor (small arrow). Less marked
enhancement is seen anteriorly (large
arrows) in the calcified tissue that, at
surgery, was also found to be tumor.
D. Coronal T2-weighted image shows
the tumor to be heterogeneous. (This
case courtesy of Dr. Ennio Del Giudice,
Napoli).

proton spectroscopy or perfusion imaging have been reported. Our Imaging Studies
experience with a single malignant astroblastoma revealed markedly The CT appearance of oligodendrogliomas in children is that of round
increased blood volume. to oval, sharply marginated masses that originate at the cortical-white
matter junction and are hypo- to isodense compared with gray mat-
Oligodendroglioma ter prior to contrast infusion. Contrast enhancement is variable,
Although they constitute 5% to 7% of brain tumors in the general but usually subtle. Calcification and cysts (Fig. 7-70) are present in
population, oligodendrogliomas are rare in children. They appear to approximately 25% to 50% (370). Because the lesions grow slowly, the
constitute approximately 1% of pediatric brain tumors; males are more inner table of the calvarium may be eroded when they are located in
commonly affected, with male: female ratios up to 2:1 (367). Affected the periphery of the brain (10). The MR appearance of oligodendro-
children most commonly present with seizures; other common symp- gliomas is completely nonspecific; they are most commonly sharply
toms include headache, visual field defects and weakness (367). In gen- defined and mildly heterogeneous, with T1 hypointensity and T2/
eral, oligodendrogliomas are slow-growing hemispheric lesions that FLAIR hyperintensity (370). Regions of calcification may be hyperin-
most commonly occur in the frontal and temporal lobes and are fre- tense (Fig. 7-70) or, if the calcification is dense enough, hypointense
quently calcified (75,76). Prognosis is related to the histological grade on T1-weighted images. Solid portions of the tumor typically enhance
of the tumor (367) and, particularly, to the tumor genotype; tumors moderately after administration of paramagnetic contrast (Fig. 7-70),
with deletions of chromosomes 1p36 and 19q13 have more favorable but enhancement may be minimal or completely absent. The pres-
outcomes and better response to antiangiogenic agents (368,369). ence of calcium and the absence of enhancement are associated with

Barkovich_Chap07.indd 710 5/6/2011 9:16:46 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 711

improved survival (370,371). A diagnosis of oligodendroglioma may sex or racial predilection. The most common presenting symptoms are
be raised when a tumor develops in the frontal lobe, and is associated headache, seizure, weakness, and cranial neuropathy; the precise pre-
with calcification, cysts, and limited or absent contrast enhancement; sentation is dependent upon the location of the granuloma (388–392).
these features separate the tumor from most astrocytomas. When
located in the temporal lobe, they are indistinguishable from ganglio- Pathology
gliomas. Anaplastic oligodendrogliomas show rim enhancement and Histologically, plasma cell granulomas are composed of myofibroblasts
central necrosis, an appearance indistinguishable from high-grade and chronic inflammatory cells (lymphocytes, eosinophils, histiocytes)
astrocytomas, ependymomas, and PNETs. with a predominance of plasma cells. When involving the CNS, they
When evaluating posttreatment oligodendrogliomas for recur- may most commonly arise in the dura, but may be located anywhere,
rence, it is important to remember that low-grade oligodendrogliomas including the cerebrum, cerebellum, hypothalamic-pituitary axis, and
have significantly elevated rCBV compared with low-grade astrocy- choroid plexuses (390–393). When intraparenchymal, they tend to be
tomas and that both low-grade and high-grade oligodendrogliomas peripherally located; however, no lobar predilection has been reported.
have similar blood volumes (372,373), consistent with the increased
microvessel density seen in both low- and high-grade oligodendroglial Imaging Studies
tumors (374). However, choline is significantly higher in high-grade On CT, plasma cell granulomas are hyperdense masses that are round
than in low-grade oligodendrogliomas and the presence of lactate or to ovoid and well-circumscribed. On MRI, they are sharply defined
lipid correlates with high tumor grade (374). Therefore, when evalu- masses that are hypointense on both T1- and T2-weighted images. They
ating oligodendroglial tumor recurrence, proton MRS is superior to show homogeneous enhancement after administration of intravenous
dynamic susceptibility-weighted perfusion imaging. contrast (390,393). Dural-based plasma cell granulomas may cause
hyperostosis or erosion of the overlying bone; thus, they may mimic
Neuroglial Hamartomas (Glioneuronal Heterotopia) meningiomas (388–390). When originating in the choroid plexus,
Cerebral hamartomas are defined as lesions composed of disorganized these signal characteristics may raise the possibility of a papilloma or,
but mature cells, usually a combination of neurons (ganglion cells), in an older child or adult, a meningioma or xanthogranuloma (393).
glia, and blood vessels. They are referred to by many names, includ- Diffusion/perfusion characteristics are not reported. The possibility of
ing neuroglial hamartomas (375), glioneuronal heterotopia (376,377), this diagnosis may be raised when a peripherally located parenchymal,
and heterotopic cerebellum (378). Affected patients typically present cavernous sinus, or choroid plexus mass exhibits low signal intensity
for medical evaluation because of seizures; neurological examination is on T2-weighted images and homogeneous contrast enhancement.
typically normal (379). They may be discovered in newborns as a result
of associated anomalies (378,380) or progressive head enlargement Lymphoproliferative Disorders
(377), and in fetuses as an incidental finding on sonography (377). In Lymphoproliferative disorders, including CNS lymphoma, can develop
pathology series, about 3% of patients who undergo epilepsy surgery in children (or adults) who have immunodeficiency syndromes, such
are discovered to have glioneuronal hamartomas; associated cortical as Wiskott-Aldrich syndrome, AIDS, and X-linked lymphoproliferative
dysplasia is often found (381–384). disease (394), as well as in those who have undergone solid organ or
Pathology of these lesions shows a mixture of mature and imma- stem cell transplantation and are immunosuppressed to prevent rejec-
ture neuronal and glial cells with variable representation from the cere- tion. As a result of the immunocompromise, lymphoid growth pro-
bellum, choroid plexus, and ependyma (375,376,380). The lesions may ceeds uncontrolled. Infection by Epstein-Barr virus is thought to play
be intracerebral or extracerebral, and sometimes extend outside of the a role, as Epstein-Barr seronegativity at the time of transplantation is a
cranium, typically through skull base foramina (375,376). They do not strong risk factor; other risk factors include concomitant cytomegalo-
invade adjacent brain. virus infection, the specific immunosuppressive regimen, the allograft
Imaging studies show lesions that are typically homogeneous or type, and young (pediatric) age (395–397). Lung, liver, and heart trans-
slightly heterogeneous. The signal intensity is isointense to gray matter plant recipients generally have higher risks than do recipients of renal
on T1-weighted images, and isointense to slightly hyperintense to gray transplants, possibly because of the different immunosuppressive regi-
matter on T2-weighted images, and variably hyperintense compared mens (398). Involvement of the CNS is less common than involvement
to gray matter on FLAIR images. The lesions are usually nonenhanc- of the thorax, abdomen, or head and neck (399). However, when the
ing, but minimal enhancement may be seen (377–379). Most com- CNS is involved, it is usually involved in isolation; therefore, a high
mon locations are the thalamus, mesial temporal lobe (Fig. 7-71A–C), index of suspicion is essential when CNS disease develops in the post
hypothalamus (Fig. 7-71D and E and see next section on “Suprasellar transplant setting. Affected patients may present with focal neurologic
Tumors”), and frontal lobe. In these locations, the major differential signs/symptoms, seizures, or signs of increased intracranial pressure
diagnosis is ganglioglioma in older patients and teratoma in neonates. (headaches, nausea, vomiting) (395–398).
Occasionally, glioneuronal heterotopia are seen in the interhemispheric Imaging typically shows multiple lesions surrounded by extensive
fissure, separating the two normal cerebral hemispheres. interstitial edema (398,399). As is typical with collections of lympho-
cytes, the lesions are isointense to slightly hyperintense compared to
Plasma Cell Granulomas (Inflammatory Pseudotumors) gray matter on T2-weighted images and slightly hyperintense to gray
Plasma cell granulomas (also called inflammatory pseudotumors or matter on FLAIR images. Ring-like enhancement is typically seen after
myofibroblastic inflammatory tumors) are unusual tumors that can administration of intravenous contrast (398). Diffusion is typically
present in nearly any organ at nearly any age. The lung is the most reduced compared with surrounding brain tissue. Differentiation of
frequent site of presentation, followed by the CNS, and the liver. Other benign lymphocytic proliferation from frank lymphoma or infection
organs, involved less frequently, are the skin, adrenals, thyroid, orbit, is usually not possible by imaging alone, so biopsy is typically per-
breast, spine, meninges, skull base, and skeletal muscle (385–389). formed. However, if reduced immunosuppression, antiviral therapies
Plasma cell granulomas of the CNS can present at any age, with equal (to control Epstein-Barr virus replication), or cytokines (which boost
distribution before and after the age of 20 years; thus, just under half the immune system and control viral load) result in diminution of the
of the cases involving the CNS present in children (390). There is no lesion load, biopsy may occasionally be avoided. The typical proton

Barkovich_Chap07.indd 711 5/6/2011 9:16:47 PM


712 Pediatric Neuroimaging

FIG. 7-71. Glioneuronal hamartomas A–C. Temporal lobe hamar-


toma. Coronal T1-weighted image (A) shows a gray matter attenuation
mass (arrows) in the region of the left amygdala. Coronal FLAIR (B) and
T2-weighted (C) images show that the mass remains isointense to gray
matter. In addition, the mass did not change during 3 years of follow-up
studies (arrows). D and E. Suprasellar hamartoma. Sagittal T1- (D) and
axial T2-weighted (E) images show a heterogeneous mass with signal of
both gray and (unmyelinated) white matter involving midbrain, both thal-
ami and hypothalamus. The mass was stable for more than 4 years.

spectroscopic characteristics of benign lymphoproliferation have not a significant number, estimated at 4% to 14%, arise in the basal ganglia
been described. and thalami, particularly in boys during the second decade of life (400–
405). Common symptoms and signs of germinomas in these regions
Germ Cell Tumors are progressive hemiparesis, fever of unknown origin, and visual dis-
Although most germ cell tumors are found in the suprasellar and turbances; some patients also have dementia, psychiatric signs, convul-
pineal regions (and will be discussed in those sections of this chapter), sions, precocious puberty, or diabetes insipidus.

Barkovich_Chap07.indd 712 5/6/2011 9:16:47 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 713

Pathology differentiated from astrocytomas by their high signal intensity relative


Germ cell tumors include both germinomatous and nongerminomatous to white matter prior to contrast infusion (10,401,408–410).
types; the latter include teratoma (mature teratoma, immature tera- On MRI, germinomas usually appear as heterogeneous, poorly-
toma, teratoma with malignant transformation), choriocarcinoma, marginated tumors, either round or lobulated, with relatively little
embryonal carcinoma, yolk sac tumors, and mixtures of these entities mass effect (411). The tumors are usually isointense to hyperintense
(406). The histologic features of germinomas are characteristic, with compared to cortical gray matter on T1-weighted, T2-weighted, and
two distinct cell populations. Areas of large polygonal or spheroidal FLAIR images. They typically, but not always, demonstrate reduced
cells with a frequently vacuolated cytoplasm and large round nuclei diffusion diffusivity (ADC) maps (412). As a result of the iron depo-
with prominent nucleoli are separated by vascular trabeculae along sition that occurs in the globi pallidi during adolescence and early
which are clustered small round cells resembling lymphocytes (76). adulthood, small tumors may appear hyperintense compared to the
Some tumor cells may have an abnormal karyotype (407). basal ganglia on T2-weighted images. Indeed, the findings of small
basal ganglia germinomas are subtle on MRI, showing only subtle T1
Imaging Studies shortening or T2 prolongation without significant enhancement (405).
The CT appearance of a germinoma is that of an isodense to hyperdense, Larger tumors demonstrate prolonged T1 relaxation times and isoin-
well-marginated mass. When the tumors are large, low-attenuation, tensity to hyperintensity compared with gray matter on T2-weighted
cystic-appearing regions of necrosis will often be present. Uniform images. Heterogeneity is common in larger germinomas of the basal
enhancement of the solid portions of the tumor is seen after intra- ganglia and thalami (Fig. 7-72); some degree of necrosis is present in
venous infusion of contrast. Germinomas of the basal ganglia can be 80% (50% are slightly necrotic, 30% are mostly necrotic) at the time of

FIG. 7-72. Germinoma of the basal gan-


glia. A. Axial T1-weighted image shows
multiple cysts (arrows) in the right basal
ganglia. B. Axial T2-weighted image at a
slightly higher level shows a solid com-
ponent (arrows) that is isointense to gray
matter, suggesting the diagnosis of small
cell tumor. C. Axial FLAIR image shows
the solid portion (arrows) remaining
isointense to gray matter. D. Postcon-
trast axial T1-weighted image shows that
the solid portion of the tumor (arrows)
enhances nearly uniformly.

Barkovich_Chap07.indd 713 5/6/2011 9:16:49 PM


714 Pediatric Neuroimaging

initial imaging (403,404,410,413). The heterogeneity is most apparent tions of CSF) may be present. No significant mass effect is noted. MR
on T2-weighted and contrast-enhanced images (Fig. 7-72). Germino- scans show heterogeneous masses that are isointense to hypointense
mas of the deep gray matter tend to be large at the time of original compared with gray matter on T1-weighted images and hypointense
imaging (403,404,413). The solid portions of germinomas strongly with a hyperintense periphery on T2-weighted images (Fig. 7-73B)
enhance after infusion of paramagnetic contrast (Fig. 7-72), as do (417,419–421). Eight of nine reported cases of postcontrast MR scans
CSF-borne metastases to the brain and spine. At the time of presenta- showed heterogeneous enhancement (416,419,421). One case pre-
tion, hemiatrophy is often present in the ipsilateral hemisphere and sented with extensive cerebral and intraventricular hemorrhage (421).
brainstem (410,414). When a relatively shortened T2 relaxation time is
present in a basal ganglia mass, particularly in association with cysts or
ipsilateral hemiatrophy, germinoma should be considered, along with Sellar and Suprasellar Tumors
lymphoma and PNET. A Systematic Approach to Tumors of the
Meningioangiomatosis Hypothalamus and Pituitary
Meningioangiomatosis is a disorder that involves the cerebral cortex Suprasellar masses in children can be separated with some degree of
and, often, the overlying leptomeninges (75). The association of this certainty based upon their imaging characteristics (Table 7-8). How-
rare, benign, hamartomatous lesions may be seen in association with ever, pediatric suprasellar masses can be differentiated to some degree
Type II neurofibromatosis (415) is controversial. It is usually sporadic before even looking at the neuroimaging study. It is important to rec-
(416). Patients typically present as children or young adults with sei- ognize that some clinical signs and symptoms indicate hypothalamic
zures, headaches, or dizziness (417), but the majority of cases are found involvement, whereas others indicate pituitary involvement. Knowl-
incidentally (418). Meningioangiomatosis found in neurofibromatosis edge of the characteristic modes of clinical presentation is important
is usually an incidental finding, does not typically cause seizures, and is if the region is to be optimally imaged. Moreover, many tumor types
often multifocal (416). Neurologic exam is typically normal (416). The present with specific clinical signs and symptoms. The most common
cause is unknown. presenting syndromes of hypothalamic and pituitary lesions and the
lesions that most commonly cause them are listed in Table 7-9. The
Pathology optimal imaging sequence for these tumors is thin section (3 mm or
The characteristic findings are cortical meningovascular fibroblastic less) precontrast T1-weighted and T2-weighted images in the sagit-
proliferation and leptomeningeal calcification (418). Histologically, tal and coronal planes, followed by postcontrast T1-weighted sagit-
the disorder is characterized by transcortical fascicles of vascular fibro- tal and coronal images. Postcontrast axial images through the entire
meninges that contain central capillaries. Adjacent cortex may contain brain should be obtained for suprasellar masses, which often spread
neurofibrillary tangles or hyalinization (418). through the CSF pathways. We have not found FLAIR sequences useful
for tumors in this region.
Imaging Studies
On CT, meningioangiomatosis appears as a peripheral mass that Normal Size of the Infundibulum (Pituitary Stalk)
is hyperdense prior to contrast administration and elicits mild to When patients have central diabetes insipidus, imaging of the hypo-
moderate edema or hypomyelination in the adjacent white matter thalamic/pituitary region is often ordered to exclude the presence
(Fig. 7-73A) (417,419). Calcifications and cysts (perhaps locula- of a mass in the hypothalamus or the infundibulum. It can often be

FIG. 7-73. Meningioangiomatosis.


A. Axial noncontrast CT scan shows a
region of hypodensity (arrows) in the
left temporoparietal region. B. Axial SE
2000/80 image shows a heterogeneous
mass with central hypointensity and
surrounding hyperintensity.

Barkovich_Chap07.indd 714 5/6/2011 9:16:50 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 715

primary site of origin cannot be determined in many cases, tumors aris-


TABLE 7-8 Suprasellar Tumors in ing from these two locations are often discussed together. Astrocytomas
of the optic chiasm and hypothalamus make up 10% to 15% of
Children supratentorial tumors in children. Males and females are approxi-
mately equally affected. The usual age at presentation is 2 to 4 years; in
Tumor Key Characteristics contrast, patients with gliomas that involve only the optic nerve typi-
Craniopharyngioma Heterogeneous solid portion cally present slightly later (6 years) (425,426). Tumors restricted to the
Rim-enhancing cysts of variable optic nerve in children also have a different prognosis and are treated
intensities differently; therefore, they should probably be considered an entity dis-
tinct from the hypothalamic/chiasmal tumors (425,426). Nonetheless,
Astrocytoma T2 hyperintensity of solid portions considering the recent studies (427–430) demonstrating spontaneous
Germ cell tumors T2 isointensity to gray matter regression of tumors of the optic nerve and chiasm, both large and
Diabetes insipidus small, both in and out of the setting of NF1, childhood tumors of the
optic nerves and chiasm should, perhaps, be treated conservatively
Langerhans cell histiocytosis Enlarged, enhancing stalk
(428,430). Optic nerve tumors are discussed more completely in Chap-
Look for osseous lesions
ter 6, within the discussion of NF1, because about 15% of children with
Hypothalamic hamartoma Isointense to gray matter on T1, T2 NF1 develop optic system gliomas (431); a brief discussion is included
No enhancement. Sometimes cysts. in this section for convenience.
Arachnoid cyst Isointense to CSF The most common symptom of hypothalamic/optic pathway
No enhancement gliomas is diminished visual acuity, which is noted in almost 50% of
Suprasellar pituitary Intrapituitary mass
adenoma Expansion of sella turcica
Rathke cleft cyst Intrasellar, smooth walled TABLE 7-9 Common Clinical
Variable intensity. No enhancement. Presentations of Hypothalamic
Lymphocytic hypophysitis Identical to germ cell tumor or and Pituitary Lesions
(rare) Langerhans cell histiocytosis
Granuloma (sarcoidosis/TB) Involved infundibulum Hypopituitarism
Hypointense on T2 images Craniopharyngioma
Kallmann syndrome (see Chapter 5)
Septo-optic dysplasia (see Chapter 5)
Pituitary hypoplasia (see Chapter 5)
Idiopathic
difficult to determine the presence of a mass, as the findings at the time
of initial presentation may be absent or very subtle. A few facts can Diabetes insipidus
help in making the proper diagnosis. Most important is to remember Langerhans cell histiocytosis (histiocytosis X)
that diabetes insipidus occurs due to dysfunction of supraoptic or Germ cell tumors
paraventricular nuclei of the hypothalamus. Infiltration of these nuclei Craniopharyngiomas
can occur at a time when the infundibulum is still normal in size. As Lymphocytic hypophysitis
the infundibulum lacks a blood–brain barrier and, therefore, normally Tuberculosis (see Chapter 11)
enhances after intravenous administration of paramagnetic contrast, Sarcoidosis
one might find no imaging abnormality at the time of the initial MR Hypothalamic/pituitary malformations
study. However, a normal scan does not necessarily imply absence of Precocious puberty (boys before age 9 y, girls before age 8 y)
tumor (422)! It is very important to reimage the patient at regular (3–6 Hamartoma of the tuber cinereum
month intervals) so that the cause of the hypothalamic abnormality, Hypothalamic glioma
be it tumor, granuloma, or lymphocytic infiltration, can be detected Choriocarcinoma
and treated soon. It is also very helpful to know that the infundibu- Teratoma
lum should be no more than 2.7 mm at the pituitary insertion and Increased intracranial pressure
no more than 3.8 mm at the level of the optic chiasm on coronal and Hydrocephalus
sagittal MR images (423); greater thickness is highly suspicious for Head trauma
pathologic infiltration (423,424). Finally, the infundibulum should Hypoxic-ischemic brain injury
taper smoothly from it largest portion at the median eminence of the
tuber cinereum to its smallest portion as the insertion into the neuro- Delayed puberty (boys after age 14 y, girls after age 13 y)
hypophysis. Any abrupt changes in size should raise suspicion for an Hypothalamic astrocytoma
infundibular mass. Langerhans cell histiocytosis
Kallmann syndrome (see Chapter 5)
Chiasmatic/Hypothalamic Astrocytomas Pituitary adenoma
and Optic Nerve Tumors Amenorrhea
Gliomas originating in the optic chiasm often enlarge and involve the Pituitary adenoma
hypothalamus; similarly, astrocytomas arising in the hypothalamus Rathke cleft cyst
often grow anteriorly or inferiorly to involve the chiasm. Because the

Barkovich_Chap07.indd 715 5/6/2011 9:16:51 PM


716 Pediatric Neuroimaging

FIG. 7-74. Optic nerve glioma. Axial CT using bone algorithm


shows the large left optic nerve tumor (open black arrows) and an
expanded left optic canal (closed black arrows).

patients, and is associated most strongly with involvement of post- Imaging Studies
chiasmal portions of the optic pathways (432). The most common The ready availability of fat suppression techniques on modern MR
sign is optic atrophy. Endocrine dysfunction, most commonly short scanners (441) has made MRI the procedure of choice for evaluating
stature secondary to reduced growth hormone, is found in about tumors of the optic nerves and chiasm. CT displays the intraorbital
20% (433). Large tumors produce hydrocephalus secondary to exten- portion of the nerve, contrasted by orbital fat, very nicely; however, the
sion into the anterior third ventricle and obstruction at the level of intracranial portions of the optic nerves and the chiasm are not as well
the foramina of Monro. These large tumors are seen primarily in seen by CT as by MRI unless subarachnoid contrast is administered.
infants who may present initially with macrocephaly. A combination When fat suppression techniques are used, MRI shows all portions of
of emaciation, pallor, alertness, and hyperactivity (the diencephalic the optic nerves, the optic chiasm, the optic tracts, the hypothalamus,
syndrome) is found up to 20% of affected patients less than 3 years and the third ventricle. In addition, MRI allows the structures to be
of age (8,76,434). viewed in any plane; the coronal and sagittal planes are especially use-
ful for evaluating tumors in the sellar and suprasellar regions. Finally,
Pathology because of the possibility of radiation effects on the growing orbits and
Depending upon the series, 20% to 50% of patients with astrocy- brain (see discussion in Chapter 1) CT of the orbits should not be the
tomas of the optic chiasm/hypothalamus have clinical evidence or first imaging method chosen if others are available.
a positive family history of NF1. NF1 patients may develop tumors If MRI is not available or cannot be used, CT remains adequate
primarily involving the optic nerves or the optic chiasm/hypothala- for evaluation of the intraorbital portions of the optic nerves. The
mus. When the optic nerve is primarily involved, the affected nerve enlargement of the optic nerve by tumor can be smooth and fusiform
expands in a fusiform fashion. A large majority of tumors originating but is more commonly lobulated. The use of bone windows often
within the optic nerve grow extremely slowly and are histologically reveals enlargement of the optic canals (Fig. 7-74) as a result of pres-
classified as JPAs, histologically identical to pilocytic astrocytomas sure erosion from the enlarged nerves. When the mass involves the
that occur in other regions of the brain; malignant change is intracranial optic nerves, optic chiasm, optic tracts, or hypothalamus,
extremely rare in these tumors (425,426,435). Indeed, optic pathway it almost always appears as a low density, lobulated suprasellar mass
tumors may involute spontaneously, both within (429) and outside prior to the administration of IV contrast. The tumors that grow
the setting of NF1 (428,436,437). This regression may relate to the along the optic nerves in patients with NF1 have a unique appear-
finding that the growth potential of pilocytic astrocytomas decreases ance in that the nerves have a characteristic downward kinking a few
progressively with patient age (438). However, tumors originating millimeters behind the globe, possibly as a result of nerve elonga-
from the optic chiasm and the hypothalamus may have a higher his- tion (442). Hemorrhage, calcification, and cyst formation are very
tologic grade and be frankly invasive (8,425,426,439). Astrocytomas unusual in tumors of the optic nerves prior to radiation therapy.
originating in the optic chiasm and hypothalamus may also spread After intravenous contrast administration, the mass enhances het-
throughout the subarachnoid spaces via the CSF, even if the tumor erogeneously and to a variable degree. When the tumor is very large,
is of low histologic grade (280,281). Prognosis may be worse if the the enhancement can sometimes be seen extending posteriorly from
tumor is multicentric at presentation (280,281). These more aggres- the mass along the optic tracts to the region of the lateral geniculate
sive tumors (WHO grade II) have a slightly different histological bodies (443,444).
appearance, tend to affect younger children and to occur more com- MRI is optimal for showing the relationship of the mass to the
monly in the hypothalamic/chiasmatic region; they are designated chiasm, hypothalamus, and infundibulum. Moreover, when fat sup-
pilomyxoid astrocytoma (PMA) (briefly described in the section on pression and paramagnetic contrast agents are used, the intraorbital
“Cerebellar Astrocytomas” earlier in this chapter) (440). and intracanalicular portions of the nerve are demonstrated well

Barkovich_Chap07.indd 716 5/6/2011 9:16:51 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 717

FIG. 7-75. Optic nerve glioma restricted to orbit. A. Coronal


T1-weighted image shows the markedly enlarged right optic nerve
(arrows). B. Postcontrast fat-suppressed coronal T1-weighted image
shows marked enhancement of the mass. Fat saturation is essential
in performance of contrast-enhanced studies of the orbits or head
and neck. C. Sagittal T1-weighted image shows rope-like enlarge-
ment (arrows) of the intraorbital optic nerve.

(Figs. 7-75 and 7-76). MRI also helps to differentiate tumors in which optic tracts (Fig. 7-77C). The anterior cerebral artery, stretched over
the optic nerve itself is infiltrated by tumor from those in which the the top of the mass, often creates a small indentation in the superior
tumor predominantly infiltrates the subarachnoid space, leaving surface of the tumor mass (Figs. 7-78 and 7-79). High signal inten-
the nerve itself relatively unaffected (435). When changes are largely sity extending from the optic chiasm to the lateral geniculate bodies
restricted to tubular enlargement of the optic nerves and chiasm, the on T2-weighted images may represent extension of tumor or edema
tumor typically appears homogeneous precontrast and enhancement within the optic tracts (442,446). PMAs are very similar on imaging to
is variable after intravenous administration of paramagnetic contrast pilocytic ones. It appears that they are more often hemorrhagic (25%)
(Fig. 7-76). and more commonly disseminate via the CSF (15%) (147,440,447).
Astrocytomas of the optic chiasm and hypothalamus are almost Some studies have shown some differences in optic pathway gliomas
always hypointense on T1-weighted sequences and hyperintense on in patients with NF1 as compared to patients without NF1. In patients
T2-weighted and FLAIR sequences (Figs. 7-77 to 7-79). When large with NF1, the orbital portion of the optic nerve is the most commonly
and bulky, tumors of the chiasm and hypothalamus are typically het- involved site, followed by the chiasm; extension beyond the optic path-
erogeneous, with large cystic and solid components; the solid portions way is rare (2%). In contrast, optic pathway gliomas in patients without
typically enhance markedly after contrast administration (Fig. 7-79) NF1 affect the chiasm (91%) much more commonly than the orbital
(445). Although sagittal images best demonstrate the enlarged optic nerve (32%); more than 2/3 extend beyond the optic pathway into
chiasm and hypothalamus, the coronal plane is optimal for visualiz- brain parenchyma. Cystic components are uncommon in NF1 (<10%)
ing tumor invasion of the cerebral hemispheres and identification of but common (66%) in patients without NF1. Progressive enlargement
tumor in the optic canals, intracranial optic nerves, optic chiasm, and is much less common in NF1 (50% vs. 95% outside of NF1) (448).

Barkovich_Chap07.indd 717 5/6/2011 9:16:51 PM


718 Pediatric Neuroimaging

FIG. 7-76. Bilateral optic gliomas with intracranial extension to optic chiasm. A–D. Postcontrast axial T1-weighted images show bilateral enlarged intraor-
bital optic nerves (small black arrows) extending through the optic canals (closed white arrows) and to the chiasm (open white arrows). Minimal enhancement
of the nerves and chiasm is present.

Barkovich_Chap07.indd 718 5/6/2011 9:16:53 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 719

FIG. 7-77. Pilocytic astrocytoma involv-


ing most of optic pathways and extending
into basal ganglia. A. Sagittal T1-weighted
image shows a large mass (arrows) origi-
nating in the region of the optic chiasm/
hypothalamus. The mass erodes the planum
sphenoidale (arrowheads). B. Postcontrast
axial T1-weighted image shows the mass
extending anteriorly towards the optic nerves
(arrowheads) and posteriorly towards the
optic tracts (arrows). C. Postcontrast fat-sup-
pressed coronal T1-weighted image shows
uniform enhancement of the enlarged optic
nerves (arrows). D. Axial T2-weighted image
shows the hyperintense tumor (arrows)
extending from the optic radiations into the
temporal lobes.

FIG. 7-78. Homogeneous chiasmatic/hypothalamic glioma. A. Sagittal T1-weighted image shows a lobulated suprasellar mass that erodes the posterior
planum sphenoidale and tuberculum sella (open arrows). Indentation of the superior surface of the mass (closed arrow) by the anterior cerebral artery is
characteristic. B. After administration of paramagnetic contrast, the tumor enhances uniformly.

Barkovich_Chap07.indd 719 5/6/2011 9:16:54 PM


720 Pediatric Neuroimaging

FIG. 7-78. (Continued) C. Axial T2-weighted image shows the tumor to be hyperintense.

FIG. 7-79. Heterogeneous large chias-


matic/hypothalamic PMA. A. Sagittal
T1-weighted image shows a large mass
involving the suprasellar region, the
anterior two thirds of the third ventricle,
anteroinferior interhemispheric fissure
(closed arrow) and the prepontine cistern
(open arrow). Significant hydrocephalus
results from obstruction of the foram-
ina of Monro. The optic chiasm cannot
be discerned as a separate structure. B.
After infusion of paramagnetic contrast,
heterogeneous enhancement is observed.
The superior portion of the tumor
(arrows) is a large cyst. C. Axial postcon-
trast T1-weighted image shows hetero-
geneous enhancement of the mass. The
dilated anterior interhemispheric fissure
(arrows) was the result of a cyst with non-
enhancing walls extending anteriorly from
the tumor. D. Axial T2-weighted image
shows the tumor extending into the inter-
peduncular cistern (arrows) and splaying
the cerebral peduncles. The tumor has a
prolonged T2-relaxation time.

Barkovich_Chap07.indd 720 5/6/2011 9:16:56 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 721

Proton MR spectroscopy of suprasellar astrocytomas shows cysts, when present, are small and contain straw-colored fluid (457,458).
increased choline and diminished N-acetyl aspartate. This finding may Because the tumor commonly originates in the pituitary stalk, both the
be useful in the differentiation of astrocytomas from craniopharyn- dorsum and tuberculum sellae are often eroded. It has been postulated
giomas, which show no normal metabolites (only a lipid peak at 1–2 that lobulated craniopharyngiomas with large, hyperintense cysts on
ppm), and from pituitary adenomas, which show only a choline peak T1-weighted MR scans are adamantinomatous, whereas the smaller,
or no metabolites at all (449). round, primarily solid craniopharyngiomas with hypointense cysts on
T1-weighted images are of papillary histology (456).
Differential Diagnosis Surgeons divide craniopharyngiomas into three groups based on the
The differential diagnosis of chiasmal/hypothalamic astrocytomas consequences of their location upon surgical approach: sellar, prechias-
includes craniopharyngiomas, germ cell tumors, hypothalamic gan- matic suprasellar, and retrochiasmatic suprasellar (453,454). Sellar cran-
gliogliomas (316), and granulomatous diseases such as tuberculosis iopharyngiomas are usually small; they may involve the pituitary gland
and sarcoidosis. Presentation before age 5 years favors the diagnosis of but usually do not distort the optic nerves, chiasm, or anterior cerebral
astrocytoma, as does association with cutaneous or radiologic stigmata arteries. Prechiasmatic suprasellar craniopharyngiomas grow anteriorly
of NF1. In addition, clinical history can be helpful. Diabetes insipidus between the optic nerves, displacing the chiasm posteriorly and elevat-
is usually present at the time of presentation of germ cell tumors and ing the anterior cerebral arteries; they rarely compress the third ventricle.
granulomatous diseases; moreover, these masses typically show isoin- Retrochiasmatic suprasellar craniopharyngiomas can push the chiasm
tensity to gray matter on T2-weighted images and reduced diffusiv- forward against the tuberculum sellae, fill the third ventricle, and extend
ity compared with normal brain, in contrast to the hyperintensity and into the interpeduncular and prepontine cisterns. Rarely, craniophar-
relatively high diffusivity seen in astrocytomas. Pediatric craniophar- yngiomas may develop as clival, nasopharyngeal, or sphenoidal masses
yngiomas can be differentiated from astrocytomas by their primarily (460,461). Recent work has suggested a possible role of an immune
cystic appearance, by the higher signal intensity of the cysts, and by response in craniopharyngioma cyst formation (462); if further investi-
the more granular, heterogeneous appearance of their solid component gations support this, nonsurgical treatment may improve outcomes.
(see the following section).
Imaging Studies
Craniopharyngioma The CT appearance of pediatric (adamantinomatous) craniophar-
Craniopharyngiomas are tumors that are postulated to arise from rem- yngiomas is characteristic (10,463). They are most frequently supra-
nants of the craniopharyngeal duct (450,451). They may arise anywhere sellar in location. Ninety percent will have a cystic component and
along the pituitary stalk from the floor of the third ventricle to the 90% will be at least partially calcified. The calcification may be a thin,
pituitary gland; rarely, they arise below the sella in the sphenoid sinus, circumferential rim (usually around the cyst), or chunks of calcium
presumably along the path of the craniopharyngeal canal (451,452). within the solid portion of the tumor. Intravenous administration of
Craniopharyngiomas account for 3% of all intracranial tumors, 15% iodinated contrast results in enhancement of 90% of craniopharyn-
of supratentorial tumors, and 50% of suprasellar tumors in children. giomas. Therefore, a cystic, contrast-enhancing suprasellar lesion with
Although the incidence of craniopharyngiomas peaks between 10 and calcification is almost certainly a craniopharyngioma (Fig. 7-80). If any
14 years of age, a second peak occurs in the fourth to the sixth decade
of life. Males are more commonly affected than females. Symptoms vary
with the location of the tumor, size of the tumor, and the age of the
patient. Symptoms typically consist of headaches and visual field defects
secondary to compression of the optic chiasm and tracts. In addition,
anterior pituitary dysfunction (growth failure resulting from compres-
sion of the anterior pituitary and the hypothalamic-pituitary portal
system) and hypothalamic dysfunction (usually manifest as diabetes
insipidus) are often noted (8,445,453,454). Hypothalamic dysfunction
at the time of diagnosis, severe hydrocephalus, and age less than 5 years
at presentation are associated with high hypothalamic morbidity (455).

Pathology
Craniopharyngiomas are divided into two histologic types, adaman-
tinomatous and papillary types; some have mixed histology. Ada-
mantinomatous craniopharyngiomas generally occur in children and
adolescents. They are lobulated masses with multiple (often very large)
cysts that often encase the vessels of the circle of Willis (456). Adaman-
tinomatous tumors commonly calcify and are more likely to involve the
hypothalamus (457,458); indeed, most appear to arise in the floor of the
third ventricle (459). They vary greatly in size; small tumors present as
solid or partly cystic nodules in the tuber cinereum, infundibulum, or
sella, whereas large tumors are primarily cystic, often extending upward
into the third ventricle or posteriorly and inferiorly into the interpe-
FIG. 7-80. CT of craniopharyngioma. Axial contrast-enhanced CT scan
duncular and prepontine cisterns to the level of the pons. The cyst fluid
shows a low attenuation suprasellar mass with multiple areas of calcifica-
may be either straw-colored or a brownish, motor-oil-like substance tion (arrows). Large chunks of calcified tumor are seen in the posterior
that is rich in cholesterol crystals (8,76,445). Papillary craniopharyn- aspect of the tumor but only thin curvilinear calcification is seen in the cyst
giomas generally present in adults and will be discussed only briefly. walls. In addition, the cyst walls show a rim of enhancement around the low
They are predominantly solid and seem to originate mostly in the sella; attenuation cystic center (c).

Barkovich_Chap07.indd 721 5/6/2011 9:16:57 PM


722 Pediatric Neuroimaging

FIG. 7-81. Small craniopharyngioma. A. Axial noncontrast


CT shows a calcified suprasellar mass (arrows). B. Sagit-
tal T1-weighted image shows a loculated multicystic mass
(arrows) in the suprasellar region. C. Coronal FLAIR image
does not show the lesion well. The solid portion of the tumor is
difficult to differentiate from the hyperintense cyst and neither
is well differentiated from the hyperintense edema in the sur-
rounding tissue. D. Axial T2-weighted image shows the solid
component of the cyst (white arrow) as hypointense and an
associated cyst (white arrowhead) as hyperintense compared to
surrounding CSF. E. Postcontrast fat-suppressed T1-weighted
image shows a rim of enhancement (small arrows) around
an inferiorly located cyst and heterogeneous enhancement
(arrowheads) in the more superior portion of the tumor.

two of these components are present, craniopharyngioma is a likely of initial presentation, and the solid portion is generally isodense with
diagnosis. (Epi)dermoid tumors can have the CT density of a cyst brain, small tumors are sometimes difficult to identify on CT.
with foci of calcification but (epi)dermoids, as stated earlier, do not Pediatric craniopharyngiomas are also characteristic on MR stud-
enhance with IV contrast. Suprasellar arachnoid cysts can be differen- ies, typically appearing as multilobulated, multicystic suprasellar masses
tiated from craniopharyngiomas by their absence of calcification and (Figs. 7-81 to 7-84). The cystic areas may be isointense or may have
enhancement. Because craniopharyngiomas are often small at the time high or low signal intensity with respect to brain tissue on T1-weighted

Barkovich_Chap07.indd 722 5/6/2011 9:16:58 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 723

FIG. 7-82. Retrochiasmatic multiloculated craniopharyngioma with extension into interpeduncular cistern. A. Sagittal T1-weighted image shows a
suprasellar mass with multiple areas of hyperintensity extending posteriorly into the interpeduncular cistern and distorting the midbrain. B and C. Sagit-
tal and axial postcontrast T1-weighted images show heterogeneous solid portion of tumor (black arrows in B) immediately superior to the pituitary gland
enhances heterogeneously. A thin rim of enhancement (white arrows in B and C) is seen around the large cysts. Hydrocephalus is present secondary to
obstruction of the foramina of Monro by the tumor. D. Axial T2-weighted image shows the hyperintense cysts (arrows) extending into the interpedun-
cular cistern and splaying the cerebral peduncles. The irregular margin of the tumor with the cerebral peduncles does not necessarily imply invasion by
the tumor.

sequences; the T1 hyperintensity, when present, is the result of very high prepontine cisterns and from edema in surrounding tissue on 2DFLAIR
protein content (464). Although both the cystic and solid components sequences. The solid portions of the tumor have a granular appearance
tend to have a high signal intensity on T2-weighted sequences, the cys- on precontrast T1-weighted images; they may also show heterogeneity
tic areas tend to have higher signal intensity than the solid components as a result of small cysts and calcifications. The solid portions enhance
(Figs. 7-81 and 7-83) (445,465). In contrast, cystic components and heterogeneously after administration of paramagnetic contrast
solid components are difficult to distinguish from flowing CSF in the (Figs. 7-82 and 7-84); the thin walls of the cysts nearly always enhance

Barkovich_Chap07.indd 723 5/6/2011 9:16:59 PM


724 Pediatric Neuroimaging

FIG. 7-83. Multicystic prechiasmatic craniopharyngioma. A. Sagittal


T1-weighted image shows a mass (straight arrows) expanding the sella
and compressing the pituitary gland (curved arrow). The floor of the
third ventricle and optic chiasm are displaced posteriorly and superiorly.
B and C. Postcontrast coronal T1-weighted images show the smaller cyst
(open arrows) to be hyperintense compared to the larger cyst. Both cysts
show thin rims of enhancement. The solid portions of the tumor (closed
arrows) show greater enhancement. D and E. Axial FLAIR image shows
differing intensity of the cysts and lower intensity of the solid tumor por-
tions (arrows).

(Figs. 7-82 and 7-83) (445). Occasionally, craniopharyngiomas are appearance and the inherent T1 shortening of the cysts, if present,
entirely solid (Fig. 7-85); the solid tumors are usually of papillary his- should aid in this diagnosis.
tology and are found almost exclusively in adults. They have a hetero- Proton MR spectroscopy of pediatric craniopharyngiomas differs
geneous appearance and enhancement characteristics that are identical from that of other suprasellar tumors (449). The spectra show no nor-
to the solid portion of cystic craniopharyngiomas. mal metabolites, only a broad lipid spectrum at 1–2 ppm. This should
It is important to realize that craniopharyngiomas extend into allow differentiation from suprasellar astrocytomas, which show a large
the middle cranial fossa, anterior cranial fossa, or posterior fossa choline peak and reduced NAA peak (449).
(prepontine cistern) in about 25% of affected patients (giant cran-
iopharyngiomas, Fig. 7-84) (466,467). This pattern of tumor exten- Differential Diagnosis
sion should therefore suggest a diagnosis of craniopharyngioma; Rathke cleft cysts and hemorrhagic pituitary adenomas are the only
thus MRI is particularly important in those patients presenting with other lesions that originate in this area and have high signal intensity
symptoms referable to the posterior fossa in order to point to the on T1-weighted sequences. Since Rathke cleft cysts tend to be small,
proper diagnosis and proper site of tumor origin. Very rarely, cran- intrasellar lesions or small lesions situated immediately anterior to the
iopharyngiomas can arise primarily in the third ventricle or in the insertion of the infundibular insertion into the pituitary gland, and
optic chiasm (468,469). The characteristic lobulated, multicystic they are approached transsphenoidally, the differentiation between

Barkovich_Chap07.indd 724 5/6/2011 9:17:01 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 725

FIG. 7-84. Giant cystic craniopharyngioma. A. Sagittal T1-weighted image shows very large hyperintense lobulated suprasellar mass. The sella turcica is
enlarged beyond recognition. B. Axial T2-weighted image shows the lobulated cyst to be hyperintense compared to CSF. A fluid-fluid layer (arrows) is pres-
ent. C. Coronal T1-weighted image shows smaller, cysts (arrows) that are hypointense compared with brain. The left temporal fossa is enlarged by the cyst.
D. After administration of paramagnetic contrast, all of the cysts show rim enhancement.

an intrasellar craniopharyngioma and a Rathke cleft cyst is entirely T1-weighted images, differentiation from suprasellar astrocytoma may
academic. Hemorrhagic adenomas can generally be differentiated by be difficult. The presence of calcification, the granular and heteroge-
the marked hypointensity (from deoxy- or methemoglobin) of the neous appearance of the solid portion of the tumor, enhancement of
hemorrhage on T2-weighted spin-echo or susceptibility-weighted cyst walls, and the enlargement of the sella turcica by extension of the
sequences. When the cysts of a craniopharyngioma are dark on tumor into the sella all suggest the diagnosis of craniopharyngioma.

Barkovich_Chap07.indd 725 5/6/2011 9:17:02 PM


726 Pediatric Neuroimaging

FIG. 7-85. Papillary craniopharyngioma in an adult. A. Axial


contrast-enhanced CT scan shows an enhancing mass in the supra-
sellar region (black arrows) situated between the two posterior cli-
noid processes (white arrows). B. Sagittal T1-weighted image shows
a heterogeneous mass within the sella and extending upward into
the suprasellar region (white arrows). The optic chiasm (open white
arrow) is slightly elevated by the mass. C. Coronal T1-weighted image
shows the mass sitting within the sella turcica and causing a slight
upward displacement of the optic chiasm (open arrows).

Hypothalamic Hamartomas (Hamartomas of precocious puberty and epilepsy appear to be more common when
the Tuber Cinereum) tumor diameter is more than 10 mm (472,474,475). Some authors
Hypothalamic hamartomas are rare congenital malformations that have postulated that precocious puberty is mostly seen in patients
are composed of normal neuronal tissue and are located in the tuber with pedunculated hamartomas suspended from the tuber cinereum,
cinereum (the region between the mamillary bodies and the median whereas epilepsy is seen with intrahypothalamic hamartomas (476).
eminence of the hypothalamus). Boys are affected more commonly Indeed, gelastic seizures may be present in the presence of smaller intra-
than girls. The most common presenting symptom is isosexual pre- hypothalamic hamartomas and in the absence of precocious puberty
cocious puberty; indeed, hypothalamic hamartomas are the most (477). Other symptoms associated with hypothalamic hamartomas
common cause of central precocious puberty (470). The precocious include neurodevelopmental delay, and hyperactivity. The hands and
puberty usually manifests itself prior to the age of two years in boys feet should be examined to look for postaxial polydactyly, which sug-
and slightly later in girls (471); the diagnosis, however, is frequently gests the diagnosis of Pallister-Hall syndrome, an autosomal dominant
not made until one or two years later when the MRI is performed. The disorder related to mutations of the GLI3 gene on chromosome 7p13
other common presenting symptom is epilepsy, typically of the gelastic (478,479). Affected patients are commonly large for their chronologi-
type (“laughing seizures”), but patients may initially manifest partial cal age (480,481). Although therapy is primarily medical, some authors
complex seizures or even infantile spasms, with the result that the hypo- propose that some patients with medically refractory seizures may be
thalamic region is not adequately examined. Ultimately, most patients cured by resection of the tumor (482). Indeed, ictal SPECT studies and
manifest many types of seizures, including atypical absences, gener- ictal depth electrode recordings indicate that the gelastic seizures origi-
alized tonic-clonic, partial motor, and drop attacks (472,473). Both nate from the hamartoma (473,477,483,484). Moreover, there seems to

Barkovich_Chap07.indd 726 5/6/2011 9:17:04 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 727

FIG. 7-86. Hypothalamic hama-


rtoma within the floor of the third
ventricle. A. Postcontrast CT scan
shows soft tissue mass (arrows) fill-
ing suprasellar and interpeduncular
cisterns. B. Sagittal T1-weighted
image shows soft tissue mass (arrow)
in the floor of the third ventricle. C.
Coronal FLAIR image shows the
gray matter-intensity mass (arrow-
heads) filling the floor of the third
ventricle. D. Axial T2-weighted
image shows the mass (arrow) in the
floor of the third ventricle remain-
ing isointense to gray matter.

be a strong correlation between the presence/onset of seizures and the Classification


presence/onset of developmental abnormalities in this group of chil- Valdueza et al. (493) have proposed a clinico-topographical classifi-
dren (485). Therefore, surgical therapy is becoming more common in cation of hypothalamic hamartomas. Type Ia hamartomas are small,
patients with intractable epilepsy and developmental disorders (486), with a pedunculated attachment to the tuber cinereum, while type Ib
particularly with new, safer approaches being developed (487–491). hamartomas are small pedunculated masses attached to a mamillary
body. Both are asymptomatic or associated with precocious puberty.
Pathology
This group of patients is treated with LHRH analogs.
Hypothalamic hamartomas are well defined, round to oval masses that Type IIa hamartomas are sessile masses, usually larger than 1.5 cm
usually project from the floor of the third ventricle into the lumen of in diameter, attached to the floor of the third ventricle and mamillary
the third ventricle or into the suprasellar or interpeduncular cistern. bodies. Patients typically present with gelastic or generalized seizures.
They may be pedunculated, attached to the tuber cinereum or the Type IIb hamartomas are large sessile masses, usually larger than
mamillary bodies by a thin stalk, or sessile, presenting as a mass within 1.5 cm, that distort the walls and floor of the third ventricle. Patients
the hypothalamus. They range in size from a few millimeters to several have mental and behavioral disorders in addition to gelastic and mixed
centimeters in diameter (492). epilepsy (493,494).
Histologically, hypothalamic hamartomas are composed of a het-
erotopic collection of large and small neurons, astrocytes, and oligo- Imaging Findings
dendroglial cells in proportion to normal neural tissue. Myelinated CT is sensitive only to the presence of large hypothalamic hamartomas.
and unmyelinated axons are seen in bundles, suggesting connections The small, sessile hamartomas arising in the floor of the third ventricle
to other parts of the brain (472). They closely resemble the histologic cannot be seen on axial CT studies. Those hamartomas visible on CT
pattern of the tuber cinereum. Calcification is rare, and hemorrhage is appear as homogeneous, sharply marginated, round masses within the
not described in these lesions (76,480,481). suprasellar and interpeduncular cisterns (Fig. 7-86); they are isodense

Barkovich_Chap07.indd 727 5/6/2011 9:17:05 PM


728 Pediatric Neuroimaging

FIG. 7-87. Pedunculated hypothalamic hamartoma. A. Sagittal T1-weighted image shows a mass (arrow) extending caudally from the tuber cinereum of
the hypothalamus. B. Coronal T1-weighted image shows this pedunculated hamartoma (arrow) clearly outlined by CSF. C. Axial T2-weighted image shows
isointensity of the mass (arrow) to gray matter. D. Sagittal T2-weighted image shows the mass (arrows) as slightly hyperintense compared with cortex;
hamartomas may appear slightly more hyperintense on RARE (FSE, TSE) images compared with conventional spin echo.

with brain tissue. Enhancement does not occur after intravenous on inversion-prepared gradient-echo T1-weighted sequences (475),
infusion of iodinated contrast (480,481,495). Rarely, cystic compo- and isointense to slightly hyperintense on T2-weighted sequences
nents can extend into the adjacent temporal fossa; under these cir- (Fig. 7-87) (496). They range in size from a few millimeters to 3 or 4
cumstances, the solid, nonenhancing tumor may be difficult to identify cm in diameter (480,492). Larger hamartomas are often heterogeneous
prior to shunting of the cyst. on both T1- and T2-weighted images and regions may be quite hyper-
MRI is the study of choice for patients with precocious puberty intense on T2-weighted and FLAIR sequences (Fig. 7-89); the regions
and for patients with epilepsy. The MR appearance of hypothalamic of hyperintensity seem to correspond to an increased glial component
hamartomas is typically that of a well defined, round to ovoid mass histologically (497). No enhancement is seen after intravenous admin-
that lies within the hypothalamus. They typically involve the region of istration of paramagnetic contrast (496). Occasionally, large associated
the mamillary bodies and may extend anterosuperiorly into the third cysts (Fig. 7-90) may be seen in the suprasellar cistern that may extend
ventricle (Fig. 7-86), inferiorly to be suspended from the floor of the into the middle cranial fossa. The diagnosis is made by the characteristic
third ventricle (Fig. 7-87), or into the lateral wall of the hypothalamus location, isointensity to normal brain, and lack of enhancement of the
(displacing the postcommissural fornix and hypothalamic gray matter solid portion of the mass (498–500).
anteriorly) (Fig. 7-88) (475). The hamartomas are isointense compared We have performed proton MR spectroscopy on a single hypotha-
with gray matter on spin-echo T1-weighted sequences, hypointense lamic hamartoma in a neonate; as expected the spectrum was similar

Barkovich_Chap07.indd 728 5/6/2011 9:17:06 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 729

FIG. 7-88. Hypothalamic hamartoma in the wall of the third ventricle. A. Sagittal T1-weighted image shows a small mass (arrow) in the inferior third
ventricle. B. Coronal T2-weighted image shows that the slightly heterogeneous mass (arrows) sits within the right lateral wall of the third ventricle.

to that of normal immature (neonatal) brain tissue. Reports in the skull base, and calvarium. In this section, we consider involvement of
literature suggest that the myo-inositol is slightly higher and NAA the pituitary stalk, the most common intracranial manifestation of the
lower than in normal adjacent brain tissue in the thalamus and fron- disorder. Diabetes insipidus develops when more than 80% of neurons
tal lobe (475,501). The increased myo-inositol seems to be associated in the paraventricular and supraoptic nuclei of the hypothalamus are
with the astrogliosis that is often seen in the hamartomas histologically destroyed (502). It is present in 5% of patients with LCH at the time of
(475,497), which may, in turn, be the result of the epileptic seizures that diagnosis, but in up to 25% of LCH patients and up to 50% of patients
originate from that hamartomas. with multisystemic disease on follow-up examinations (249,503,504).
CNS involvement is most common in patients with multisystemic dis-
Langerhans Cell Histiocytosis ease (249). Involvement of the CNS in LCH starts with the development
LCH was discussed briefly in the “Posterior Fossa Tumors” section of of granulomas in the subarachnoid space, with subsequent invasion of
this chapter, pertaining to its involvement of the brainstem, cerebellum, the hypothalamus and infundibulum. These lesions are characterized

FIG. 7-89. Large, heterogeneous hypothalamic hamartoma. A. Axial T1-weighted image shows a large mass in the suprasellar and interpeduncular cisterns
with slight hypointensity (arrows) on the left side. B. Coronal T2-weighted image better shows the heterogeneity of the mass, with marked hyperintensity
(arrows) in its left lateral portion.

Barkovich_Chap07.indd 729 5/6/2011 9:17:07 PM


730 Pediatric Neuroimaging

FIG. 7-90. Cystic hypothalamic hamartoma. A. Sagittal T1-


weighted image shows a mass (white arrows) in the suprasellar
cistern. A low intensity mass (black arrows) elevates the frontal
lobes. B. Axial T2-weighted image shows that the gray matter-in-
tensity mass (black arrows) is surrounded by CSF (white arrows)
in the suprasellar cistern and in the anterior and middle cranial
fossae. C. Coronal postcontrast T1-weighted image shows that
the suprasellar mass does not enhance. A small hypointensity
(arrow), presumably a cystic or necrotic area, is seen within the
mass.

by diffuse parenchymal infiltration by CD1a+ histiocytes, CD68+ infundibulum (Fig. 7-91) can be appreciated in both the sagittal and
macrophages, T cells and B cells with surrounding inflammation and coronal planes (509). Larger hypothalamic masses are easily identi-
nearly complete loss of neurons and axons (250,504). Ultimately, such fied, generally centered in the superior aspect of the stalk (Fig. 7-92).
granulomas may develop throughout the brain parenchyma (175,249), Small or large, the mass enhances markedly after intravenous infusion
meninges, choroid plexus (505), pineal gland (506), and spinal cord of paramagnetic contrast (Figs. 7-91 and 7-92). Prior to the onset of
(507). Brain parenchymal lesions in LCH, which are very rare, are dis- diabetes insipidus, the posterior pituitary retains its normal T1 hyper-
cussed briefly in the “Posterior Fossa Tumors” section of this chapter. intensity. After the onset of diabetes insipidus, the high signal in the
Calvarial, dural, and plexal lesions are discussed in the section on posterior pituitary on T1-weighted images is not present (Figs. 7-91
“Extraparenchymal Tumors.” and 7-92) (510). Diffusion imaging and spectroscopy have not been
When the pituitary stalk and hypothalamus are involved by LCH, used to aid this diagnosis.
the involvement varies in size from mild thickening of the infundibu- The differential diagnosis of a thickened, enhancing pituitary stalk
lum (508) to a frank hypothalamic mass. Mild thickening of the or a larger mass centered in the stalk is large, and includes germ cell
infundibulum is difficult to see on CT without the use of intravenous tumors, lymphocytic hypophysitis, lymphoma, and granulomatous
contrast, especially if images are obtained only in the axial plane. diseases such as tuberculosis and sarcoidosis, in addition to LCH. In the
When images are obtained in the coronal plane, both the thickening absence of bone involvement by the LCH, the other differential con-
and the enhancement of the infundibulum can be appreciated. MRI siderations need to be strongly considered in children with diabetes
is the imaging modality of choice, as even mild thickening of the insipidus (422).

Barkovich_Chap07.indd 730 5/6/2011 9:17:09 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 731

FIG. 7-91. Small Langerhans cell histiocytosis and lymphocytic hypophysitis. A and B. Langerhans cell histiocytosis. Sagittal T1-weighted image (A) shows
an enlarged infundibulum (arrows) and absence of the posterior pituitary gland (the “pituitary bright spot”). Postcontrast T1-weighted image (B) shows
uniform enhancement (arrows) of the enlarged infundibulum. C and D. Lymphocytic infundibulohypophysitis. Sagittal T1-weighted image (C) shows an
enlarged infundibulum (arrow). Postcontrast image shows marked enhancement of the infundibular mass. Note that the hyperintense posterior pituitary is
seen despite the presence of the infundibular lesion.

Pediatric Pituitary Tumors amenorrhea (514). In contrast to adults, pituitary tumors in children
Tumors of the pituitary gland are uncommon in children, with pedi- are more commonly macroadenomas than microadenomas. This fact
atric patients comprising only about 2% of those with pituitary ade- may reflect a lack of symptoms in boys and premenarchal girls with
nomas (511–513). When pituitary tumors do become symptomatic prolactin secreting tumors. It might also result from a shorter time to
in the pediatric age group, adolescents are usually affected (512,513). progression in pediatric pituitary adenomas (512,515,516); the shorter
The most common hormonally active pituitary adenomas in the pedi- time to progression suggests that pediatric pituitary adenomas are
atric age group are prolactin-secreting adenomas (typically result- more biologically aggressive tumors (515). Children with large pitu-
ing in delayed menarche), ACTH-secreting adenomas (resulting in itary tumors may present with short stature, secondary to compromise
Cushing disease), and growth hormone-secreting adenomas (result- of hypothalamic-pituitary function, or with visual disturbance if the
ing in gigantism) (513). About 25% of pediatric pituitary adenomas tumor extends into the suprasellar cistern (453,511,512). Rarely, chil-
are nonfunctioning (453,512); nonfunctioning adenomas typically dren and adolescents may present with pituitary apoplexy, a condition
present with delayed puberty, short stature, or (in females) primary caused by sudden enlargement of a pituitary adenoma secondary to

Barkovich_Chap07.indd 731 5/6/2011 9:17:10 PM


732 Pediatric Neuroimaging

FIG. 7-92. Large Langerhans cell histiocytosis lesion. Precontrast (A) and contrast-enhanced (B) sagittal T1-weighted images show a large hypothalamic
mass (arrows) that uniformly enhances after administration of contrast.

extensive tumor infarction or hemorrhage. Pediatric patients with can result in invasion of the cavernous sinuses (520). After intravenous
pituitary apoplexy manifest acute headache and visual loss, identical to contrast administration, microadenomas will typically remain hypoin-
their adult counterparts (517,518). tense compared to the enhancing gland on early images (519). However,
if imaging is delayed, the adenoma will typically become isointense, or
Imaging Findings even hyperintense, compared with the surrounding normal gland (521).
The imaging appearance of pituitary adenomas in children is identi- Therefore, the patient should be imaged as soon as possible after admin-
cal to that in adults. MRI is the imaging modality of choice (519,520). istration of contrast. Dynamic scanning, in which multiple images are
Precontrast and postcontrast thin section (3 mm or less) coronal and acquired through several sections of the pituitary gland during the infu-
sagittal T1 and T2 images should be acquired through the sella and sion of contrast, can be useful in detecting enhancing tumors while still
suprasellar regions. T2-weighted images are useful to look for cysts hypointense (before they become isointense) to the surrounding gland
or evidence of hemorrhage. Microadenomas will appear as small (522,523); we routinely perform dynamic scanning when looking for
(<1 cm) hypointense masses (Fig. 7-93) within the gland on noncontrast microadenomas. Macroadenomas typically enhance uniformly and
scans. Secondary signs include deviation of the pituitary stalk to the intensely (519). Caution must be exercised when examining the ado-
side opposite the tumor and upward convexity of the gland. Macroad- lescent gland, as the hormonal changes during adolescence result in an
enomas will expand the gland and, if the diaphragma sella is sufficiently enlargement of the gland, with upward convexity of the superior surface
large, will dumbbell into the suprasellar cistern, where the optic chiasm (see Chapter 2 and Fig. 2-18) (524). A focal lesion must be identified
and infundibulum can be compressed. Lateral extension of the tumor before an adenoma is diagnosed on the basis of the imaging study.

FIG. 7-93. Pituitary microadenoma in a patient with Cushing syndrome. Postcontrast sagittal (A) and coronal (B) images show a small, nonenhancing
mass (arrows) in the anterior pituitary lobe.

Barkovich_Chap07.indd 732 5/6/2011 9:17:11 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 733

In pituitary apoplexy, a sellar mass with suprasellar extension is that about two thirds present with manifestations of pituitary
identified. The mass shows homogeneous or heterogeneous high sig- dysfunction, ranging from isolated hormonal deficiencies (typically
nal intensity on T1-weighted images secondary to the intratumoral growth hormone or prolactin) to generalized pituitary hypofunction;
hemorrhage. The diagnosis is made by a combination of the history visual disturbances are present in about half and headaches in about
and neuroimaging appearance (525). Pituitary apoplexy is an emergent 30% (526).
condition, as permanent visual loss may result if the acute compression of CT scans typically show a round or lobulated intrasellar or supra-
the optic chiasm is not rapidly relieved. If this appearance is identified, the sellar mass with attenuation values similar to CSF and characteristi-
referring physician must be contacted immediately or the patient should cally without any evidence of calcification (529). MRI shows a round to
be referred to the nearest emergency department. ovoid, sharply defined mass that typically lies either between the ante-
rior and posterior pituitary lobes (Fig. 7-94) or immediately anterior
Rathke Cleft Cysts to the insertion of the pituitary stalk into the gland (Fig. 7-95). The
Rathke cleft cysts are benign epithelium-lined cysts that arise primar- cyst has a variable signal intensity, ranging from isointense to hyperin-
ily in the sella turcica and contain mucoid material. They are rare in tense compared to CSF on T1-weighted images and from isointense to
children. Although nearly always asymptomatic, they may reportedly slightly hypointense compared to CSF on T2-weighted images (530).
produce symptoms by compression of the pituitary gland or supra- No enhancement is seen after administration of paramagnetic contrast
sellar structures. They are thought to derive from remnants of Rathke (Fig. 7-94); however one must be aware of the fact that in some cases the
pouch; thus, they may be embryologically related to craniopharyn- cyst is surrounded by a layer of stretched enhanced pituitary tissue that
giomas (450,526). Recent experience with high quality neuroimaging may lead to the wrong diagnosis of enhancing cyst. Not uncommonly,
suggests that cysts within the pituitary fossa are common particularly the cyst is hyperintense compared to normal gland on precontrast MR
in children and that most of these cysts (presumably Rathke cleft cysts) images and hypointense compared to enhancing gland on postcontrast
are asymptomatic (527). Indeed, they are reported in up to 33% of images (Fig. 7-94). When a sharply demarcated mass situated posterior
routine autopsy series (528). Of the few with symptoms, it is estimated to the anterior pituitary lobe has these signal characteristics, Rathke

FIG. 7-94. Rathke cleft cyst. A. Sagittal T1-weighted image shows


a mass (arrows), situated between the anterior and posterior pitu-
itary lobes that is slightly hyperintense compared to normal gland.
B. After contrast administration, the mass (arrows) remains unchanged
as the gland enhances. This change from relative hyperintensity to
relative hypointensity is characteristic of a Rathke cleft cyst. C. Coro-
nal T2-weighted image confirms the absence of blood by showing the
lesion (arrows) to be hyperintense (blood would be hypointense).

Barkovich_Chap07.indd 733 5/6/2011 9:17:12 PM


734 Pediatric Neuroimaging

FIG. 7-95. Rathke cleft cyst. A. Sagittal T1-weighted image shows an ovoid hyperintense mass (arrows) directly anterior to the infundibulum. B. Coronal
T1-weighted image shows the mass (arrows) situated between the pituitary gland and the optic chiasm.

cleft cyst should be strongly considered. It is distinguished from a here. Suprasellar germ cell tumors have imaging characteristics similar
hemorrhagic adenoma by the absence of hypointensity on T2-weighted to suprasellar LCH and lymphocytic neurohypophysitis. They origi-
or susceptibility-weighted images (Fig. 7-94C). nate in the hypothalamus and grow into the infundibulum, most com-
Rarely, Rathke cleft cysts can become infected, resulting in a pitu- monly causing diabetes insipidus, which is usually the reason affected
itary abscess (531). Affected patients typically have visual symptoms, patients seek medical attention. Imaging characteristics vary with
often accompanied by fever. The offending organism may or may not histology and with the size of the tumor at the time of imaging. Ger-
be identified. On imaging, the characteristic appearance is that of a minomas, by far, the most common histologic type in the suprasellar
rim-enhancing mass with central T1 and T2 prolongation (531). region, have typical imaging features when they are small, appearing
on MR as thickening of the infundibulum and uniformly enhancing
Lymphocytic Hypophysitis after administration of contrast (Fig. 7-96). When small, they can-
Lymphocytic hypophysitis is an autoimmune inflammatory infil- not be differentiated from lymphocytic hypophysitis or LCH. CT is
tration of the pituitary gland by lymphocytes and plasma cells that often normal. With slightly larger germinomas, CT shows homoge-
leads to gradual destruction of the gland. It can be divided into two neous masses of gray matter attenuation that uniformly enhance after
major forms: adenohypophysitis, which involves mostly the anterior administration of iodinated contrast (Fig. 7-97). MRI shows masses of
pituitary gland, and infundibuloneurohypophysitis, which involves gray matter intensity in the midline of the hypothalamus that enhance
primarily the posterior pituitary gland, infundibulum, and hypothala- uniformly. Typically, the high signal intensity of the posterior pituitary
mus (532,533), although any combination of anterior gland, posterior lobe will not be seen on sagittal T1-weighted images due to block-
gland, and infundibulum may be affected (534). It is primarily a disease age of the infundibulum by the mass. Large suprasellar germinomas
of adults, but children may be affected. A slight female predominance are much more heterogeneous (Fig. 7-98), probably due to necrosis
has been noted, with the mean age of diagnosis being 12 years (534). in the more central regions of the tumor. They are much bulkier and
Presentation depends upon the portions of the gland that are affected. often extend down the infundibulum into the sella turcica, displacing
Diabetes insipidus is the most common symptom (534), but patients the pituitary gland (Fig. 7-98). When large, the MR appearance of a
may present with hypopituitarism, headache or impaired vision. suprasellar germinoma is very similar to that of a suprasellar astrocy-
Hypothalamic-pituitary dysfunction, most commonly diabetes insipi- toma; however, isointensity of solid portions of the tumor to gray mat-
dus, elevated prolactin, and reduced gonadotropins, ACTH, and growth ter on T2-weighted images, reduced diffusion, and tumor extension
hormone may be found on endocrinologic testing (534–537). MRI (the down the infundibulum suggest the presence of a germinoma (445).
neuroimaging study of choice) shows an enlarged median eminence, The differentiation is more easily made from the history; patients with
infundibulum, and, sometimes, pituitary gland. The enlarged regions suprasellar germinomas nearly always have diabetes insipidus at the
typically show uniform enhancement after administration of para- time of presentation, whereas patients with suprasellar astrocytomas
magnetic contrast. The imaging findings can be identical to those of rarely develop diabetes insipidus until later in the course of the disease
suprasellar germinomas and granulomatous conditions such as LCH (445). Imaging is more helpful in differentiating suprasellar germino-
(Fig. 7-91C and D), sarcoidosis, and tuberculosis (534–536,538). Dif- mas from craniopharyngiomas; germinomas rarely have large cystic
ferentiation must be made by clinical presentation, laboratory studies, components and rarely calcify (445).
or biopsy (536,537). It is important to recognize that at the time that the child pres-
ents with diabetes insipidus, the suprasellar germinoma may be small
Suprasellar Germ Cell Tumors (Fig. 7-96) or not yet be visible on an imaging study. In such patients, a
Germ cell tumors are discussed in the following section on tumors of the repeat imaging study should be obtained in 3 to 6 months and, if still
pineal region, and only their imaging characteristics will be described negative, a second repeat examination should be obtained after another

Barkovich_Chap07.indd 734 5/6/2011 9:17:13 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 735

FIG. 7-96. Small suprasellar germinoma in a patient with diabetes insipidus. Sagittal (A) and coronal (B and C) postcontrast images reveal a slightly thickened
infundibulum, with the enhancing mass (arrows) extending superiorly into the tuber cinereum of the hypothalamus and inferiorly into the pituitary gland.

FIG. 7-97. Moderate sized suprasellar germinoma. A. Sagittal T1-weighted image shows a mass (arrows) in the hypothalamus/floor of the third ventricle.
Note that the pituitary gland is small and the high signal intensity of the posterior pituitary gland is not seen. B and C. Sagittal T2-weighted (B) and coronal
FLAIR (C) images show that the mass (arrows) is homogeneous and has gray matter intensity. D. Coronal postcontrast T1-weighted image shows uniform
enhancement (arrows) of the mass.

Barkovich_Chap07.indd 735 5/6/2011 9:17:14 PM


736 Pediatric Neuroimaging

FIG. 7-98. Large suprasellar germinoma. A. Axial noncontrast CT image shows the suprasellar mass (arrows) that is hyperdense compared to surround-
ing brain. Small, round cell tumors are typically hyperdense compared to normal brain. B. After administration of intravenous iodinated contrast, the
germinoma uniformly enhances. C. Sagittal T1-weighted image shows the mass (arrows) extending from the inferior third ventricle downward in to the sella
turcica, filling the suprasellar cistern. D. Postcontrast sagittal image shows slightly heterogeneous enhancement of the tumor.

3 to 6 months. A uniformly enhancing mass will often develop in the tumor is still small; at this stage, many of them are curable with radia-
infundibulum; biopsy of the mass will show a germinoma (most com- tion and chemotherapy.
mon), LCH, or lymphocytic hypophysitis (422).
Germ Cell Tumors
Suprasellar Arachnoid Cysts Most tumors arising in the pineal region are germ cell tumors, a group
Arachnoid cysts are discussed in Chapter 5. of neoplasms that comprise about 3% to 8% of pediatric brain tumors
(539,540). They are considerably more common among Asian popu-
lations, where they comprise approximately 10% of pediatric brain
Pineal Region Masses
tumors, than populations of Europe or North America, where the
Pineal region masses are an important group of pediatric brain tumors figure is closer to 2% to 4% (77,541). The incidence of germ cell tumors
because most affected children present with hydrocephalus when the appears to be increased in patients with Klinefelter syndrome (542).

Barkovich_Chap07.indd 736 5/6/2011 9:17:16 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 737

Of germ cell tumors, approximately 65% are germinomas (sometimes of the solid portions of the tumor is seen after intravenous infusion of
incorrectly referred to as atypical teratoma, pinealoma, or dysgermi- contrast. Germinomas in the suprasellar and basal ganglia regions can
noma), 26% are nongerminomatous (16% teratomas, 6% endodermal be differentiated from astrocytomas by their high attenuation prior to
sinus tumors [also called yolk sac tumors] or embryonal cell carcinoma, contrast infusion (Fig. 7-98) (10,401,408,409).
and 4% choriocarcinomas), and 9% are mixed (540). Other series On MRI, germinomas usually appear as well-marginated tumors,
have a greater percentage of mixed histology (541). Germ cell tumors either round or lobulated, that demonstrate isointensity to hypoin-
develop most commonly in the pineal region, with about half of all tensity compared with gray matter on T1-weighted, T2-weighted,
intracranial germ cell tumors being found in this region (539–541). and FLAIR images (Figs. 7-97 and 7-99) (411). The T2 hypointensity
Patients with pineal region germ cell tumors most commonly present presumably results from the diminished free water content of these
with intracranial hypertension secondary to hydrocephalus, Parinaud tumors. Diffusion of solid portions is often reduced compared with
sign, Argyll-Robertson pupillary deficit, or diplopia and possibly in the cerebral hemispheric parenchyma on diffusivity maps. The perfusion
first decade, precocious puberty (541). About 30% of germ cell tumors and spectroscopy characteristics of these tumors are not known.
develop in the suprasellar/hypothalamic region; this group of patients When a relatively shortened T2 relaxation time is present in a
typically presents with diabetes insipidus, visual disturbances, or amen- pineal region tumor in an adolescent, the diagnosis of germinoma
orrhea (541). Other, less common, locations include the basal ganglia is suggested; in younger children, pineoblastomas can have a similar
(increased intracranial pressure, hemiparesis, discussed in the section MRI appearance (although with lower Dav than germinoma) (549,550).
on “Tumors of Cerebral Hemispheres”), cerebellopontine angle (cranial Some internal heterogeneity is often seen in large germinomas (Figs.
neuropathy), cerebellum (increased intracranial pressure), corpus callo- 7-98 and 7-99), probably secondary to focal necrosis or microcystic
sum (diplopia), and spinal cord (myelopathy) (402,539–541,543–545). change. The solid portions of germinomas strongly enhance after infu-
sion of paramagnetic contrast (Figs. 7-97 to 7-100) (411), as do CSF-
Germinomas borne metastases to the brain (Fig. 7-100) and spine. When large, the
Germinomas are the most common neoplasm in the pineal region, MR appearance of a suprasellar germinoma is very similar to that of
accounting for over 50% of neoplasms in this area. Most germinomas a suprasellar astrocytoma; however, isointensity of solid portions
occur in the second and third decades of life, with a peak incidence in of the tumor to gray matter on T2-weighted images, reduced diffusion,
the latter half of the second decade. Patients with pineal germinomas and tumor extension down the infundibulum suggest the presence
show approximately a 10:1 male predominance and most commonly of a germinoma (445). The differentiation is more easily made from
present with hydrocephalus or the Parinaud syndrome (paralysis of the history; patients with suprasellar germinomas nearly always have
upward gaze), resulting from compression of the aqueduct of Sylvius diabetes insipidus at the time of presentation, whereas patients with
and midbrain, respectively (546). Approximately 35% of intracranial suprasellar astrocytomas rarely develop diabetes insipidus until later in
germinomas occur in the suprasellar region. The imaging appearance the course of the disease (445). Imaging is more helpful in differenti-
of these tumors was briefly discussed in the previous section on “Sellar ating suprasellar germinomas from craniopharyngiomas; germinomas
and Suprasellar Tumors”; a more wide-ranging discussion is given rarely have large cystic components and rarely calcify (445).
here. Suprasellar germinomas affect males and females with equal fre-
quency; they commonly cause symptoms indicative of hypothalamic Teratomas
involvement such as diabetes insipidus, emaciation, or precocious Although the pineal region is the most common site of intracranial
puberty. It is of interest that patients with pineal germinomas fre- teratomas, they may also be situated in a parapineal region or in the
quently have symptoms indicative of hypothalamic involvement; these suprasellar region. Other than in neonates and infants (see section on
symptoms most likely result from spread of tumor through the CSF of “Tumors in the First Year of Life” later in this chapter) pineal region
the third ventricle to the infundibular recess with subsequent invasion teratomas are much less common than germinomas. As with germi-
of the hypothalamus. Some authors, however, believe that the cause of nomas, pineal teratomas occur almost exclusively in males; however,
these symptoms is multifocality of the tumor (8,76,402,539,540,547). in contradistinction to germinomas, teratomas in the suprasellar
Between 4% and 14% of intracranial germinomas tumors originate in location have a marked male predominance. The peak incidence of
the basal ganglia or thalamus (discussed earlier in this chapter under presentation is in the second decade. Teratomas are similar to other
“Tumors of the Cerebral Hemispheres”). pineal region germ cell tumors, in that affected patients may present
with the Parinaud syndrome, hydrocephalus, and hypothalamic symp-
Pathology toms, the exact presentation being dependent upon tumor location
Pineal germinomas usually are well-defined lesions restricted to the (8,408,541,547,548).
pineal gland; rarely they occur in a parapineal location. Occasionally,
adjacent structures, especially the quadrigeminal plate or the thalami, Pathology
are infiltrated by tumor. Suprasellar germinomas are round or lobulated Benign teratomas are usually sharply marginated, extremely hetero-
masses that arise in the pituitary stalk and floor of the third ventricle, geneous masses with a rounded or irregular, lobulated outline. Cystic
subsequently compressing and invading the optic chiasm. They may areas and recognizable teratoma elements, such as cartilage, bone, hair
extend upward to infiltrate the walls of the third ventricle, mimicking and fat, are common. In contrast to those in other parts of the brain,
an infiltrating glioma. Metastatic spread occurs early and frequently via pineal region teratomas tend to be noninvasive (8,76). Malignant tera-
the CSF, resulting in many pial nodules or, sometimes, a sugar-coated tomas typically have less well differentiated cells and, therefore, less
appearance (8,76,401,408,547,548). well differentiated features. When highly malignant (composed pri-
marily of anaplastic cells), the masses have a much more homogeneous
Imaging Studies appearance and may invade surrounding brain (8,76).
The CT appearance of a germinoma is that of an isodense to hyper-
dense, well-marginated mass, typically occurring in either the pineal or Imaging Studies
suprasellar region. Occasionally, the tumor may originate in the deep As with teratomas in other parts of the brain and body, the hallmark of
cerebral nuclei, cerebellum, or other regions. Uniform enhancement these tumors is their heterogeneity. On CT, they are sharply marginated

Barkovich_Chap07.indd 737 5/6/2011 9:17:17 PM


738 Pediatric Neuroimaging

FIG. 7-99. Pineal germinoma. A. Sagittal T1-weighted image shows a heteroge-


neous mass in the pineal region (arrows). B. Axial T2-weighted image shows the
tumor mass to have heterogeneous signal intensity with areas of short T2 relax-
ation time (arrows). C. After intravenous infusion of gadolinium-DTPA, there is
marked contrast-enhancement of the tumor (arrows).

masses distinguished by areas of fat and calcification in combination malignant teratomas are poorly marginated, homogeneous masses that
with cystic and solid areas (Fig. 7-101A). The MR appearance is also have irregular shape and elicit vasogenic edema in surrounding tissues.
usually one of an extremely heterogeneous mass, usually arising from Calcification and fat are not seen. Significant enhancement is typically
the tuber cinereum or infundibulum (Fig. 7-101). The presence of fat, seen after administration of paramagnetic contrast (237).
calcium, cysts, and soft tissue give a variegated appearance on T1- and
T2-weighted images (Figs. 7-101 and 7-102) (445,550). Immature Other Germ Cell Tumors
or malignant teratoma has a similar pattern to that of mature tera- Choriocarcinoma, embryonal cell carcinoma, and endodermal sinus
toma, but the cystic components and calcifications tend to be less and tumors are highly malignant germ cell tumors that occur in the
smaller, respectively. The tumor margins may be obscure in malignant pineal region. Affected children typically present with either hydro-
teratoma, and perifocal edema may be observed in some cases (551). cephalus and/or the Parinaud syndrome. Embryonal cell carcino-
Enhancement after infusion of intravenous contrast may be seen in mas and endodermal sinus tumors are heterogeneous masses that do
solid portions; its presence does not necessarily indicate malignant not have specific findings that allow them to be diagnosed by imag-
degeneration. An extremely heterogeneous mass in the pineal or supra- ing (237,411,551). Choriocarcinomas are notoriously hemorrhagic;
sellar area should be strongly suggestive of a benign teratoma, partic- therefore, a hemorrhagic tumor in the pineal region (where most
ularly if it contains fat or calcium. Highly malignant teratomas have tumors have a very low tendency to hemorrhage) is suggestive of a
a very different appearance than their benign counterparts. Highly choriocarcinoma (408,547,550,551). Tumor markers, in particular

Barkovich_Chap07.indd 738 5/6/2011 9:17:17 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 739

FIG. 7-100. Pineal region germinoma


with CSF-borne metastases. A. Sagittal
T1-weighted image shows a large pineal
region mass that has invaded the quadri-
geminal plate (arrows). B. After intrave-
nous infusion of paramagnetic contrast,
heterogeneous enhancement of the pineal
region mass is seen (large white arrows).
In addition, a small focus of enhance-
ment is seen in a thickened infundibu-
lum (small white arrow). The patient had
diabetes insipidus as a result of metasta-
sis of tumor to the pituitary stalk. C and
D. Coronal T1-weighted images show
the enhancing tumor mass in the pineal
region (closed arrows) and a subependy-
mal metastasis in the atrium of the right
lateral ventricle (open arrow).

placental alkaline phosphatase (PLAP), alpha fetoprotein (AFP), and Pineal Parenchymal Tumors
the beta subunit of human chorionic gonadotropin (b-HCG), are Pineocytomas and pineoblastomas are both tumors that arise from
often secreted by germ cell tumors and can be assessed by serological pineal parenchymal cells. Both tumor types are considerably less
and CSF analyses. PLAP is secreted primarily by germinomas, b-HCG common in children than pineal germ cell tumors. Pineocytomas
by choriocarcinomas, AFP by endodermal sinus tumors, and both affect both sexes equally; they may be found at any age, but are very
AFP and b-HCG by embryonal cell carcinomas (552). Therefore, the uncommon in children. They are composed of relatively mature cells
specificity of preoperative diagnosis is aided by analysis of the tumor and are usually circumscribed, noninvasive, slowly growing tumors.
markers (551–553). An important concept is that 10% (or more) of Because they are rare, the natural history of pineocytomas is not
germ cell tumors are of mixed histology; for example there may be well-defined (554). Some that remain circumscribed and noninva-
components of germinoma, embryonal cell carcinoma, and teratoma sive are found after several years of clinical symptoms or are dis-
in the same tumor. Therefore, a needle aspiration of the tumor does closed at postmortem examination. Others metastasize extensively
not obtain adequate tissue to properly characterize the mass; open through the cerebrospinal fluid and behave more like the primitive
biopsy is necessary (551,553). tumor of this group, the pineoblastomas. Some evidence indicates

Barkovich_Chap07.indd 739 5/6/2011 9:17:18 PM


740 Pediatric Neuroimaging

FIG. 7-101. Pineal region teratoma. A. Axial noncontrast CT scan


shows a large mass in the pineal region and extending anteriorly into
the third ventricle (black arrows). Foci of calcification (open white
arrows) and fat (solid white arrows) are present as well. B. Sagittal
T1-weighted image shows the large heterogeneous tumor mass
(arrows) within the third ventricle. The high intensity regions rep-
resent fat. Calcification is not well seen on MR images. Note the
extreme heterogeneity of the tumor. C. Axial T2-weighted image
shows the markedly heterogeneous tumor (arrows) with areas of
high signal intensity, low signal intensity, and intermediate signal
intensity occupying the pineal region and the third ventricle.

FIG. 7-102. Suprasellar teratoma in an


infant with growth delay. A. Axial non-
contrast CT shows a fatty suprasellar mass
(arrow) with a focus of calcification within
it. B. Sagittal T1-weighted image shows a
heterogeneous mass (arrow) extending from
the mammillary body to the infundibulum.
Both fatty components and nonfatty com-
ponents (open arrows) are present. Mild
enhancement of the nonfatty components
was seen on the postcontrast scan.

Barkovich_Chap07.indd 740 5/6/2011 9:17:19 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 741

TABLE 7-10 Pineal Region Tumors in Children


Tumor Key Characteristics
Germ cell tumors Most common solid pediatric tumor
Germinomas most common: gray matter intensity
Imaging varies with histology
Pineal parenchymal tumors Solid portions: gray matter intensity
Dermoids and epidermoids Similar to imaging characteristics in other locations
Pineal region gliomas Look for extension from adjacent brain
Pineal cysts Most common pineal mass
Cyst with rim of enhancing tissue
Vein of galen varices (see Chapter 12) Look for signal void from flow

that pineocytomas in children are aggressive tumors that should be the margins of the pineoblastomas are less sharply demarcated (558).
treated (555). Pineoblastomas are primitive, small round cell tumors They are usually isointense to hypointense on T1-weighted sequences
that are highly cellular and are best classified as PNETs, grade IV and isointense to hyperintense compared with gray matter on FLAIR
embryonal tumors. They are highly malignant, and demonstrate local and T2-weighted images (Figs. 7-103 and 7-104); they demonstrate
invasion as well as distant spread through the cerebrospinal fluid. reduced diffusivity compared with normal brain parenchyma. Necrotic
They may be found at any age but are more common in the pediatric or cystic areas (hypointense on T1-weighted images and hyperintense
age group, with boys affected more frequently than girls (8,76,556). on T2-weighted images) are commonly seen in larger pineoblastomas
As with most pineal region tumors (Table 7-10), affected children (Fig. 7-103). The solid portions of the tumor enhance intensely after
are brought to attention by signs and symptoms of hydrocephalus or intravenous contrast administration (Fig. 7-104) (561,562). Pineoblas-
Parinaud syndrome (556). tomas very rarely bleed; the author has seen one such case. As with
other pineal region tumors, pineoblastomas are known to metastasize
Imaging Studies frequently through the CSF; postcontrast scans of the entire neural
On CT, both pineocytomas and pineoblastomas are isodense to hyper- axis are essential to detect and define this subarachnoid tumor, which
dense to normal brain prior to contrast infusion (Fig. 7-103). In con- enhances markedly (564). The perfusion and spectroscopy characteris-
trast to germinomas, which displace the calcified pineal gland, pineal tics of these tumors are not published. In our limited experience, pine-
parenchymal tumors usually contain areas of calcification within them; oblastomas have significantly increased blood volume compared with
this feature is an important differentiating point (557). Pineoblasto- normal parenchyma.
mas are usually more lobulated than pineocytomas (558). Both show
marked enhancement, with pineocytomas having generally more uni- Dermoids and Epidermoids
form enhancement, while pineoblastomas have more heterogeneous Dermoids and epidermoids commonly occur in the pineal region.
enhancement after the infusion of intravenous contrast (554,555,558– Their characteristics are described earlier in this chapter.
560). In this author’s experience, pineoblastomas more commonly
have hypodense, nonenhancing foci within them and are larger and Pineal Region Gliomas
more irregular in shape at the time of presentation than pineocytomas Gliomas, primarily astrocytomas, can also occur in the pineal region.
or germinomas (Fig. 7-103). They frequently extend inferiorly into the Gliomas rarely develop from astrocytes within the pineal gland itself
posterior fossa and can invade the cerebellar vermis. Moreover, they (565). Much more commonly, the tumors arise from adjacent brain
can extend anteriorly into the third ventricle or laterally into the walls parenchyma, such as the quadrigeminal plate or thalamus, and grow
of the third ventricle. Pineocytomas tend to be smaller and much less into the pineal region secondarily (76,557). Tumors originating from
invasive (10,408,409). the quadrigeminal plate are considered a subgroup of brainstem
The MR appearance of pineal parenchymal tumors is nonspecific tumors and are discussed more fully earlier in this chapter. Patients
(550,561,562). However, the diagnosis should always be considered typically present with headaches, nausea, vomiting, or sleepiness sec-
when a tumor with proper MR characteristics is found in the pineal ondary to hydrocephalus. Other than those from hydrocephalus, they
region of a girl or woman and suggested if no other predisposing have few, if any, abnormalities on neurological examination; thus a
conditions (other primary malignancy, immunocompromised state) significant delay often occurs between the onset of symptoms and the
are present. Pineocytomas are typically hypointense on T1-weighted time of diagnosis (193,566). Histologically, most of the tumors in this
images and hyperintense on T2-weighted images compared with gray region are pilocytic astrocytomas (566); they are quiescent and rarely
matter; they may be isointense with CSF on these sequences. They are progress (193).
differentiated from pineal cysts by the presence of trabeculae within
the tumor mass and by diffuse, homogeneous enhancement after intra- Imaging Studies
venous contrast administration (558,563). Pineoblastomas are usually Imaging studies reveal a mass, originating most commonly in the
larger and more lobulated than pineocytomas (Figs. 7-103 and 7-104); quadrigeminal plate, but sometimes in the thalamus, medial temporal

Barkovich_Chap07.indd 741 5/6/2011 9:17:21 PM


742 Pediatric Neuroimaging

FIG. 7-103. Pineoblastoma. A. Noncontrast CT scan shows a mass in the


pineal region (arrows). A small low attenuation center is seen within the
mass. Pineoblastomas are usually iso- to hyperintense with respect to brain
on CT. B. Sagittal T1-weighted image shows the mass extends from the
pineal region well below the tentorial incisura into the posterior fossa (large
arrows). There are multiple small cystic/necrotic areas within the tumor
(small black arrows). Note the inferior displacement of the fourth ventricle
(small white arrows). C. Axial T2-weighted image shows the solid portion of
the tumor to be isointense with gray matter (arrows).

FIG. 7-104. Pineoblastoma. A. Sagittal


T1-weighted image shows the presence
of hydrocephalus, caused by an iso- to
slightly hypointense mass (arrows) in
the pineal region. B. Axial T2-weighted
image shows the mass (arrows) to be
gray matter intensity with small cen-
tral regions of hyperintensity that likely
represent necrosis

Barkovich_Chap07.indd 742 5/6/2011 9:17:21 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 743

FIG. 7-104. (Continued)


C. Postcontrast sagittal
T1-weighted image after
shunting of ventricles shows
that the lobulated mass
enhances heterogeneously. A
small cyst is seen inferiorly
(arrow) where the tumor
enters the enlarged Sylvian
aqueduct. D. Axial calculated
Dav image shows reduced dif-
fusion of the mass (arrows)
compared with normal cere-
bral parenchyma.

lobe, or the medial occipital lobe, and growing into the pineal region whether growth on an imaging study, without the development of clin-
(Fig. 7-105). If the tumor is very large at the time of the initial imaging ical symptoms, is in indication for therapy (193,197).
study, the site of origin may become obscured; under these circum-
stances, differentiation of a primary pineal tumor from a parapineal Pineal Cysts
tumor growing into the pineal region is impossible. The tumors are Small nonneoplastic cysts of the pineal gland are common, incidentally
typically hypodense on CT, hypointense on T1-weighted images, and found in up to 40% of autopsies (567–569). These lesions are uncom-
hyperintense on T2-weighted and FLAIR images prior to contrast mon in infancy; they seem to develop during adolescence and their
administration. Enhancement is variable; those arising in the tectum incidence is highest in the third decade (570). Their observation on
enhance little, if at all, while those arising from the thalamus may imaging studies is nearly always incidental, as they are essentially never
enhance mildly or moderately. Most tumors are stable in size, but symptomatic unless complications, such as hemorrhage, ensue. Their
slow enlargement may be seen on sequential studies; it is controversial cause is not known, and authors have postulated cystic degeneration

FIG. 7-105. Pineal region astrocytoma. A. Sagittal T1-weighted image shows hydrocephalus (note expanded anterior recesses of the third ventricle) and
a hypointense, poorly marginated mass (arrows) in the posterior third ventricle, extending into the aqueduct and tectum to the pineal region. B. Axial
T2-weighted image shows the mass (arrows) to be hyperintense compared to surrounding brain tissue.

Barkovich_Chap07.indd 743 5/6/2011 9:17:23 PM


744 Pediatric Neuroimaging

FIG. 7-106. Pineal cyst. A. Sagittal T1-weighted image


shows a sharply marginated ovoid mass in the pineal
region (large arrows). The mass is isointense with CSF.
Note that there is some compression of the superior col-
liculi (small arrows). In the absence of the Parinaud syn-
drome or hydrocephalus, such compression should not
cause alarm. These findings are classic for a pineal cyst.
B. After administration of paramagnetic contrast, the
rim of normal pineal gland around the cyst (arrows)
enhances.

of the gland, enlargement of microscopic cysts under hormonal influ- Extraparenchymal Tumors
ence, and enlargement of the embryonic pineal cavity (571).
Although many are too small to see on routine imaging studies, Supratentorial extraparenchymal tumors are rather uncommon in
pineal cysts are, nonetheless, a frequent observation on MR studies of children. By far, the most common are tumors originating from the
the brain. They can be identified in up to 23% of brain MR examinations choroid plexuses of the lateral ventricles and those originating in
(572–575). Pineal cysts are infrequently noted on CT scans, other than the calvarium, the most common of which is LCH. Key features of
in retrospect, because they are isointense with CSF in the surrounding supratentorial extraparenchymal tumors are outlined in Table 7-11.
cisterns. On MRI, they are most commonly appreciated on the sagittal
Choroid Plexus Tumors
images, where they cause pineal enlargement that frequently results in
a slight impression on the superior colliculi or posterior third ventricle Choroid plexus papillomas and carcinomas are rare tumors that
(Fig. 7-106A). Most pineal cysts are small (2–4 mm in diameter) and arise from the epithelium of the choroid plexus in both children and
are incidental findings (575,576). When imaged sequentially, they may
enlarge, shrink, or disappear entirely (576). If the MR appearance is
classic for pineal cyst, these changes should not cause concern. TABLE 7-11 Supratentorial
When larger than 1 cm, pineal cysts may cause some diagnostic Extraparenchymal Tumors
difficulty, especially when hemorrhagic or heterogeneous (577). An
important factor in the differentiation of cysts from neoplasms is Tumor Key Features
that cysts are rarely, if ever, associated with the Parinaud syndrome
and they are essentially never the cause of hydrocephalus (574). In Choroid plexus papilloma Intraventricular origin,
general, the MR appearance of pineal cysts is that of isointensity with lobulated, T2 hypointensity
CSF on T1-weighted images and slight hyperintensity with respect to Choroid plexus carcinoma Intraventricular origin, invasion
CSF on T2-weighted and FLAIR images. The hyperintensity on the of parenchyma, necrosis
T2-weighted sequences is probably the result of loss of phase coher- Tumors of the meninges Meningeal origin
ence of the protons in the moving CSF in the surrounding cisterns;
the phase coherence of the protons within the cysts results in a higher Infantile myofibromatosis Calvarial or dural origin
relative signal intensity. When hemorrhage is present in the cyst, high Sharply marginated
signal intensity is seen in the cyst on both T1- and T2-weighted images Isointense to gray matter
and the cyst may enlarge, resulting in symptoms from local mass effect. Lymphoma/leukemia Gray matter intensity on T2
After intravenous administration of paramagnetic contrast, a crescent Knowledge of systemic disease
or rim of normal pineal gland surrounding the cyst typically enhances
Langerhans cell histiocytosis Dural, skull, or choroid plexus
(Fig. 7-106B). The scan should not be delayed after contrast adminis-
origin
tration as, because no blood–brain barrier is present in the pineal, the
Marked T2 hypointensity
cyst itself may enhance after prolonged delays, making differentiation
from tumors impossible (578). The keys in making the diagnosis of Epidermoids and dermoids Same characteristics as in other
pineal cyst by MRI are the absence of clinical manifestations, normal locations
ventricular size, the characteristic location, the isointensity with CSF Arachnoid cysts (see Chapter 5) Sharp margins, isointense to CSF
on T1-weighted images, the absence of any internal structure within
the lesion, and the presence of the typical rim of enhancement around Calvarial tumors Centered in calvarium
the cyst.

Barkovich_Chap07.indd 744 5/6/2011 9:17:24 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 745

adults. They comprise approximately 5% of supratentorial tumors Anaplastic papillomas cannot be differentiated from carcinomas based
in children and less than 1% of all primary intracranial tumors. on gross pathologic features; the distinction is purely based on charac-
Choroid plexus tumors can occur at any age, but most are found in teristics of individual cells. Both aggressive papillomas and carcinomas
infants, or during the first 5 years of life. Papillomas of the choroid show a marked propensity to metastasize through the CSF pathways,
plexus are most often found in the first year of life; they are typically sometimes forming large subarachnoid masses (8,75,76).
discovered because of the resultant severe hydrocephalus. A marked
male predominance has been noted for papillomas (8,579–582), but Imaging Studies
carcinomas affect males and females more equally (583). Choroid Choroid plexus tumors are usually easy to identify and diagnose, as
plexus carcinomas make up 30% to 40% of choroid plexus tumors few pediatric tumors arise in the lateral ventricles. Although it is usu-
in children; affected children tend to present at a somewhat older ally easy to differentiate a choroid plexus papilloma from a carcinoma,
age than those with choroid plexus papillomas, but usually within it is important to remember that this differentiation is histologic, not
the first 3 to 5 years of life. Focal neurological deficits secondary to radiologic. Therefore, tumors with a more benign appearance may
local brain involvement by tumor are more common than in patients ultimately be diagnosed histologically as carcinomas and tumors with
with choroid plexus papillomas; however, most patients with choroid aggressive, invasive imaging characteristics may turn out to be anaplas-
plexus carcinomas still present with signs and symptoms of hydro- tic papillomas. Nonetheless, it is important to properly characterize the
cephalus (583). tumors preoperatively by neuroimaging, as this characterization may
affect surgical approach to the tumor.
Pathology On CT, choroid plexus papillomas typically appear as lobulated,
The most common site of origin of choroid plexus tumors in the pediat- isodense or mildly hyperdense intraventricular masses (Fig. 7-107) with
ric age group is the lateral ventricle, the left side being more commonly occasional punctate foci of calcification. The most common location
involved than the right; tumors are rarely bilateral (579,581). The third of the tumor is the trigone, but papillomas may be located anywhere
and fourth ventricles are less commonly involved in children, although along the choroid plexus, including the temporal frontal horns of the
the fourth ventricle is the most common site of origin of choroid plexus lateral ventricle, third ventricle, fourth ventricle, or lateral recess of the
tumors in adults. Exceptionally, tumors may originate from chor- fourth ventricle with extension into the cerebellopontine angle cis-
oid plexus in the cerebellopontine angle (584). Grossly, tumors of the tern. Homogeneous enhancement is seen after infusion of IV contrast.
choroid plexus are dark pink or red, globular masses with an irregular Although usually unilateral, bilateral papillomas have been reported
papillary surface resembling a cauliflower. They often cause consider- (579). Occasionally, papillomas grow through the ependyma and into
able enlargement of the involved lateral ventricle. An important point the adjacent white matter, inciting surrounding edema. These aggres-
is that choroid plexus papillomas are differentiated from carcinomas sive papillomas have a more irregular appearance, more closely resem-
based upon histological, not gross pathological, criteria. In general, bling a carcinoma; histologically, they show a higher degree of anaplasia
papillomas are well-marginated and separate from the surrounding than the less aggressive papillomas. Choroid plexus carcinomas usually
brain. Small areas of hemorrhage may be present and multiple gritty appear much more aggressive than papillomas. Carcinomas have irreg-
foci of calcification are frequently found. Some choroid plexus papil- ular contours, demonstrate mixed density prior to contrast enhance-
lomas show evidence of anaplasia and invasion of the adjacent brain. ment and have a prominent, but variable, enhancement pattern. Cysts

FIG. 7-107. Choroid plexus papilloma. A. Axial noncontrast CT images show hydrocephalus and a hyperdense, lobulated mass (arrows) in the left lateral
ventricle. Note surrounding vasogenic edema. B. Sagittal T2-weighted image shows that the very lobulated mass is extremely heterogeneous and has mul-
tiple small areas of hyperintense cysts/necrosis.

Barkovich_Chap07.indd 745 5/6/2011 9:17:25 PM


746 Pediatric Neuroimaging

FIG. 7-107. (Continued) C. Axial FLAIR image show the heterogeneous mass with sur-
rounding edema. The branching areas of central hypointensity are characteristic. Sur-
rounding vasogenic edema does not necessarily imply malignancy, but should inspire
a search for invasion of the cerebral hemispheres. D. Postcontrast axial T1-weighted
image shows robust enhancement of the solid portion of the tumor. Note the character-
istic central, rather low intensity, branches extending outward from the center. The pos-
terior fluid (f) is likely a tumor cyst rather than trapped area of ventricle in light of its
hyperintense signal on FLAIR (in C). E. Calculated diffusivity map shows similar diffu-
sion characteristics to normal cerebral parenchyma. This can help differentiation from
PNET and ATRT which have reduced diffusivity. F. Collapsed view from time of flight
MRA shows enlarged posterior choroidal branches (arrow), indicating a choroid plexus
origin of the tumor. G. Relaxivity curve from dynamic susceptibility-weighted perfu-
sion study shows no return to baseline of curves 1 and 3, which were placed over the
tumor. This indicated absence of blood–brain barrier and strongly suggests an extra-
parenchymal tumor. Contrast with parenchymal curve 2. H. Proton MR spectrum (TE =
135 milliseconds) shows only a choline peak (Ch); absence of NAA also suggests and
extraparenchymal tumor.

Barkovich_Chap07.indd 746 5/6/2011 9:17:25 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 747

and hemorrhage are often present within the tumor. Carcinomas ependymomas, or choroid plexus carcinomas. The central portions
nearly always grow through the ventricular wall into the brain and of papillomas are usually somewhat hypointense compared to gray
incite vasogenic edema (Fig. 7-108) (10,11,579,582,585,586). matter on T2-weighted and FLAIR images, often with a well-defined
The typical MR appearance of a choroid plexus papilloma branching pattern (Figs. 7-107D, E and 7-109B), a characteristic that
on T1-weighted sequences is that of a homogeneous, lobulated may aid in their preoperative diagnosis. Foci of hemorrhage or cal-
intraventricular mass (Fig. 7-107C). The surface of the mass typi- cium are occasionally present. Cysts or septations may develop within
cally has a papillary appearance that distinguishes it from the smooth the ventricle (Fig. 7-109C); these are likely the result of inflammation
surfaces of most other intraventricular tumors such as meningiomas, generated by the tumor mass, possibly as a result of recurrent small

FIG. 7-108. Choroid plexus carcinoma. A. Axial noncontrast CT shows a large, high attenuation mass involving the right lateral ventricle and the right
cerebral hemisphere. Foci of calcification (open arrows) and cystic or necrotic areas (closed arrows) are seen within the tumor mass. B. After infusion of
iodinated contrast, there is heterogeneous enhancement of the tumor mass. C and D. Different patient. Axial T1-weighted images show a heterogeneous
mass filling the left ventricular trigone. Foci of hemorrhage and necrosis are present.

Barkovich_Chap07.indd 747 5/6/2011 9:17:27 PM


748 Pediatric Neuroimaging

FIG. 7-108. (Continued) E and F. Axial T2-weighted images show the extremely heterogeneous tumor mass eliciting vasogenic edema (arrows) in adjacent
white matter. G and H. Postcontrast T1-weighted images show heterogeneous enhancement of the tumor, with both cystic and necrotic regions.

hemorrhages. Uniform, intense enhancement follows intravenous is not seen. When the surrounding brain parenchyma is extensively
administration of paramagnetic contrast (Figs. 7-107 and 7-109) invaded, it is sometimes difficult to determine whether the tumor origi-
(581,585). Third and fourth ventricular papillomas are more com- nated in the ventricle or in the parenchyma. Under these circumstances,
mon in adults but can develop in children (Fig. 7-109). They have an it is difficult to make the proper diagnosis. Carcinomas often have areas
appearance similar to that of lateral ventricular papillomas, lobulated of high and low signal intensity on both T1- and T2-weighted images,
intraventricular masses that calcify and cause hydrocephalus. Fourth resulting from hemorrhage and cyst formation. The frontal, occipital,
ventricular papillomas may originate within or grow through the or temporal horn of the affected lateral ventricle becomes encysted in
fourth ventricular outlet foramina, sometimes presenting as a cerebel- more than 80% of patients (37,580). Hydrocephalus is present in more
lopontine angle or cerebellomedullary angle mass (Fig. 7-110). than 80% of patients at the time of diagnosis and leptomeningeal dis-
Choroid plexus carcinomas are typically heterogeneous in appear- semination of tumor is present at the time of diagnosis in nearly half
ance, particularly on T2-weighted images, and usually invade the sur- of affected patients (587).
rounding brain through the ventricular wall, inciting vasogenic edema As with all intraventricular tumors, choroid plexus tumors (papil-
(Fig. 7-108) (587). The well-defined branching pattern of the stroma lomas and carcinomas) can spread throughout the neuroaxis via the

Barkovich_Chap07.indd 748 5/6/2011 9:17:28 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 749

FIG. 7-109. Choroid plexus papilloma of the fourth ventricle.


A. Sagittal T1-weighted image shows a fourth ventricular mass
(large arrows) with associated cysts. The mass is somewhat
heterogeneous and extends inferiorly (small arrows) through the
foramen of Magendie. B. Axial T2-weighted image shows
the mass (arrows) to be heterogeneous, with some areas of very
low signal intensity and other areas (probably cysts) appear-
ing very hyperintense. C. Postcontrast coronal T1-weighted
image shows the tumor mass (large arrows) extending inferiorly
through the foramen of Magendie (small arrows). One of the
walls of the tumor-associated cysts is seen (arrowheads).

FIG. 7-110. Fourth ventricular choroid plexus papilloma extending


through foramen of Luschka. A and B. Postcontrast coronal T1-weighted
images show the lobulated enhancing tumor (arrows) extending out the
left foramen of Luschka into the cerebellomedullary angle cistern and into
the cervical subarachnoid space.

Barkovich_Chap07.indd 749 5/6/2011 9:17:29 PM


750 Pediatric Neuroimaging

cerebrospinal pathways (588). Therefore, all affected patients should sarcomas (594), extraosseous Ewing sarcomas (596), malignant fibrous
have a contrast-enhanced scan of the brain and spine with careful histiocytomas (597), and meningeal melanomas (591,598). In addition
attention for small tumors of the surface of the brain and spinal cord, to their rarity, several features characterize meningiomas in children.
and along cranial and spinal nerves. A higher percentage of meningiomas in the pediatric age group are
Proton MR spectroscopy of choroid plexus papillomas is remark- intraventricular (590–592). Cystic areas are commonly associated with
able for the complete absence of an NAA or creatine/phosphocreatine pediatric meningiomas, being found in more than 12% of affected
peak. Only a choline peak is seen on long TE spectra (Fig. 7-107H), patients (599–601). Meningiomas in childhood sometimes originate
but a very prominent peak of myo-inositol is seen on short TE (45). in the Sylvian fissure and lack a dural attachment, a characteristic that
Carcinomas have higher choline levels and show elevation of lactate may imply origin from heterotopic meningeal rests within the brain
(589), but myo-inositol is not increased (45). Perfusion imaging of the (602,603). Pediatric meningiomas are often very large, grow rapidly,
tumor shows high blood volume with persistence of contrast within and are more frequently malignant than adult meningiomas; in several
the tumor interstitium due to absence of blood–brain barrier within series more than 50% of pediatric meningiomas were malignant
the mass, resulting in no return of the perfusion curve to baseline. (8,76,590,592,604). A high mortality rate and high incidence of sar-
comatous degeneration have been reported (76,590,592). Nonetheless,
Differential Diagnosis outcome is usually good if the tumor is completely resected and if the
Choroid plexus papillomas are easily diagnosed on both CT and MRI patient does not have NF2 (593,602,605).
by their characteristic location and appearance. The diagnosis of chor- Extraosseous Ewing sarcomas are very rare tumors that are histo-
oid plexus carcinoma is more difficult because of their resemblance to logically identical to PNETs but are distinguished by their dural origin/
PNETs, ATRT, and ependymomas. attachment, by strong membrane expression of the MIC-2 gene prod-
uct (CD99), and by demonstration of the chromosomal translocation
Tumors of the Meninges t(11,22)(q24;q12) (606).
Intrinsic meningeal tumors are uncommon in childhood, comprising
only 1% to 2% of primary brain tumors. In contrast to adults, in whom Imaging Studies
a female predominance exists, males and females seem to be equally Meningiomas, plasma cell granulomas, meningeal fibromas and
affected in the pediatric age group. No definite age peak is noted myofibromas, and meningeal sarcomas have nearly identical imag-
within the pediatric population. The clinical presentation is variable ing features; they can only be differentiated by histologic examination
and depends upon the site of the tumor; headaches and focal neuro- (220,591,594,607). The diagnosis of malignant fibrous histiocytoma
logical deficits are the most common reasons for initial presentation may be suggested if the child presents with hemorrhage into the tumor
(590–594). The presence of a meningioma in a child should raise sus- (608). CT studies have revealed a heterogeneous appearance of these
picion for neurofibromatosis type 2 (see Chapter 6) and should initiate tumors. Approximately one half of patients with meningiomas have
a search for other meningiomas or schwannomas. associated hyperostosis and half have intratumoral calcification. All
pediatric meningeal-based tumors are isodense to hyperdense on pre-
Pathology contrast CT and the solid portions of the tumor show diffuse contrast
Meningeal tumors of childhood include meningiomas (which are, enhancement (Fig. 7-111). Cystic and necrotic changes are more fre-
by far, the most common), plasma cell granulomas (discussed ear- quent in pediatric patients than in adults (599–601), possibly because
lier in this chapter in the section on “Tumors of the Cerebral Hemi- of the larger size at the time of diagnosis. Rapid growth or the pres-
spheres”), meningeal fibromas and myofibromas (595), meningeal ence of atypical CT features, such as hemorrhage, heterogeneous

FIG. 7-111. Meningioma. A. Axial con-


trast-enhanced CT scan shows a mass
within the atrium of the left lateral ven-
tricle. The periphery (white arrows) is
enhancing whereas the central portion
(open black arrows) is densely calcified.
B. Axial T2-weighted image shows the
meningioma (closed white arrows) in the
atrium of the left lateral ventricle. The
densely calcified center (open arrow) is of
very low signal intensity.

Barkovich_Chap07.indd 750 5/6/2011 9:17:31 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 751

enhancement, cyst formation, or poorly defined margins, suggests a of dural-based masses in neonates and infants, an age group in which
malignant tumor such as meningeal sarcoma, extraosseous Ewing sar- meningiomas are extremely rare.
coma, or meningeal PNET (594,596); however, benign meningiomas
can have many of these features. Intraventricular meningiomas arise Leukemia and Lymphoma
from the choroid plexuses and have the same CT characteristics as Tumor deposits in the brain and meninges from systemic malignan-
those originating in the dura (590,591,609). cies are rare in childhood. The exceptions to this rule are childhood
On MRI, pediatric meningeal tumors are generally large masses leukemia and lymphoma. Leukemias account for 30% of all childhood
that arise from the dura, the choroid plexuses, or from ectopic dural malignancies and are the most common cancers diagnosed in children
rests in the Sylvian fissures. They typically have smooth, well-defined (614). Involvement of the CNS may be asymptomatic or may manifest
margins (whether benign or malignant) and are isointense to gray mat- with irritability, headaches, vomiting, seizures, or unusual weight gain.
ter on T1-weighted sequences and isointense to hyperintense to gray In advanced cases, papilledema or cranial neuropathies (typically uni-
matter on FLAIR and T2-weighted sequences. Cystic and necrotic areas, lateral involvement of cranial nerve VII) will be detected (614). The
if present, are hypointense on T1- and hyperintense on T2-weighted most common finding in leukemia is enlargement of the ventricles
images compared to the solid portion of the tumor, which shows uni- and sulci. Although some authors have suggested that the enlarged
form enhancement after intravenous infusion of paramagnetic con- CSF spaces result from radiotherapy and chemotherapy, Kretchmer et
trast. Hemorrhage, heterogeneity, reduced diffusion (compared with al. (615) found that 31% of children with acute lymphocytic leuke-
gray matter), and indistinct margins favor sarcomas or meningiomas mia have enlarged ventricles before treatment. These findings suggest
with higher degrees of malignancy (596), but this feature is not reliable. that the ventricular enlargement represents hydrocephalus, not atro-
Densely calcified areas have very low intensity, whereas areas that are phy, and is caused by the disease, not by the treatment. Meningeal and
less densely calcified may appear isointense or hyperintense on non- parenchymal metastatic disease is far less common than enlargement
contrast T1-weighted images (Fig. 7-111B). of the ventricles and sulci. Children with acute myelogeneous leuke-
As stated in the section on imaging characteristics at the beginning mia, may present with focal signs or symptoms due to the develop-
of this chapter, perfusion imaging may be useful to differentiate men- ment of chloromas, aggregates of leukemia cells that often occur in the
ingeal tumors from intraparenchymal tumors in that neuroepithelial calvarium, neck, or spine.
tumors show a return to baseline of relaxivity after the bolus of con- On CT, only 5% of leukemic patients with CNS symptoms
trast has passed through the tumor, but meningeal tumors show per- show contrast-enhancement of the meninges (10). When lympho-
sistence of abnormal relaxivity due to absence of blood–brain barrier. mas or aggregates of leukemic cells involve the brain, they appear
In addition, meningiomas tend to have large blood volumes compared on CT as masses of isodense to hyperdense signal prior to contrast
to normal brain parenchyma (54). enhancement; they enhance uniformly after contrast administration
Long echo time proton MR spectroscopy in meningiomas shows (Fig. 7-112) (614). Of interest, leukemic infiltrates may grow through
the normal neoplastic pattern of elevated choline with diminished the calvarium or an orbital wall without any apparent radiologic evi-
creatine/phosphocreatine and NAA. Short echo time spectra show an dence of bone involvement (Fig. 7-112). On MRI, these masses tend to
increase in alanine (seen as a doublet centered at 1.47 ppm), choline, be slightly hypointense with respect to brain on T1-weighted sequences
and glutamine/glutamate, whereas concentrations of myo-inositol, and isointense to slightly hyperintense on FLAIR and T2-weighted
creatine/phosphocreatine, and NAA are low (610–612). sequences. Almost all lesions show diffuse and prominent enhance-
Catheter arteriography typically shows a characteristic homoge- ment after infusion of IV contrast (Fig. 7-113). There are no character-
neous tumor blush, along with vascular supply from meningeal vessels. istic locations within the brain, and, indeed, the disease can affect the
Surgery may be aided in such cases by preoperative embolization. The cerebral parenchyma, meninges, cranial nerves, calvarium, or orbits
reader should keep in mind, however, that meningeal tumors some- (Figs. 7-112 and 7-113). Indeed, when the disease is predominantly or
times parasitize blood flow from intracerebral vessels and may, thus, entirely subarachnoid in location, the only imaging abnormality may
incorrectly suggest a primary intracerebral tumor. Similarly, intrac- be enhancement of cranial nerves (usually resulting from deposition of
erebral tumors can parasitize flow from meningeal vessels to mimic subarachnoid tumor on the nerves) on postcontrast scans. Equally or
meningeal tumors. more important, and much more frequent than direct involvement of
the disease is the effect of therapy and immunosuppression. Infection
Infantile Myofibromatosis (see Chapter 11), acute neurotoxicity (see Chapter 3), demyelination
Infantile myofibromatosis is a condition in which mesenchymal pseudo- (see Chapter 3), hemorrhage and infarction (see Chapter 4) are all-
tumors develop in the skin, subcutaneous tissues, skeletal muscle, bone, too-common sequelae of the treatment of leukemia and lymphoma
and/or visceral organs of children (595). When infantile myofibromas (614,616,617). The diagnosis of these treatment-related changes can
involve the skull and brain, they most commonly arise from the calva- sometimes be aided by the use of volumetrics, morphometry, perfu-
rium or dura (595). By radiologic criteria, these masses are very dif- sion imaging, and DWI (618).
ficult to differentiate from meningiomas when they are dural based
and from LCH when they arise within the calvarium. Infantile myo- Langerhans Cell Histiocytosis
fibromas appear on CT and MRI as well-demarcated, homogeneous LCH has been mentioned in nearly every section of this chapter. The
calvarial or dural masses that are isodense to gray matter on CT and reason for the multiple citations is the fact that LCH can arise nearly
isointense to hypointense to gray matter on T1-and T2-weighted MR anywhere in the brain and skull. This diagnosis should always be con-
sequences. If the calvarium is involved, osteolytic or sclerotic changes sidered when a mass arises from the bones of the face, skull base, or
may be identified on plain films or CT. Homogeneous enhancement calvarium in children. Rarely, LCH may result in masses based in the
is typical after administration of intravenous contrast; if the lesion is cerebral parenchyma, spinal cord, pineal gland (506), dura, or choroid
dural-based, a “dural tail” may be present (595,613). Because the imag- plexus (504).
ing characteristics of dural-based infantile myofibromas are nearly The most common imaging finding of calvarial LCH is that of
identical to those of meningiomas, differentiation is not possible by a well-defined mass involving the calvarium. On CT, a sharply cir-
imaging alone. Myofibroma should be considered as a possible cause cumscribed skull lesion is identified with differential involvement

Barkovich_Chap07.indd 751 5/6/2011 9:17:31 PM


752 Pediatric Neuroimaging

FIG. 7-112. Leukemic infiltrate (chloroma) involving the


orbit and frontal lobe. A. Axial postcontrast CT image shows
a mass (arrows) in the anteromedial right orbit, displacing
the ocular globe posteriorly and laterally. No bone destruc-
tion is present. B. Image at a slightly higher level shows the
mass (arrows) extending subcutaneously. C. At a higher level,
a large mass is seen in the right frontal lobe, resulting in sub-
stantial mass effect with subfalcine herniation (arrows) of
the right frontal lobe.

of the inner and outer tables (resulting in the classic “beveled edge” Lesions originating within the dura and choroid plexus tend to
on plain skull films). The lesion is typically of similar attenuation to be ovoid masses that are often quite large at the time first detected by
cortical gray matter; uniform, moderately intense enhancement is imaging (246). Histologically, these lesions are granulomas consisting
seen after administration of contrast (Fig. 7-114A and B). MRI shows of a mixture of histiocytes, foamy macrophages, multinucleated giant
the bone lesions as sharply defined soft tissue masses that have sig- cells, lymphocytes, plasma cells, and eosinophils (250). On imaging,
nal intensity comparable to skeletal muscle and enhance markedly the masses are characterized by isointensity to cerebral gray matter
after IV administration of paramagnetic contrast (Fig. 7-114C and D) on CT and a marked hypointensity on FLAIR and T2-weighted MR
(220,241,243,244,246,249,504). The lesion may occasionally displace images (Fig. 7-115). The reason for this marked T2 hypointensity is not
the underlying brain. known but it may be related to a severe fibrotic reaction of the choroid

Barkovich_Chap07.indd 752 5/6/2011 9:17:32 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 753

FIG. 7-113. Leukemic infiltrate involving the trigeminal nerve. Fat-suppressed postcontrast axial
T1-weighted image shows enhancement of the cisternal portion of the right trigeminal nerve
(arrowheads). Enhancement is also seen within the trigeminal cistern (arrow), which surrounds the
trigeminal ganglion.

FIG. 7-114. Langerhans cell his-


tiocytosis involving the calvarium.
A. Axial CT image using wide win-
dows for optimal evaluation of bone
shows a sharply marginated lytic area
(arrows) in the posterior left frontal
bone. B. Axial contrast-enhanced CT
image shows enhancing soft tissue
(arrows) within the calvarial defect. C.
Axial T2-weighted image of a different
patient shows a well-defined focus of
T2 prolongation (arrows) involving
the right frontal bone. D. Postcontrast
T1-weighted image reveals that the soft
tissue intensity mass (arrows) shows
uniform enhancement.

Barkovich_Chap07.indd 753 5/6/2011 9:17:32 PM


754 Pediatric Neuroimaging

FIG. 7-115. Langerhans cell histiocytosis of the choroid plexus and tentorium. A. Axial FLAIR image shows heterogeneous isointense and hypointense
masses (black arrows) in the temporal horns of the lateral ventricles. The well-defined hypointense regions (t) anterior to the masses are the trapped tem-
poral horns of the lateral ventricles. The tentorial mass (white arrowheads) is equally heterogeneous, with areas of isointensity and areas markedly hypoin-
tense compared with brain tissue. B. Postcontrast axial T1-weighted image shows somewhat heterogeneous enhancement of both the choroid plexus (black
arrows) and tentorial (white arrowheads) masses. (This case courtesy of Dr. Danielle Prayer, Vienna.)

or meningeal tissue to the LCH involvement. These lesions enhance administration of iodinated contrast. On MRI, the lesions are typically
intensely and uniformly after administration of intravenous contrast slightly hyperintense co cerebral cortex and T1-weighted images and
(Fig. 7-115) (505). The choroid plexus and dural lesions are rarely the isointense to hypointense on T2-weighted images. Enhancement is dif-
cause of clinical presentation. They are more commonly found inci- fuse, but heterogeneous (622). Nuclear medicine studies can be useful
dentally in children with known multisystemic LCH, conditions pre- to confirm the diagnosis, as sulfur colloid is taken up by macrophages
viously known as Hand-Schüller-Christian and Letterer-Siwe diseases and, thus, traces the reticuloendothelial system (623). Uptake of 99mTc
(241,246,504). sulfur colloid within the masses, therefore, strongly supports the diag-
nosis of extramedullary hematopoiesis.
Extramedullary Hematopoiesis
Extramedullary hematopoiesis refers to the development of hematopoi- Calvarial Tumors
etic tissue (blood cell development) outside of the bone marrow. It is Tumors arising from the calvarium were discussed in the “Posterior
a compensatory mechanism by which the body attempts to maintain a Fossa Tumors” section of this chapter and in Chapter 5, under the
level of erythrogenesis sufficient for its demands in the setting of chronic section entitled “Fontanel Dermoids and Other Calvarial Masses of
hemolytic anemias, hemoglobinopathies, and some myeloproliferative Childhood.” Calvarial masses from LCH were discussed in the previ-
syndromes. The most common locations for this phenomenon are the ous section.
liver, spleen, paravertebral and presacral spaces, kidneys, adrenals, lung, Neuroblastoma metastases should also be mentioned again in this
breast, thyroid, maxillary antra, and the falx cerebri (619). Rarely, it can section, because patients with neuroblastoma sometimes present with
occur in the epidural space of the head or spine (620), possible due to a calvarial mass. Neuroblastomas are common tumors of childhood
transformation of multipotential cells into marrow or direct extension that can involve the CNS either by direct extension (primarily in the
of hematopoietic cells from marrow into the epidural space (620,621). spine, see Chapter 10) or, more commonly, by blood-borne metastases.
Clinically, extramedullary hematopoiesis is usually asymptomatic, but Neuroblastoma metastases rarely involve the brain; instead, they char-
neurologic signs and symptoms can result from compression of adja- acteristically involve the orbit and calvarium. On CT, calvarial metas-
cent tissue in the brain or spine (see Chapter 10 for discussion of spinal tases from neuroblastoma show a characteristic appearance of a mass
manifestations). Headaches, seizures, hemiplegia, altered conscious- (or masses) growing inward from the bone with spicules of bone
ness, and cranial neuropathies are the most common manifestations of radiating centripetally within it (Fig. 7-116). Calvarial metastases can
intracranial disease (622). grow centripetally into the intracranial space and mimic intracranial
Imaging studies typically show multiple intracranial dural-based masses. Coronal images help to verify a dural or extradural origin of
masses, often with associated enlargement of the diploic space of the these lesions. On MRI, the metastases appear as soft tissue intensity
overlying calvarium. The masses are typically isodense to cerebral cortex masses originating within the calvarium (Fig. 7-117). They enhance
on noncontrast CT and show uniform enhancement after intravenous markedly, on both CT and MRI, after administration of contrast.

Barkovich_Chap07.indd 754 5/6/2011 9:17:34 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 755

FIG. 7-116. Metastatic neuroblastoma to calvarium. A. Axial noncontrast CT scan shows multiple intracranial hyperdense masses (arrows), some of
which appear to be intraparenchymal in origin. Bony spicules extend intracranially from the calvarium. B. After intravenous contrast infusion, the masses
heterogeneously enhance. C. Bone windows show the characteristic bony spicules radiating centripetally from the calvarium. Also notice the splitting of the
coronal and sagittal sutures (arrows), another characteristic finding in metastatic neuroblastoma.

FIG. 7-117. Metastatic neuroblastoma


to calvarium and dura. A. Axial contrast
enhanced CT scan shows calvarial metas-
tases with intracranial extension (black
arrows) in the right frontal, right pari-
etal, and midline posterior regions. Note
the displacement of the superior sagittal
sinus from the skull. B and C. Axial T2
(B) and postcontrast T1 weighted (C)
images show extensive extraparenchymal
tumor (arrows) posteriorly. D. Coronal
postcontrast T1 weighted image shows
the involvement of the calvarium and the
heterogeneity of the metastases (arrows).

Barkovich_Chap07.indd 755 5/6/2011 9:17:34 PM


756 Pediatric Neuroimaging

The elevated periosteum enhances, as well, which may be a helpful sign in the first few months after birth (Fig. 7-118) and should be consid-
in differentiating metastatic tumor from the normal hematopoietic ered congenital tumors, as their incidence of presentation drops off
marrow of childhood. steadily thereafter. Congenital tumors are usually detected because of an
enlarged head circumference or asymmetric skull growth; the anterior
fontanelle is often tense. Among congenital tumors, which represent
BRAIN TUMORS IN THE FIRST less than 2% of all childhood brain tumors, teratomas constitute 30% to
YEAR OF LIFE 50%; the remainder most often consist of astrocytomas, choroid plexus
The histologic distribution of primary brain tumors in the first year of papillomas, embryonic tumors (such as ATRT, PNET, and medullo-
life (Table 7-12) is different than the distribution of the entire pediatric blastoma), ependymomas, and craniopharyngiomas (630,634). Many
period. Although various groups report different incidences of tumors congenital tumors are now detected prenatally by ultrasound and MRI
in infants (10,13–15,624–631) and nearly any histologic type of tumor, (635). It is not surprising that the histologies of these fetal tumors are
including glioblastoma, can occur in neonates and infants (632,633), similar to those found in neonates, with teratomas being most com-
certain definite trends emerge. Supratentorial tumors predominate, mon. Astrocytomas, craniopharyngiomas, embryonal tumors (ATRTs,
constituting 60% to 70% in most series. Suprasellar lesions seem to PNETs, medulloblastomas), choroid plexus tumors, and ependymo-
be more common during the first year of life than during the pediatric mas are also found (635).
age group as a whole. Of these, the most common are suprasellar astro- The clinical manifestations of tumors presenting in the first year
cytomas (usually grade II or grade III) arising in the hypothalamus. of life differ from those of older children (15,625,627–631). Focal neu-
Choroid plexus papillomas have a much higher relative incidence in rological deficits are almost always absent because of the immaturity
the first year of life, representing 15% to 20% of all tumors in this age of the CNS. Infants with infratentorial tumors tend to present with
group. vomiting and increased sleepiness. Increasing head size and develop-
Among posterior fossa tumors, medulloblastomas (although mental delays are noted in more than one-half of affected patients.
known to peak in frequency toward the latter half of the first decade) Among infants with supratentorial tumors, an increasing head size is
are reported to be relatively common among tumors presenting during seen in about one half, vomiting and lethargy or irritability develop
the first year of life. However, the recent identification of ATRT as a dis- in approximately one third, and seizures are seen in approximately
tinct group raises the strong possibility that many of the posterior fossa 20%. Those infants with hypothalamic/chiasmal gliomas typically
tumors of infancy previously identified as medulloblastomas were, in present with decreased appetite, poor weight gain and abnormal eye
fact, ATRT (118). Therefore, it is likely that ATRT, not medulloblasto- movements.
mas, are common in the first year of life. Posterior fossa ependymomas Treatment of very young children also differs from those in the
also seem relatively more common in younger children, whereas cer- older age group (15,625,628–631). The treatment of children under
ebellar and brainstem astrocytomas are uncommon. the age of 3 years is compromised by the immaturity of the brain,
Among supratentorial tumors, PNET are reported in some series the highly malignant nature of the neoplasms, the delay in diagnosis
to be common within the first year of life, with an incidence nearly (because of a low index of suspicion and nonspecific clinical find-
equal to that of choroid plexus papillomas. However, some of these ings), the large bulk of tumor found at presentation, and the reduced
PNETs may have, in fact, been ATRT because both tumors are com- radiation dose used in this age group. A reduced radiation dosage is
posed of sheets of small round cells. The diagnoses of PNET, atypi- necessary because of the immaturity of the brain and the high inci-
cal teratoid/rhabdoid tumor, and ependymoma should be considered dence of subsequent vaso-occlusive disease and developmental retar-
when intraparenchymal hemispheric tumors are discovered in infancy; dation that occurs when radiation is given to very young children;
choroid plexus tumor should be a strong consideration if the tumor is most oncologists try not to use radiation at all in this age group. In
intraventricular. general, a conservative approach is recommended in children with
In other series, teratomas are the most frequent brain tumor diag- optic gliomas and low grade supratentorial astrocytomas. Radiation is
nosed in the first year of life. Teratomas are diagnosed most frequently deferred until the child is 3 to 4 years old, when its effects on the CNS
are better tolerated; when necessary, modified focal irradiation is used.
Presently, most centers are using surgery, postoperative chemotherapy
(systemic and intrathecal), and delayed radiation; results have been
mixed (636–639).
TABLE 7-12 Most Common Brain
Tumors in the First Year
of Life RADIATION-INDUCED TUMORS OF THE
CENTRAL NERVOUS SYSTEM
Teratoma One of the unfortunate side effects of radiation therapy for brain
Suprasellar/thalamic astrocytoma tumors is the development of secondary neoplasms. As the long-term
survival of children with brain tumors increases, so does, unfortu-
Atypical teratoid/rhabdoid tumor nately, the incidence of secondary tumors. Secondary brain tumors in
Ependymoma children differ from those in adults in several ways. Most important,
Choroid plexus tumor the latency period appears to be shorter. In adults, the time between
treatment of the primary tumor and detection of the secondary tumor
Craniopharyngioma averages 20 to 25 years, whereas in children it averages 8 years (640). The
Medulloblastoma? second major difference is in the histology of the tumors. Although the
most common secondary tumor in adults is meningioma, malignant
Primitive neuroectodermal tumor? gliomas are, by far, the most common secondary tumor in childhood
(640). Leukemia is another common complication of brain tumor

Barkovich_Chap07.indd 756 5/6/2011 9:17:36 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 757

FIG. 7-118. Congenital tumors A and B. Teratoma in a


newborn. Sagittal T1-weighted image (A) shows an enor-
mous, heterogeneous mass involving the frontal, temporal,
and parietal lobes in this newborn infant. There is marked
hydrocephalus. The brainstem is pushed posteriorly and
inferiorly (open arrows) and is kinked at the cervicomedul-
lary junction (closed arrow). Coronal T1-weighted image (B)
shows markedly dilated temporal horns (arrows) in addition
to the huge, extremely heterogeneous tumor occupying the
majority of the intracranial space. (This case courtesy of
Dr. T. Ito.) C–E. Fetal teratoma. Fetal sonogram (C) shows
a transverse view of the brain containing a large echogenic
mass (m) that seems to be centered in the midline. Sagittal
fetal MR view (D) through the brain shows the large midline
heterogeneous mass that seems to be eroding and enlarging
the sella turcica (arrows) suggesting possible origin from
the pituitary gland or stalk. Coronal view of the brain (E)
shows the suprasellar mass lifting and displacing both cere-
bral hemispheres. The margins of the tumor appear sharp,
suggesting displacement rather than invasion of the brain.
Pathology showed craniopharyngioma.

Barkovich_Chap07.indd 757 5/6/2011 9:17:36 PM


758 Pediatric Neuroimaging

irradiation, with an average latency period of about 10 years. Other nonetheless, patients with RB1 germline mutations are predisposed to
common solid secondary tumors include meningiomas, sarcomas, low the development of additional tumors elsewhere in the body. Whereas
grade gliomas, and PNETs (640). secondary sarcomas occur much more commonly in irradiated patients
(in the radiation field) in the early teenage years, melanomas, carcino-
mas, leukemias, and lymphomas tend to occur in young adults (average
TUMORS OF THE HEAD AND NECK IN age 27–29 years), often outside of the radiation field and in patients
CHILDHOOD who did not undergo radiation treatments (645). The deformities of
the face that often occurred secondary to varying degrees of growth
Masses in the head, neck, and orbit are common indications for imag-
arrest of the maxillary, nasal, and temporal bones following radiation
ing in childhood. It is very important, when possible, to differentiate
therapy (644,646,647) are much less commonly seen.
malignant tumors from developmental and benign masses. The topics
In 2% to 7% of patients with bilateral retinoblastomas, a small cell
of calvarial and skull base masses have been addressed earlier in this
tumor will eventually develop intracranially (typically in the midline);
chapter and in Chapter 5. In this section, I will concentrate on masses
this condition is referred to as “trilateral retinoblastoma.” Patients with
of the ocular globe, orbit, face, and neck.
trilateral retinoblastoma present with ocular tumor at an earlier age
(mean 6 months) than those with sporadic or unilateral tumors (648–
Ocular Tumors 650). The incidence of trilateral retinoblastoma is higher in children
with a family history of retinoblastoma than in those with no family
Retinoblastoma history (651). The intracranial tumor, which is histologically identical
Retinoblastoma is the most common intraocular malignancy in child- to the intraocular tumors, may develop in the pineal region (classically
hood, accounting for 11% of all cancers in the first year of life (641). the most common), suprasellar region, or fourth ventricle (rare) (650–
It most often presents in early childhood with leukocoria or “cat’s eye 654). It is rarely present at the time the ocular tumors are detected,
reflex.” Other conditions resulting in this presentation are listed in usually appearing after a latency period of about 2 to 3 years (648,649).
Table 7-13. Retinoblastoma is primarily a tumor of infancy; 70% to Prognosis of trilateral retinoblastoma is very poor, and the incidence
80% of retinoblastomas occur in infants less than 2-years-old. There of subarachnoid tumor spread is high (648,651,655). Recent work sug-
is no gender or race predilection (641). Multifocal tumors are com- gests that neoadjuvant intravenous chemotherapy might reduce the
mon, with a 30% incidence of bilateral tumors and a 30% incidence of incidence of this intracranial complication (656).
multifocal retinal tumors in affected children (642,643); the incidence A special mention must be made of the diffuse infiltrating retino-
of bilaterality is higher in patients younger than 1 year old at presenta- blastoma (DIR), a clinically and pathologically distinct variant that
tion (641). comprises 1% to 2% of retinoblastomas (657–660). Both clinically
Heritable retinoblastoma accounts for 30% to 60% of cases; 10% and radiologically, diagnosis of diffuse retinoblastoma is more diffi-
to 30% of these are familial and the remainder are caused by sporadic cult than that of the more common nodular type because the tumor
germline mutations. Both forms are transmitted to offspring in an assumes a relatively flat, ill-defined horizontal growth along the retinal
autosomal dominant manner with greater than 90% penetrance (641). tissue; due to the lack of vertical growth, examination shows little defi-
Most patients with retinoblastomas have a mutation on the RB1 gene at nition of a mass (660). Patients are typically older (average of ∼6 years)
chromosome 13q14; this is a tumor suppressor gene that controls cell- and present with pain, redness, or decreased vision in the affected eye;
cycle progression. More than 200 distinct mutations of this gene have glaucoma can result from neoplastic seeding of the anterior chamber,
been identified. Formerly, about 15% of patients with bilateral retino- causing obstruction of flow of the aqueous humor (660). Leukocoria
blastomas ultimately developed nonocular neoplasms, most commonly is relatively uncommon (24%). Ophthalmologic examination typi-
soft tissue sarcomas (644). The decreased utilization of radiation treat- cally shows a whitish, exudative-appearing, and flocculent material in
ment has resulted in a marked decrease in the incidence of sarcomas; the vitreous or anterior chamber, often associated with hemorrhage.

TABLE 7-13 Causes of Leukocoria in Children


Frequency
Condition Leukocoria Age Calcium CT Density Enhancement Size of Eye
Retinoblastoma 58% <12 mo (familial) ++ Very high ++ Normal
>20 mo (nonfamilial)
Persistent hyperplastic 28% Birth – High ++ Small
primary vitreous
Toxocaris infection 16% Childhood – High + Normal
Coats disease 16% Childhood – Very high – Normal to slightly small
Retinopathy of prematu- 5% Infancy ± High +/– Small (bilateral)
rity
Retinal astrocytoma 3% Childhood ± High + Normal
Medulloepithelioma <1% Childhood ± High ++ Normal

Barkovich_Chap07.indd 758 5/6/2011 9:17:38 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 759

FIG. 7-119. Bilateral retinoblastomas. A. Axial noncontrast CT


through the globes shows calcified masses arising from the retina bilat-
erally (arrows). Two discrete masses are seen in the left globe. There is
no evidence of extension of tumor outside of the globes. The pineal
region was spared in this patient. B. Axial 3D T2-weighted image shows
the masses as regions of hypointensity contrasted by the hyperintense
vitreous. C. Axial fat-suppressed postcontrast T1-weighted image shows
the mass in the left globe (arrow) as an enhancing nodular thickening.
The mass in the right globe is not well seen because of high signal in the
vitreous resulting from hemorrhage and proteinaceous exudate.

The appearance can be mistaken for uveitis, unexplained vitreous for children with suspected intracranial spread of tumor and those with
hemorrhage, hyphema, or endopthalmitis (660). No family history can bilateral retinoblastoma should be imaged by MRI, with specific atten-
be elicited in the vast majority of affected patients. tion to the suprasellar, pineal, and fourth ventricle regions. In addition,
follow-up imaging studies, which are intended to identify tumor along
Neuroimaging Findings the optic nerves or intracranially, should be MR scans of the orbit and
The usual CT appearance of retinoblastoma is that of a calcified mass brain. The orbital studies are best performed using precontrast volu-
within the globe. The mass, which may be of any size, arises from the metric fast spin-echo sequences with thin (<1 mm), overlapping par-
retina, but the borders may be ill-defined, making the retinal origin titions and 2 to 3 mm postcontrast axial and coronal fat-suppressed
unclear. Calcification, seen in 95% of these tumors, may be large, small, T1-weighted spin-echo images.
single, or multiple (Fig. 7-119). Enhancement is usually seen after con- The parenchymal tumor of trilateral retinoblastoma has CT and
trast administration. The tumor may extend through the sclera into MR characteristics identical to that of the intraocular tumor. CT shows
the orbital lymphatics or through the optic nerve intracranially; both a hyperdense mass that is usually heavily calcified and shows promi-
pathways of extension are well visualized by CT. Although retinoblas- nent heterogeneous enhancement (663). MRI shows a mass with T1
tomas can be identified by MRI (Fig. 7-119), the presence of calcifi- and T2 relaxation times similar to normal gray matter. The presence
cation is the most important radiological factor in the diagnosis of of calcification may cause the mass to appear heterogeneous. Hetero-
these tumors. As modern CT scanners often allow imaging without the geneous enhancement is seen after administration of paramagnetic
need for sedation, CT should be the primary imaging modality used contrast (652,653,663).
in patients presenting with leukocoria (642,661). However, MRI (with The appearance of DIR is different, as this form is characterized
fat saturation and contrast enhancement) shows the tumor well as an by diffuse infiltration of the various layers of the retina. Vitreous inva-
enhancing heterogeneous intraocular mass (Fig. 7-119). In addition, sion is frequent, and extension to the anterior chamber can occur via
MRI is more sensitive to spread of tumor along the optic nerve (Fig. the ciliary processes and the iris (664). No discrete nodular retinal
7-120) (seen as enhancement or enlargement along the affected nerve) mass, as typically seen in retinoblastoma, is present; instead, a dif-
(662) and to the presence of subarachnoid intracranial tumor. There- fuse, homogeneous, infiltrative uniformly enhancing mass is detected
fore, MRI is an excellent modality for the evaluation of patients with (Fig. 7-121); localized enhancement of the anterior chamber may
suspected retinoblastoma. Moreover, MRI should be the study of choice mimic medulloepithelioma (664). Calcification may be too minimal to

Barkovich_Chap07.indd 759 5/6/2011 9:17:38 PM


760 Pediatric Neuroimaging

FIG. 7-120. Bilateral retinoblastomas with spread of tumor through the optic nerve. A. Axial T1-weighted image shows retinal detachments in both globes
and enlargement of the left globe. The tumor in the left globe can be seen as a focal hypointensity (arrows) lateral to the optic nerve head. B. Postcontrast
T1-weighted image with fat suppression shows the enhancing tumor in the left globe extending (arrows) posteriorly into the optic nerve.

detect by ocular ultrasound or CT (Fig. 7-121). Typically, only a single tion is present, the abnormality cannot be radiologically differentiated
eye is involved. These tumors appear to be less biologically aggres- from Toxocaris infection or from Coats disease. Careful examination
sive than typical retinoblastomas; therefore, extension into the optic under anesthesia to search for the yellow-white lipid deposits of Coats
nerve or through the sclera is very rare. If any calcification is seen, disease within the retina, blood analysis for toxocara titers and other
the diagnosis of retinoblastoma should be suggested. If no calcifica- uveitis laboratory analyses, careful history for episodes of pica, which

FIG. 7-121. Diffuse retinoblastoma. A. Axial CT


image shows a smoothly marginated high attenuation
collection or mass (arrows) in the left ocular globe.
No calcification is seen. B. Axial 3D T2-weighted
image shows the lesion (arrows) to be hypointense
compared to vitreous (isointense to white matter).
C. Homogeneous enhancement (arrows) is seen
after intravenous infusion of paramagnetic contrast
material.

Barkovich_Chap07.indd 760 5/6/2011 9:17:39 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 761

FIG. 7-122. Coats disease.


A. Axial CT image shows cres-
centic hyperdensity (arrow-
heads) in the posterior right
globe secondary to proteina-
ceous subretinal exudate. The
globe is of normal size and no
calcification is present, sug-
gesting the diagnosis of Coats
disease. B. Axial T1-weighted
image shows that the exudate
(arrowheads) is slightly hetero-
geneous and has signal inten-
sity similar to white matter. C.
Axial T2-weighted image shows
a small linear area of marked
hypointensity (arrowheads) in
the exudate, probably repre-
senting hemorrhage. D. Post-
contrast T1-weighted image
shows absence of enhancement
(arrowheads), which helps to
differentiate this lesion from
diffuse retinoblastoma.

are correlated with toxocara canis exposure, and low birth weight (for
retinopathy of prematurity) must then be utilized to make this distinc-
tion (665).

Differential Diagnosis—Medulloepithelioma, Infection,


and Malformations
When bilateral ocular tumors are present in an infant, the diagnosis is
retinoblastoma until proven otherwise. When the tumor is unilateral,
the primary differential diagnosis is Coats disease, which is a degen-
erative proliferative disease of the retina (666). In Coats disease, a
defect is present in the endothelial cells of retinal blood vessels, result-
ing in leakage of secretions that are high in cholesterol content into
the subretinal area and causing retinal detachment and hemorrhage
(Fig. 7-122) (667–670). Absence of calcification helps to differenti-
ate Coats disease from retinoblastoma. Differentiation from diffuse
retinoblastoma is made by the absence of enhancement of the secre-
tions in Coats disease (although the detached retina does enhance) on
postcontrast MR images (Fig. 7-122) and the presence of blood (Fig.
7-122). Another helpful finding is that the affected orbit in Coats dis-
ease is slightly smaller (20% by volume) than the contralateral, nor-
mal globe (671) although this is difficult to determine without the use
of volume measurements. Otherwise, Coats disease has an identical
appearance on CT to larval granulomatosis (Fig. 7-123, also called
ocular toxocariasis or sclerosing endophthalmitis), caused by a Toxo- FIG. 7-123. Larval granulomatosis. Axial CT shows crescentic hyper-
cara canii infection (672). Both are differentiated from retinoblastoma density in the posterior right globe. The appearance is identical to that
by the absence of calcification on CT. Larval granulomatosis, which of Coats disease (Fig. 7-122). (This case courtesy Dr. Sylvester Chuang,
is histologically an eosinophilic abscess, may show focal enhancement Toronto.)

Barkovich_Chap07.indd 761 5/6/2011 9:17:40 PM


762 Pediatric Neuroimaging

FIG. 7-124. Persistent hyperplastic primary vitreous. A. Axial


CT image shows a small right globe that has an abnormal,
rounded lens and shows abnormal hyperintensity in the region
of the vitreous. B. Postcontrast image at the same level shows
areas of enhancement (arrowheads) behind the lens that may
represent enhancement of the persistent primary vitreous. C
and D. Axial 3D T2-weighted image (C) and T1-weighted image
(D) show crescentic regions of subretinal hemorrhage pos-
terior to the abnormally-shaped lens in the small right globe.
The combination of microphthalmia and retinal detachment is
strongly suggestive of PHPV. E. Persistent hyaloid canal. Axial
CT through the globes shows a hypoplastic right globe. More-
over, a streaky density is seen extending toward the head of the
optic nerve through the vitreous (arrows). This represents a per-
sistent hyaloid canal and confirms the diagnosis of persistent
hyperplastic primary vitreous.

on MRI if granulomas are present in the retina (673). The persistent cussed in Chapter 5) is almost always bilateral and symmetric, rarely
hyperplastic primary vitreous is another anomaly that results in leu- calcifies (except in advanced stages), and is usually associated with
kocoria, retinal detachment, and subretinal hemorrhage (see Chapter microphthalmia (675); a history of low birth weight or prematurity
5 and Figs. 7-124 and 7-125). Differentiation from retinoblastoma is should be sought if this diagnosis is suspected. Microphthalmia and
made by the absence of calcification and by the presence of a hypoplas- leukocoria may sometimes be associated with retinal astrocytic hamar-
tic globe on the affected side (668,674). In contrast to the appearance tomas of tuberous sclerosis (see Chapter 6). Retinal Hamar-toma may
of Coats disease, the affected globe in PHPV is visibly smaller than calcify, making the differentiation from retinoblastoma difficult by
the contralateral normal globe. Subretinal hemorrhage (Fig. 7-124) is ultrasound and by CT. A search for cortical and subependymal hama-
often present in the affected globe. Occasionally, a persistent hyaloid rtomas in the brain will often help to make the proper diagnosis (676).
canal can be seen as an enhancing linear structure extending from A very rare cause of leukocoria is medulloepithelioma, a rare embryo-
the posterior aspect of the lens to the optic nerve head, confirming nal intraocular neoplasm of young children that arises from the med-
the diagnosis (Fig. 7-124E). Retinopathy of prematurity (also dis- ullary epithelium of the ciliary body. This tumor sometimes calcifies,

Barkovich_Chap07.indd 762 5/6/2011 9:17:41 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 763

FIG. 7-125. Medulloepithelioma. Axial T2-weighted image (A) shows a


hypointense mass (arrow) in the anterior chamber near the ciliary body.
Coronal (B) and axial (C) postcontrast T1-weighted images show uni-
form enhancement of the mass and thickening of surrounding soft tissues.
Medulloepitheliomas can mimic inflammatory disease, both clinically and
on imaging.

due to the presence of a hyaline cartilage component found in 30% of Vascular Masses
teratoid medulloepitheliomas (677). Medulloepitheliomas can be dif- Orbital hemangiomas and venolymphatic malformations are best
ferentiated from classic retinoblastomas if they originate from the cili- thought of as hamartomatous malformations that arise from an anlage
ary bodies but, when they arise from the retina, differentiation may be of vascular mesenchyme that is capable of differentiating into both
impossible. Medulloepithelioma arising from the ciliary body may be lymphatic vessels and blood vessels, as well as other mesenchymal tis-
difficult to differentiate from DIR, as discussed above; in the author’s sues (683,684).
experience, medulloepithelioma is a more sharply defined mass than
DIR. Ultrasound may be useful, as it can show cystic collections within Hemangioma
the tumor. On CT, medulloepitheliomas appear as dense, irregular, Hemangiomas, formerly referred to as capillary hemangiomas (685),
enhancing masses that may be calcified. On MRI, they are slightly are the most common vascular masses to present in the orbits of chil-
hyperintense to vitreous on T1-weighted images and hypointense on dren. These malformations, which have a slight (3:2) female predomi-
T2-weighted images; the solid portions exhibit marked, homogeneous nance, typically present at birth or in the first few months of life; they
enhancement after intravenous administration of paramagnetic con- then enter a proliferative phase, with rapid growth that lasts up to
trast (Fig. 7-125) (670). If cystic areas are present, the enhancement 10 months. They typically cease growth during the second year of life
may appear heterogeneous. and then slowly involute over the subsequent 5 to 10 years (683,684).
Hemangiomas can develop anywhere in the orbit; anterior lesions are
most common detected clinically, but some retrobulbar hemangiomas
Extraocular Orbital Masses
are asymptomatic and may only be detected by imaging. As the mass
Extraocular orbital masses in children include a wide range of patholo- enlarges, it may cause complications including proptosis, amblyopia,
gies, including vascular masses (venolymphatic malformations— optic nerve stretching, bleeding, and corneal ulceration (686). Some
lymphangiomas, varices, venous malformations, and hemangiomas), orbital hemangiomas may be associated with cerebral and vascular
rhabdomyosarcomas and (rarely) rhabdomyomas, neurofibromas and anomalies of the PHACES syndrome (see Chapter 6).
schwannomas, infections, cysts, dermoids, cephaloceles, gliomas, men- CT scans of orbital hemangiomas show a diffuse, lobulated, usually
ingiomas, LCH, leukemic infiltrates, lymphoma, Ewing sarcoma, and poorly marginated mass that may be located anywhere in or around the
metastatic neuroblastoma (678–682). Many of these masses can develop orbit. The mass may be outside the cone of extraocular muscles (extra-
in any compartment (intraconal, conal, or extraconal) of the orbit, so conal, more common), inside the muscle cone (intraconal, less com-
they cannot be distinguished by topology. Dermoids and cephaloceles mon) or both. Rarely, they spread intracranially through the orbital
were discussed in Chapter 5 and infections are discussed in Chapter 11. fissures. Diffuse enhancement is seen after intravenous administration
Optic gliomas, meningiomas, and LCH were discussed earlier in this of contrast. MRI shows the tumor as well-marginated, with slightly
chapter. The remaining pathologies will be discussed briefly. hyperintense signal intensity compared to extraocular muscles and

Barkovich_Chap07.indd 763 5/6/2011 9:17:42 PM


764 Pediatric Neuroimaging

FIG. 7-126. Orbital hemangioma (formerly capillary heman-


gioma). A. Coronal T1-weighted image shows a mass (arrowheads)
inferior to the right ocular globe, that is both inside and outside of
the muscle cone. B. Coronal T2-weighted image shows the lesion
(white arrowheads) to be of soft tissue intensity. A curvilinear
flow void is present within the mass. C. Postcontrast T1-weighted
image shows that the hemangioma enhances uniformly other than
the curvilinear vascular structures.

hypointense signal intensity compared to fat on T1-weighted images both the lymphatic and venous systems (688). Nonetheless, some are
(Fig. 7-126). Moderate to marked enhancement, usually homogeneous, primarily lymphatic while others are primarily vascular; therefore, the
is observed after administration of paramagnetic contrast (Fig. 7-126). disorders are discussed separately within this section with the under-
On T2-weighted images, hemangiomas are hyperintense compared to standing that these malformations essentially always contain both
muscle and fat, and hypointense compared to fluid. Curvilinear flow components.
voids, representing blood vessels, are typically present within and at the Most lymphatic malformations (formerly called lymphangiomas)
periphery of the mass (Fig. 7-126); this finding is useful in differentiat- present during childhood; they are the most common vascular mass
ing hemangiomas from more aggressive lesions, such as rhabdomyo- of the orbit presenting between the ages of 1 and 15 years. Patients
sarcomas. Dark, fibrous septa may be seen between the hyperintense typically present with recurrent episodes of proptosis that result from
lobules on T2-weighted images. The affected orbit is often enlarged, recurrent hemorrhages within the mass (possibly due to vascular
probably as a result of the congenital nature of the lesion (684,687). As thromboses [689]) or from reactions of the lymphatic system due to
the mass involutes, fat develops in the mass, and it becomes more hyper- upper respiratory infections; other complications include astigmatism,
intense on T1-weighted images and less hyperintense on T2-weighted corneal exposure, hyperopia secondary to pressure on the posterior
images. Uniform, intense enhancement is typically seen after intrave- globe, strabismus, glaucoma, and compressive neuropathy (690). Such
nous administration of paramagnetic contrast (686). Hemangiomas episodes can result in poor visual outcome (691). The malformation
are most easily differentiated from other orbital tumors by the curvi- may involve any part of the orbit, including eyelids, conjunctiva, sclera,
linear flow voids within and at the periphery of the mass. intraconal spaces, and extraconal spaces. Anterior lesions may extend
to the forehead and cheek, while posterior lesions may extend through
Combined Venous Lymphatic Malformations—Lymphatic the superior orbital fissure into the cavernous sinus or middle cranial
Malformations (Lymphangiomas) and Venous fossa (692,693). This permeation of the intra- and extraconal compart-
Malformations (Cavernous Hemangiomas) ments makes complete resection nearly impossible.
Recent work has shown that orbital lymphatic malformations and Pathologically, lymphatic malformations are composed of abnor-
venous malformations are part of a spectrum of disorders involving mal venous channels that are associated with enlarged lymphatics,

Barkovich_Chap07.indd 764 5/6/2011 9:17:43 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 765

particularly in the eyelids and conjunctiva. Histology shows thin-walled


channels lined by epithelium and containing serous fluid or blood. The
spaces range from large cysts to sponge-like microcystic spaces, often
containing a mixture of both (688).
On CT, lymphatic malformations are lobulated, heterogeneous,
poorly defined lesions that cross anatomic boundaries such as the
conal fascia and orbital septum (Fig. 7-127). Small focal calcifications,
representing phleboliths, are occasionally seen. Areas of hyperdensity,
representing acute hemorrhage, may be present, especially after recent
exacerbation of proptosis. Enhancement is heterogeneous, usually with
weak enhancement along the periphery and in the stromal framework
(678,687). On MRI, lymphatic malformations appear as lobulated
masses containing septa that divide it into multiple cysts and lobules
that have varying sizes and intensities on both T1- and T2-weighted
images. The majority of the mass is typically hypointense to muscle
on T1-weighted images and hyperintense to muscle on T2-weighted
images (Fig. 7-128). The variations of intensity are the result of differ-
ing amounts of blood and protein in the cysts. Fluid-fluid levels may
be present within the cysts (Fig. 7-128); if present, they are strongly
suggestive of lymphatic malformation. Heterogeneous enhance-
ment follows intravenous administration of paramagnetic contrast
FIG. 7-127. Lymphatic malformation (formerly called lymphangiomas). (694). Some lesions have multiple components, each with different
A and B. Axial noncontrast CT images show an irregular, lobulated mass appearances; some areas have characteristics suggesting lymphatic
(arrows) in both the extra- and intraconal spaces of the left orbit, causing malformations and others more suggestive of venous malformations
mild proptosis. (Fig. 7-128) (694).

FIG. 7-128. Lymphatic malformation. A. Sagittal


T1-weighted image shows an irregular mass (arrows),
primarily intraconal, in the right orbit. An extraorbital
component (arrowheads) is seen extending superiorly
above the orbit. B. Axial 3D T2-weighted image shows
two distinct components of the lesion. The intraorbital
component (arrows) is multiloculated and cystic in
appearance with fluid-fluid levels, more compatible with
a lymphatic component, while the supraorbital compo-
nent (arrowheads) is more homogeneous and of lower
signal intensity, more compatible with a venous com-
ponent. C. Postcontrast axial T1-weighted image shows
heterogeneous enhancement (arrows and arrowheads).

Barkovich_Chap07.indd 765 5/6/2011 9:17:44 PM


766 Pediatric Neuroimaging

FIG. 7-129. Orbital venous malformation (formerly called


cavernous hemangioma). A. Axial T1-weighted image shows an
intraconal mass (m) in the right orbit. Venous malformations can
be intraconal, extraconal, or both. B. Coronal T2-weighted image
shows the mass to be slightly heterogeneously hyperintense. C.
Coronal T1-weighted image after infusion of paramagnetic con-
trast shows diffuse, slightly heterogeneous enhancement.

Venous malformations of the orbit (also called cavernous developmental venous anomalies or cavernous malformations, but AV
malformations, formerly known as cavernous hemangiomas [685]) malformations and AV fistulae may rarely be found; the intracranial
are uncommonly identified in children, becoming symptomatic more anomaly may be ipsilateral or contralateral to the orbital lesion (690).
commonly in young and middle-aged adults (683,695). They typically Up to14% of these patients may present with intracranial hemor-
present with painless proptosis that remains unchanged with Valsalva rhage (691). Katz et al. found that the orbital lesions in patients with
maneuvers, differentiating them from orbital varices, which enlarge (noncontiguous) intracranial vascular anomalies were diffuse with
during a Valsalva maneuver (683). When located anteriorly, they pres- superficial and deep components (100%) involving the intraconal and
ent earlier and often extend to the forehead, temporal region, or cheek. extraconal spaces (57%). The affected orbit and the inferior orbital fis-
On CT, venous malformations appear as homogeneously enhancing sure were expanded with extension into the pterygopalatine fossa in
round or ovoid masses that are usually intraconal and spare the orbital 82%, while the superior orbital fissure was always involved, with exten-
apex. Calcified phleboliths are frequently seen within the mass (687). sion into the middle cranial fossa in 43%. Anterior extension into soft
On MRI, venous malformations appear as homogeneous, well defined, tissues of the face and forehead and palatal involvement were relatively
round or ovoid, slightly lobulated masses that are usually intraconal common. In contrast, lymphatic malformations without intracranial
(Fig. 7-129). They are isointense to the orbital muscles on T1-weighted vascular anomalies were more anterior, less diffuse, less likely to extend
images and hyperintense to the orbital muscles on T2-weighted into the soft tissues of the face, have associated vascular lesions, or have
images; often variation in signal intensity is present among the cysts. If a poor visual outcome (691).
fluid-fluid levels are present, the dependent portion is typically more
hypointense on T2-weighted images. The lesions enhance diffusely, but Orbital varices
somewhat heterogeneously (Fig. 7-129) after intravenous infusion of Orbital varices are venous malformations that occur in the retrob-
paramagnetic contrast (684). ulbar region. They typically present in later childhood or adulthood
It should be noted that intracranial vascular malformations are with intermittent proptosis that is exacerbated by a Valsalva maneuver,
seen very commonly in patients with periorbital lymphatic and venous bending over, or lying prone. On CT and MRI, varices are identified
malformations; an incidence of up to 70% is reported (690). Therefore by a curvilinear configuration, the presence of calcified phleboliths,
contrast-enhanced study of the brain should be obtained along with intense enhancement after contrast administration, and an increase in
the orbital study. The vascular malformations are most commonly size in the prone position or with a Valsalva maneuver. On occasion,

Barkovich_Chap07.indd 766 5/6/2011 9:17:45 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 767

FIG. 7-131. Bilateral colobomas with cysts. Axial T1-weighted image


shows bilaterally small globes with cysts extending from the posterior mid-
line of the globes. The left globe and colobomatous cyst are hyperintense
FIG. 7-130. Coloboma with cyst. Axial T1-weighted image obtained with a secondary to subretinal hemorrhage. (This figure courtesy Dr. Sylvester
3 cm surface coil shows the shrunken, malformed globe (closed black arrows) Chuang, Toronto.)
with lens (small white arrows) and posterolateral cyst (large white arrows).

images obtained in a supine position without Valsalva may be normal; lymphocytic orbital infiltrates other than granulocytic sarcomas are
if the proper history is obtained, further scans should be obtained dur- uncommon in children. About half are bilateral at the time of presenta-
ing maneuvers. CT is a better study because of the shorter imaging tion; bilaterality of orbital masses in children is uncommon, so this finding
times that allow scanning to be performed during a Valsalva maneuver should arouse suspicion of a granulocytic sarcoma. On MRI, granulocytic
(684,687). sarcomas are round to oval shaped, slightly lobulated lesions that are
hypointense compared to muscle on T1-weighted images and isoin-
Orbital Cysts tense to hyperintense compared to muscle on T2-weighted images
When retro-orbital cysts are seen in infants and children, the most com- (698,699). They enhance intensely and homogeneously after adminis-
mon cause is a severe colobomatous malformation of the globe. Ocular tration of intravenous contrast.
colobomas are anomalies of the globe that result from a failure of the
posterior midline of the globe (the choroidal fissure) to close properly, Metastatic Neuroblastoma
with consequent localized defects in the uvea, retina, and optic nerve Metastatic neuroblastoma, although already mentioned twice in this
(682,696). Colobomas and their embryogenesis are discussed in Chap- chapter regarding calvarial and skull base masses, must be discussed
ter 5, in the section on “Anomalies of the Eye.” If the malformation in this section, as well, as it is the most common tumor to metastasize
isn’t recognized, the orbital cyst may mimic a more aggressive orbital to the orbit in infants and young children (683). As many as 8% of
mass. The cyst may be small with a normal sized globe, moderate in patients with neuroblastoma present with orbital symptoms; bilateral
size with a slightly small globe, or large with a small, malformed globe orbital lesions are noted in as many as 40% of patients with metastatic
(Figs. 7-130 and 7-131). MRI and CT are useful in making an accurate disease (683,684). Patients typically present with rapidly progressive
diagnosis, in characterizing the cyst, in determining the presence of proptosis. CT shows a soft tissue mass that typically arises from a par-
defects in the globe, and in assessing the brain for other anomalies. tially destroyed orbital wall; the posterior superolateral wall seems to
Both the cyst and globe are isointense to normal vitreous on all imag- be involved most commonly, but any site in the orbital wall can be
ing sequences. Contrast enhancement is not seen (682,696,697). affected. The bone typically shows a permeated pattern or elevation
of the periosteum secondary to rapid growth of tumor (Fig. 7-132).
Leukemia (Granulocytic Sarcoma, Chloroma) MRI shows focal or extensive soft tissue, often partially hemorrhagic,
Granulocytic sarcomas are solid tumors of immature granulocytes that expanding the orbital wall and growing into the orbit. Tumor growth
develop in some patients with myelogenous or myelomonocytic leuke- is often multidirectional and can extend superiorly into the anterior
mia (698). They most commonly occur in the skin, bones, sinuses and cranial fossa or laterally, displacing or invading the temporalis muscle
orbits, but rarely involve the CNS. Although granulocytic sarcomas (Fig. 7-132). Nonhemorrhagic portions of tumor typically remain
arising in other areas usually develop during relapses in children with isointense to gray matter on both T1- and T2-weighted images (see
known leukemia, those arising in the orbit may be the initial manifes- Fig. 7-132). The tumor enhances markedly after intravenous infusion
tation of leukemia (698,699). After testing, most affected patients are of contrast. A search should be made for other metastases of the calva-
found to have simultaneous evidence of bone marrow or peripheral rium, skull base, and petrous pyramids.
blood disease. Although most patients present with proptosis, some
may have pain and chemosis, suggesting an inflammatory disorder. Langerhans Cell Histiocytosis
CT shows poorly-defined, homogeneous soft tissue masses that may LCH has also been mentioned multiple times in this chapter, as the
be intraconal or extraconal. The masses typically infiltrate the orbital disease can affect nearly any part of the brain and any of the bony
fat and conform to orbital structures; bone erosion is very rare. These structures of the head and neck (241,242,246,250,504). Like metastatic
characteristics are typical of all lymphocytic orbital infiltrates; however, neuroblastoma, it most commonly involves the posterolateral part of

Barkovich_Chap07.indd 767 5/6/2011 9:17:46 PM


768 Pediatric Neuroimaging

FIG. 7-132. Metastatic neuroblastoma to orbit. A. Axial CT shows a


mass (large arrows) arising within the lateral wall of the orbit. Periosteal
reaction (small arrows) is indicated by irregular thickening of the
bone. B. Coronal T2-weighted image shows the mass to be isointense
to gray matter and to have an intraorbital (small arrows), intracranial
extradural (large arrows) and extracranial (arrowhead) component. C.
Coronal postcontrast fat-suppressed T1-weighted image shows uniform
enhancement of the mass. Note the destruction of bone resulting from
intracranial (large white arrows) and extracranial (arrowhead) exten-
sion of the mass through the orbital walls. Normal orbital structures
are compressed medially by the intraorbital component (small white
arrows) of the mass.

the orbital wall, at the suture between the frontal bone and the greater tissue that are only seen in the setting of NF1. A more extensive
wing of the sphenoid bone. Affected children typically present with discussion of the imaging characteristics of neurofibromas is given in
either proptosis or a rapidly enlarging periorbital mass. Periorbital Chapter 6, in the section on “Neurofibromatosis Type 1.” Neurofibromas
edema or ecchymosis may raise suspicion for child abuse. CT shows a tend to grow centripetally along the nerve from the periphery toward the
sharply marginated osteolytic soft tissue mass; the sharp margination center. When they involve the orbital region, plexiform neurofibromas
of the bone lesion distinguishes it from the permeative pattern seen present with proptosis, visual loss, and marked facial asymmetry (683).
in leukemia or metastatic neuroblastoma. The soft tissue component They appear on both CT and MRI as serpiginous, irregular, poorly
typically uniformly enhances after intravenous contrast enhancement. marginated masses that surround and engulf normal orbital structures
MRI shows a soft tissue mass that has signal intensity similar to skeletal (Fig. 7-133). Neurofibromas cross normal fascial boundaries such as
muscle and uniformly enhances after intravenous administration of the orbital septum and, thus, may be periorbital as well as intraorbital.
paramagnetic contrast. If LCH is considered a possible diagnosis, the The involved orbit is often enlarged and the adjacent greater sphenoid
brain should be examined to look for involvement, most commonly in wing is often partially absent; the bone may become progressively more
the pituitary stalk (see section on “Sellar and Suprasellar Tumors”), and deficient, suggesting active loss of bone (700). Although any portion
the skull and bones in the remainder of the body should be evaluated for of the orbit may be involved, the superior portion, particularly that
other sharply marginated lytic lesions or, in the spine, vertebra plana. If region close to the first division of the fifth cranial nerve, is most often
other characteristic lesions are found, biopsy might be avoided. involved. Such masses may extend through the superior orbital fissure
into the cavernous sinus, which may also be expanded (Fig. 7-133). On
Plexiform Neurofibroma CT, the masses appear as serpiginous lesions, isodense with the extraoc-
Plexiform neurofibromas are highly vascular, poorly defined, and dif- ular muscles that do not respect fascial boundaries. On MRI, it is easier
fusely infiltrative masses involving peripheral nerves and connective to appreciate the multiple serpiginous enlarged nerve fascicles coursing

Barkovich_Chap07.indd 768 5/6/2011 9:17:47 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 769

FIG. 7-133. Orbital plexiform neurofibroma. A. Parasagittal T1-weighted image shows numerous irregular curvilinear streaks in the expanded right orbit
of this patient with NF1. B. Postcontrast axial image shows some enhancement of the curvilinear structures. Extension is seen through the enlarged superior
orbital fissure (open arrow) into the cavernous sinus (closed arrows). This patient also had a parenchymal astrocytoma (black arrows).

through the orbit, isointense with muscle on T1-weighted images and Dermoids and Epidermoids
hyperintense compared with muscle on T2-weighted images with fat Dermoids and epidermoids were discussed extensively earlier in this
saturation. Contrast enhancement is variable and is usually heteroge- chapter. They can develop anywhere in the orbit, but are most com-
neous (684,687). MRI is the imaging study of choice because of the monly seen anteriorly, near the ocular globe and near the sutures
ability to analyze for cavernous sinus involvement without the use of between the bones that comprise the orbit. As in other parts of the
contrast and the ability to analyze the brain for other manifestations of head, neck, and brain, dermoids and epidermoids are sharply margin-
NF1. In the setting of NF1, no significant differential diagnosis exists. ated masses. The presence of fat signal (Fig. 7-134) is quite characteristic

FIG. 7-134. Orbital der-


moids. A and B. Axial image
(A) and sagittal reformation
(B) from a CT scan show a
sharply demarcated fat den-
sity mass (arrows) superior
to the left ocular globe. C
and D. Axial precontrast and
postcontrast fat-suppressed
T1-weighted images in a dif-
ferent patient show the high
signal of the dermoid (arrow)
to be suppressed, confirming
the fatty nature of the lesion.

Barkovich_Chap07.indd 769 5/6/2011 9:17:48 PM


770 Pediatric Neuroimaging

of dermoids and water signal is very characteristic for epidermoids on and solid masses. CT, which can also generally be performed without
both CT and MRI. sedation, reliably differentiates solid from cystic congenital masses
and often allows characterization of the masses. However, children
Rhabdomyosarcomas/Rhabdomyomas generally have less fat and, without the fat planes, CT provides less
Rhabdomyosarcomas are discussed in the following section. contrast in the pediatric neck than the adult neck. Also, as discussed
many times in this book, exposure of young children to ionizing radi-
ation should be avoided unless medically necessary. Thus, if surgical
Masses of the Face and Neck
intervention is anticipated, children at our institution are usually also
Masses of the pediatric face and neck involve a wide spectrum of both studied by MRI.
congenital and acquired lesions. A full discussion of these masses is A nice review of a large series of masses involving the pediatric face
beyond the spectrum of this book, which is concerned primarily with and neck was given by Vazquez et al. (701). They published a review of
the CNS and its coverings. Readers are referred to books on otolar- 145 pediatric patients from age 1 day to age 18 years who presented to
yngology for more information on these topics. A brief overview of their practice with neck masses. The results of this review are summa-
masses in this region is warranted and will follow, along with brief dis- rized below and in Table 7-14.
cussions of the most common lesions (see Table 7-14). An important Congenital lesions was the largest group, comprising 40% of these
concept is that neck masses in children are most commonly associated masses. These included 19 thyroglossal duct cysts (31%), 15 lymp-
with inflammatory disease, congenital anomalies, and benign neo- hangiomas (25%), 8 branchial cleft anomalies (13%), 6 dermoids
plasms. Malignant neck masses are relatively much less common than (10%), 6 cases of tracheal stenosis (10%), 3 teratomas, 2 lingual thyroid
in adults. glands, and 2 cases of thyroid gland hypogenesis.
Ultrasound is often the first modality used in the evaluation of Benign tumors and tumorlike conditions was the second largest
pediatric neck masses. Ultrasound can be performed without seda- group of pediatric neck and face masses. This group comprised 19% of
tion and is the most reliable method to differentiate between cystic the cases and included 15 cases of fibromatosis colli (56%), 6 aggressive
fibromatoses (22%, most common in the tongue, around the mandible,
or within the infratemporal fossa), 3 cervicothoracic lipoblastomatoses
(11%), 1 parathyroid adenoma, 1 juvenile laryngotracheal papilloma-
TABLE 7-14 Most Common Pediatric tosis, and 1 plexiform neurofibroma.
Neck Masses The third largest group, malignant tumors (18%), was almost
identical in size to the benign tumor group. The most common tumor
in this group was lymphoma (10 tumors, 38%). Other common malig-
Congenital masses (40%)
nant tumors were soft tissue sarcomas (23%, of which two thirds were
Thyroglossal duct cysts
rhabdomyosarcomas), carcinomas (19%, most commonly thyroid car-
Lymphangiomas
cinoma), and neuroblastoma (arising from the cervical sympathetic
Branchial cleft anomalies
chain, 19%).
Dermoid tumor
Inflammatory lesions were the next largest group of masses
Tracheal stenosis
(12%). Cervical adenitis from mycobacterial infections or cat scratch
Teratomas
fever (10 cases, 62%) was the most common cause in this group. In
Lingual thyroid gland
this regard it is important to remember that more than 90% of myco-
Benign Tumors/Tumor Like Conditions (19%) bacterial cervical lymphadenitis in children is due to nontuberculous
Fibromatosis coli mycobacterial (usually Mycobacterium avium-intracellulare); this pre-
Aggressive fibromatosis dilection is in marked contrast to adults in whom tuberculous cervi-
Cervicothoracic lipoblastomatosis (702) cal lymphadenitis is more common (704). The authors also reported
Plexiform neurofibroma 3 cases (19%) each of sialadenitis and retropharyngeal abscess (from
Juvenile laryngotracheal papillomatosis tonsillitis or trauma).
Neoplasms (18%) Masses of vascular origin (10%) comprised the smallest group. Jug-
Lymphoma ular vein varices (fusiform dilatations of the vein secondary to presence
Soft tissue sarcoma (especially rhabdomyosarcoma) of jugular venous valves) accounted for 60% of these vascular masses.
Neuroblastoma Cervical hemangiomas were the only other diagnosis of significant fre-
Carcinoma (especially thyroid) quency, comprising 33% of the masses in this group. The authors note,
however, that a pulsatile cervical mass in a child should raise suspicion
Inflammatory Masses (12%) for a cervical carotid artery aneurysm, which has the greatest clinical
Cervical adenitis significance. Cervical carotid aneurysms may be congenital (usually
Sialadenitis secondary to connective tissue disorder such as Ehlers-Danlos, Marfan,
Retropharyngeal abscess Kawasaki, or Maffucci syndromes), posttraumatic, or postinfectious.
Inflammatory myofibroblastic tumor (plasma cell granuloma,
inflammatory pseudotumor) (703) Thyroglossal Duct Cyst
Vascular Masses (10%) Thyroglossal duct cysts arise from remnants of the embryonic thyro-
Jugular vein varices glossal duct and account for up to 70% of congenital neck masses in
Hemangiomas children (705). The thyroid gland originates in the third week of fetal
Cervical carotid artery aneurysms life from a median endodermal thickening in the floor of the primi-
tive pharynx. The thyroid primordium develops at the foramen cecum,
Adapted from Vazquez et al. (701). a midline depression that is located between the anterior two thirds
and posterior one third of the tongue. The developing gland descends

Barkovich_Chap07.indd 770 5/6/2011 9:17:49 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 771

FIG. 7-135. Bland and infected thyroglossal duct cysts. A. Axial


postcontrast CT image shows an uninfected cyst (arrows) within
the left thyroid strap muscle. Note the thin, nonenhancing wall.
B. Axial CT image shows a cyst (arrows) that is exophytic, extend-
ing anterior to the strap muscles. C. Postcontrast CT image of an
infected cyst shows a thick, enhancing wall (arrows) with some
obscuration of surrounding fat planes.

along the thryroglossal duct, passing anterior to the hyoid bone and hypoechoic). CT shows a low attenuation, well-defined, nonenhanc-
laryngeal cartilages, then anterior to the thyrohyoid membrane until ing mass; the wall may show a thin rim of enhancement (Fig. 7-135A).
it reaches its final position by the seventh gestational week. Thyro- MRI usually shows sharply marginated, round or ovoid masses that
glossal duct cysts are found along the course of descent of the thyroid manifest hypointensity on T1-weighted images and hyperintensity on
gland, typically found in the midline and occasionally in a parame- T2-weighted images (Fig. 7-136) (709). Cysts in the tongue base are
dian position. They are classified by location as above the hyoid bone best seen on sagittal images, while those in the strap muscles are best
(20%–25%), at the hyoid bone (15%–45%), or inferior to the hyoid seen on axial images. It is important to remember that if the cyst is
bone (35%–65%). Children typically present with a mobile, midline, infected, or has hemorrhaged, high signal may be seen on T1-weighted
nontender mass that elevates with swallowing. If the cyst is infected, a images and low signal intensity may be present on T2-weighted images.
history of recent growth of the cyst or of local pain and tenderness may Hemorrhage and infection may also result in thickening and marked
be elicited. Typically, the mass elevates with swallowing or with protru- enhancement of the cyst wall on both CT (Fig. 7-135C) and MRI. A
sion of the tongue (706). history of recent discomfort or local edema, indicative of infection, and
Imaging studies may show the thyroglossal duct cyst anywhere the characteristic location of the cyst will usually allow diagnosis, even
along the path of the embryonic thyroglossal duct (from the median after infection has occurred.
lobe of the thyroid gland to the hyoid bone and then to the foramen If surgery is planned, it is important to ensure that the affected
cecum in the midline of the tongue base [707]). Most are in or near child has thyroid tissue outside of the cyst. Any type of imaging (ultra-
the midline (705). A common paramedian location is within the strap sound, CT, or MRI) can be used to ensure that the normal thyroid
muscles in front of the thyroid cartilage, where the lesions may be gland is present.
completely intrinsic (Fig. 7-135A) or exophytic (Fig. 7-135B). These
may extend all the way to the hyoid bone, where they typically termi- Branchial Cleft Cysts
nate at the site of attachment of the duct to the bone. Another com- Branchial cleft cysts are the result of abnormal development of the
mon location is in the midline of the tongue base (708). Ultrasound branchial apparatus, which consists of five paired regions on the
shows thin-walled cysts with variable internal echogenicity (usually ventrolateral surface of the embryonic head between the third and

Barkovich_Chap07.indd 771 5/6/2011 9:17:49 PM


772 Pediatric Neuroimaging

FIG. 7-136. Thyroglossal duct cyst. A. Sagittal


T2-weighted image with fat suppression shows a hyperin-
tense mass at the base of the tongue. An extension along the
remnants of the thyroglossal duct (arrow) is seen extending
inferiorly. B. Axial T2-weighted image with fat suppression
shows the ovoid, sharply marginated, thin walled, mark-
edly hyperintense mass (arrow) within the tongue base. C.
Coronal postcontrast T1-weighted image with fat suppres-
sion shows the thin-walled mass (arrows) with complete
lack of enhancement.

seventh gestational weeks. The five branchial arches consist of a core decades of life; males and females are equally affected. Rarely, cranial
of mesenchyme, covered externally with ectoderm and internally with neuropathies may develop (712).
endoderm. As a result, adjacent arches are separated by ectodermal About 10% of branchial cleft cysts arise from the first bran-
clefts externally and endodermal pouches internally (710,711). Most chial cleft, probably as a result of incomplete obliteration of the cleft
anomalies encountered by neuroradiologists are the results of malde- between the mandibular process of the first arch and the second arch.
velopment of the clefts; anomalies of the branchial arches and endo- Such cysts typically occur either anterior or posterior to the pinna of
dermal pharyngeal pouches can also occur, but usually cause fistulas the ear (Fig. 7-137), sometimes extending downward to the angle of the
that are discovered and recognized shortly after birth (bilateral in up to mandible. It may course lateral to or through the parotid gland in close
30%) and, therefore, are not discussed here. Branchial cleft cysts most association with, and lateral to, the facial nerve. These lesions should
often present in the same manner as thyroglossal duct cysts, typically be suspected when a young patient has a cyst adjacent to the external
with painless swelling or with recent enlargement of a longstanding auditory canal, superficial to or deep within the parotid gland, or adja-
soft, mobile, nontender mass. Although they may be discovered at any cent to the pinna and extending into the anterior neck to the level of
age, patients are most commonly discovered in the second to fourth the angle of the mandible.

Barkovich_Chap07.indd 772 5/6/2011 9:17:50 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 773

to its external opening at the anterior border of the sternocleidomas-


toid muscle. From here, the fistula passes anterior to the vagus nerve
and above the hypoglossal nerve, but posterior to the glossopharyngeal
nerve and the internal carotid artery. It then crosses over the hypoglos-
sal and superior laryngeal nerve back to the region of the thyrohyoid
membrane, which it crosses to enter the pyriform sinus.
Fourth branchial cleft anomalies occur along a track that starts
along the anterior border of the sternocleidomastoid muscle. Almost
all of these anomalies are fistulas and sinuses, almost always originat-
ing in the pyriform sinus and extending inferiorly, sometimes hooking
around the aortic arch (on the left) or the subclavian artery (on the
right).
On ultrasound, branchial cleft cysts may appear anechoic (∼40%),
homogenously hypoechoic with internal debris (∼25%), pseudosolid
(∼10%), or heterogeneous (∼25%) (716). On CT and MRI, branchial
cleft cysts appear as thin-walled cysts that have signal intensities nearly
identical to that of water. When they occur in characteristic locations
(Figs. 7-137 to 7-139), the diagnosis is straightforward. Pointing of the
FIG. 7-137. First branchial cleft cyst. Axial T1-weighted image shows a
cyst medially towards the carotid sheath or posteriorly deep to the ster-
cyst (arrows) anterolateral to the parotid gland in the subcutaneous fat.
nocleidomastoid muscle (Fig. 7-138) is a characteristic that strengthens
the diagnosis. Difficulties in interpretation result primarily from either
atypical location of the cysts or complications of the cysts, such as infec-
Second branchial cleft cysts are by far the most common (90% to tion or hemorrhage. When assessing cervical cysts in atypical locations,
95% of all branchial anomalies). The second branchial cleft runs deep it is important to keep in mind that second branchial cleft cysts may arise
to the platysma muscle between the second and third arch structures by anywhere from the parapharyngeal space to the anterior wall of the ster-
ascending along the carotid sheath and passing medially between the nocleidomastoid muscle, along a path that extends between the internal
internal and external carotid arteries, above the glossopharyngeal nerve and external carotid arteries (713). When branchial cleft cysts become
and below the stylohyoid ligament. A cyst may develop anywhere along infected, the walls thicken and enhance and the contents become more
this pathway. They are divided into four types: (a) deep to the plat- proteinaceous. Under these conditions, it becomes difficult to differen-
ysma and superficial to the sternocleidomastoid muscle; (b) between tiate the infected cyst from a necrotic lymph node (709,713). Differen-
the sternocleidomastoid muscle and submandibular gland (Figs. 7-138 tiation can often be made by the fact that infected branchial cleft cysts
and 7-139), displacing the carotid sheath medially or posteromedially; incite less surrounding edema than necrotic, infected nodes. However,
(c) between the internal and external carotid arteries, extending to the lymph nodes from nontuberculous mycobacterial infections can strongly
lateral wall of the pharynx; and (d) adjacent to the pharyngeal wall resemble infected cysts, as they incite relatively minimal inflammatory
(713–715). reaction and they occur in similar locations (717). Therefore, nontuber-
Third branchial anomalies are very rare. They usually consist of culous mycobacterial infection should always remain in the differential
cysts or fistulas. The cysts are most frequently located just proximal diagnosis of any cervical cyst with a thick wall.

FIG 7-138. Second branchial


cleft cysts. Axial CT images show
characteristic location of the cysts
(c) between the sternocleidomas-
toid muscle and the subman-
dibular gland and characteristic
pointing (arrows) of the cysts
medially (A) and posteriorly (B).
Sometimes these lesions extend
medially between the internal
and external carotid arteries to
the pharyngeal mucosa.

Barkovich_Chap07.indd 773 5/6/2011 9:17:51 PM


774 Pediatric Neuroimaging

FIG. 7-139. Second branchial cleft cyst.


Axial T1-weighted (A) and T2-weighted
(B) images show a well-defined homoge-
neous lesion (arrows) with long T1 and
long T2 relaxation times lying between
the submandibular gland and sterno-
cleidomastoid muscle.

Thymic Cysts lymphatic components and lymphatic malformations almost always


Thymic cysts are rare remnants of the third branchial pouch and, per- have venous components.
haps, should be classified as a variant of branchial cleft cysts (709). Lymphatic malformation is the name given to venolymphatic
During the fifth gestational week, the thymus develops from the malformations with predominantly lymphatic components. They are
ventral wings of the third branchial pouch. During the seventh and malformative lesions, composed of thin-walled sacs of various sizes
eighth weeks, thymic primordia detach from the pharynx and migrate containing lymphatic fluid that are generally discovered in infancy.
caudomedially to the anterior mediastinum along the thymopharyn- Clinically, lymphatic malformations present as soft, nontender neck
geal duct, which subsequently involutes. Failure of complete involu- masses that can develop anywhere in the neck, face, or oral cavity.
tion can result in formation of a cyst anywhere along the tract of the The imaging characteristics vary depending on the size of the cysts
thymopharyngeal duct from the angle of the mandible to the upper and the presence or absence of hemorrhage. Some of the lesions have
mediastinum. very small cystic spaces and may appear solid; these are referred to
Affected patients present with dysphagia, respiratory distress, as lymphangioma simplex. If the cystic spaces are of moderate size
hoarseness, or an enlarging lower neck mass. Imaging studies reveal a (several mm to 1 cm), with thin intervening septae, then they are
largely cystic mass; ultrasound often shows intralesional debris if the called cavernous lymphatic malformations (Fig. 7-140). The sep-
fluid is hemorrhagic or proteinaceous (709). Up to 50% are continu- tae may enhance on MRI (Fig. 7-141). If the cystic spaces are very
ous with the mediastinal thymus via direct extension of the cyst or by large, the lesion is referred to as a cystic hygroma (Fig. 7-141). One
connection to a solid cord or vestigial remnant of thymic tissue (718). important characteristic of lymphatic malformations and other con-
Lesions are almost always adjacent to or within the carotid sheath, fre- genital lesions of the neck (venous malformations, hemangiomas,
quently splaying the carotid artery and jugular vein. neurofibromas, branchial cleft cysts) is that they cross anatomic
fascial boundaries (719). Infections and neoplasms are unlikely to
cross these boundaries until late in the course of the disease; there-
Venolymphatic Malformations fore, the presence of a mass in multiple fascial compartments suggests
As discussed in the section on “Extraocular Orbital Masses,” the a congenital lesion. Fluid-fluid levels are common in cystic hygromas
development of the venous and lymphatic systems is interrelated. (Fig. 7-140). As a result of their crossing of fascial planes, lymphatic
As a result, the entities formerly referred to as lymphangiomas and malformations often involve multiple muscles, nerves, and blood ves-
cavernous hemangiomas have been found to be strongly interrelated, sels. MRI is the preferred method for imaging these lesions because
with both containing both venous and lymphatic components. As it allows better assessment of the spatial extent of the lesion and its
a consequence, the nomenclature of disorders of these systems has relationship to adjacent structures (709); in addition, MRI is better
changed (685). The lesions formerly known as lymphangiomas and able to identify intralesional hemorrhage and fluid-fluid levels. As
those formerly known as cavernous hemangiomas are now combined with other cystic masses of the neck, the diagnosis of lymphatic mal-
into an entity known as venolymphatic malformation (688). As dis- formations only becomes difficult if hemorrhage or infection ensue.
cussed earlier, many of these malformations are primarily lymphatic, Infection causes increased echogenicity (US), increased attenuation
while others are primarily venous. Therefore, lymphatic malforma- (CT), and T1 and T2 shortening (MRI) as well as enhancement and
tions and venous malformations will be discussed separately, while thickening of the cyst wall. Hemorrhage results in rapid enlargement
keeping in mind that venous malformations nearly always have of the malformation. After hemorrhage, echogenicity increases on

Barkovich_Chap07.indd 774 5/6/2011 9:17:52 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 775

FIG. 7-140. Venolymphatic malformation. A. Parasagittal


T1-weighted image shows a low signal intensity, lobulated mass
(arrows) in the left side of the neck below the ear. B. Fat-suppressed
T2-weighted image shows the lateral portion of the mass (arrows)
to be very hyperintense (presumably fluid) with multiple septa. A
more hypointense, presumably solid medial component (s) can be
detected. C. Fat-suppressed postcontrast T1-weighted image shows
the medial, presumably solid component (s) to enhance while the
presumably cystic lateral component (arrowheads) does not.

sonography, attenuation increases on CT, and the fluid changes from They typically present with a painless, slow growing mass, often in
water intensity to the intensity of blood-breakdown products on the face or in the skeletal muscles of the head or neck (masseter,
MRI. When large, lymphatic malformations may be detected in the pterygoid, trapezius, sternocleidomastoid) that has a bluish discolor-
fetus; imaging features are nearly identical to those seen postnatally, ation and remains unchanged with Valsalva maneuvers. They range
except that the cysts are smaller due to surrounding amniotic fluid greatly in size, from a few millimeters in diameter to involvement of
(Fig. 7-141). much of the face. When they are large, the airway may be compressed
Venous malformation is the name given to venolymphatic mal- (720). On CT, venous malformations appear as homogeneously
formations with predominantly venous components. These lesions enhancing round or ovoid masses that often cross fascial planes. Cal-
were formerly known as cavernous hemangiomas [685]), but they are cified phleboliths are frequently seen within the mass (687). On MRI,
not hemangiomas as they do not involute over time and may involve venous malformations appear as heterogeneous, well-defined, round
bone. They are uncommonly identified in children and become or ovoid, slightly lobulated masses that are isointense to the muscle
symptomatic more commonly in young and middle aged adults (683). on T1-weighted images, hyperintense to muscle on T2-weighted

Barkovich_Chap07.indd 775 5/6/2011 9:17:53 PM


776 Pediatric Neuroimaging

FIG. 7-141. Large infantile venolymphatic malformation (cystic hygroma).


A and B. Sagittal and axial T2-weighted images of a fetus show a multiloc-
ulated hyperintense lesion (arrows) involving the face and neck. Note the
marked enlargement of the face due to the large lymphangioma. C. Post-
contrast sagittal T1-weighted image shows multiple large loculations in the
lower face, neck and upper chest. The airway appears open. D–F. Axial T1
(D), T2 (E) and postcontrast T1 (F) images show that the septa between
the loculations are best demonstrated on T2 and postcontrast images. Note
the fluid-fluid layer (arrow) in E and F; it is a very characteristic finding in
venolymphatic malformations.

Barkovich_Chap07.indd 776 5/6/2011 9:17:54 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 777

FIG. 7-142. Venolymphatic malformation. A. Axial


T1-weighted image shows a mass (arrows) in the left maxillary
region. B. Fat-suppressed axial T2-weighted image shows the
lesion to be heterogeneously hyperintense. C. Administration of
contrast shows heterogeneous enhancement.

images, and enhance strongly after intravenous infusion of paramag- Response to ethanol sclerotherapy varies with grade, with best response
netic contrast (Fig. 7-142) (684). It is important to recall, however, in grade 1 lesions, and worst response in grade 3 lesions.
that variable amounts of lymphatic malformation may result in T1
shortening of part of the venous malformation and the presence of Infantile and Congenital Hemangiomas
fluid-fluid levels (Fig. 7-143). These findings should not confuse Hemangiomas have already been discussed in the section on “Extraoc-
the reader but should strengthen the diagnosis of venolymphatic ular Orbital Masses.” Referred to as hemangiomas of infancy, juvenile
malformation. hemangiomas, or capillary hemangiomas, these are common masses,
Goyal et al. have proposed a classification for venous malforma- occurring in 4% to 10% of white infants. Females are affected more
tions. They define grade 1 as well-defined lesions ≤ 5 cm, grade 2A as often than males. They are generally small at birth, and then go through
well-defined lesions greater than 5 cm, grade 2B as ill-defined lesions a proliferative phase during the first year of life, after which they slowly
≤ 5 cm, and grade 3 as ill-defined lesions greater than 5 cm (721). involute (involuting phase from 1 to 7 years, followed by involuted

Barkovich_Chap07.indd 777 5/6/2011 9:17:55 PM


778 Pediatric Neuroimaging

FIG. 7-143. Venolymphatic malformation. A. Parasagittal T1-weighted image shows a sharply defined hyperintense mass (arrows) in the floor of the
mouth. This suggests a hemorrhagic or proteinaceous cyst. B. Coronal T1-weighted image shows that the lesion has multiple components separated by
septae. Components 1, 2, and 3 are hyperintense, suggesting hemorrhagic or proteinaceous cysts. Component 4, which is hypointense, may be solid or a cyst
filled with transudate. C. Axial T2-weighted image shows marked hyperintensity, which unfortunately does not aid in characterizing the lesion. D. Postcon-
trast coronal T1-weighted image (compare to B) shows enhancement of the septae but not of components 1 to 4. The lack of enhancement of component
4 suggests that it is cystic. E. Axial postcontrast fat-suppressed T1-weighted image at the level of component 4 shows a fluid-fluid level (arrows) confirming
its cystic nature.

phase after age 8 years) (722). Some authors postulate that they result after paramagnetic contrast administration (Figs. 7-144 and 7-145).
from emboli of placental cells, because their microvessels share an In older children, as the hemangioma involutes, more and more fat is
unusual set of antigens with placental microvasculature (GLUT1, LeY, seen within the mass, resulting in a more heterogeneous appearance
FcgRII, and merosin) (723). (Fig. 7-145). Curvilinear signal voids can almost always be identified;
Infantile hemangiomas are often multiple and can develop any- they are larger and more numerous in younger patients. As heman-
where on the body. They appear on CT as well-defined masses of giomas often involve the parotid gland, assessment of the location of
intermediate attenuation; they show intense uniform enhancement the facial nerve is important if surgical resection is being considered.
after intravenous administration of iodinated contrast. On MRI, In the last decade, several authors have described a group of
they are isointense to muscle on T1-weighted images, hyperintense hemangiomas that develop in utero, seem to have an almost equal
on T2-weighted images, and show uniform intense enhancement gender distribution, are usually solitary and have a predilection for

Barkovich_Chap07.indd 778 5/6/2011 9:17:56 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 779

FIG. 7-144. Infantile hemangioma. A. Axial T1-weighted image


shows a large soft tissue–intensity mass (arrows) in the lateral aspect
of the right neck. Multiple foci of signal void, representing blood ves-
sels, are present within the mass. B. Axial T2-weighted image with fat
suppression shows that the hemangioma becomes uniformly hyper-
intense. The margins of the mass are better seen on this image. C.
Postcontrast T1-weighted image with fat suppression shows uniform
enhancement.

specific cutaneous locations, on the head or on a limb near a joint (685, clefting and/or Supraumbilical raphe) (725–727) (see Chapter 6).
722–724). Some of these congenital hemangiomas involute rapidly Therefore, a thorough examination of these patients should always be
(usually within the first 14 months after birth) and are referred to as carefully performed on these patients and the brain and intracranial
rapidly involuting congenital hemangiomas, while others remain very and cervical vessels should be assessed when imaging studies of the
vascular and do not involute; the latter are referred to as noninvolut- head or neck are performed.
ing congenital hemagiomas (685,722–724). The precise relationship
among these congenital hemangiomas and infantile hemangiomas is Neurofibromas and Schwannomas
currently a matter of research (722). From an imaging perspective, it Neurofibromas and schwannomas have been discussed in Chapter 6
suffices to say that there are not yet any imaging characteristics that will and earlier in this chapter. It will briefly be repeated here for the conve-
definitively separate these lesions. nience of the reader. Neurofibromas are masses composed of fibroblasts
Hemangiomas of the face are commonly associated with associated and nerve cells; they occur most commonly in NF1, but may occur spo-
anomalies, a condition that has been called PHACES syndrome (Pos- radically. The neck is a common location for neurofibromas, with an
terior fossa malformations, facial Hemangiomas, Arterial anomalies, estimated occurrence of 25% to 30% in patients with NF1 (728). They
Cardiac anomalies and aortic coarctation, Eye anomalies, and Sternal are usually ovoid, sharply marginated masses that typically occur in

Barkovich_Chap07.indd 779 5/6/2011 9:17:57 PM


780 Pediatric Neuroimaging

FIG. 7-145. Involuting infantile hemangioma.


A. Axial T1-weighted image shows a well-defined
mass (arrows) in the posterior left neck. Multiple
foci of hyperintensity represent fat from involution
of the hemangioma. B. Axial T2-weighted image
with fat suppression shows heterogeneous hyper-
intensity of the mass. The heterogeneity results
from the fatty degeneration of portions of the mass.
C. Postcontrast T1-weighted image with fat sup-
pression shows nearly uniform enhancement.

the region of the carotid sheath, although they can develop anywhere to those in other regions of the body. They develop in the same loca-
that nerves run. Solitary neurofibromas have slightly greater signal tions as neurofibromas, being most common in and around the carotid
intensity than skeletal muscle on T1-weighted sequences. On FLAIR sheath but may be seen anywhere in the neck. They are well-defined,
and T2-weighted sequences, the periphery of the lesions tends to be usually ovoid, masses that may cross fascial planes. On CT, the masses
of high signal intensity with respect to muscle, whereas the center of are isodense with the neck muscles. On MRI, they are isointense with
the lesions is often of low signal intensity (see Fig. 7-146 and examples muscle on T1-weighted images and hyperintense compared with mus-
in Chapter 6) (729–731); this has been referred to as the “target sign” cle on T2-weighted and FLAIR images. Contrast enhancement is vari-
(732). The central area of decreased intensity is probably related to the able but usually homogeneous.
known dense central core of collagen within these lesions (729,731). Plexiform neurofibromas are seen only in the setting of NF1.
Collagen has a low mobile proton density and therefore is of low signal They are locally aggressive congenital lesions composed of tortu-
intensity on T2-weighted images. Enhancement is variable, but usually ous cords of Schwann cells, neurons, and collagen in an unorganized
heterogeneous, after contrast administration. intercellular matrix (733). They tend to progress along the nerve of
Schwannomas are tumors of Schwann cells that are most com- origin (usually small, unidentified nerves), causing distortion and
monly sporadic, but are seen with increased prevalence in NF1 and compression of the adjacent structures. On CT, plexiform neurofibro-
NF2. Imaging manifestations schwannomas of the neck are similar mas tend to be of low attenuation and generally do not enhance after

Barkovich_Chap07.indd 780 5/6/2011 9:17:59 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 781

FIG. 7-146. Neurofibroma. A. Sagittal T1-weighted image shows an


ovoid, well-defined mass (n) in the region of the carotid sheath. The
mass is isointense to skeletal muscle. B. Axial T2-weighted image with
fat suppression shows the mass (n) to be largely hyperintense with areas
of central hypointensity, presumably the result of collagenous stroma
within the tumor. C. Postcontrast coronal T1-weighted image with fat
suppression shows heterogeneous enhancement of the mass (n).

administration of intravenous contrast. On MRI, the curvilinear masses Teratoma


are heterogeneous, crossing facial boundaries throughout the neck Although neonatal head and neck teratomas are reportedly very rare
(Fig. 7-147). They are nearly always surrounded by fat, as nerves in the (735,736), 2 to 3 teratomas of the neonatal head and neck are seen per
neck mostly run through fat. They are mostly of low signal intensity as year in our department, accounting for up to 10% of all neonatal tera-
compared with brain on T1-weighted images and high signal inten- tomas (735,736). Most are detected prenatally by obstetrical sonogra-
sity on T2-weighted images (734). When fat suppression is used, high phy (performed for polyhydramnios due to difficulty in swallowing in
quality T2-weighted images nicely show the multiple swirling enlarged up to 55% of patients); thus, the neonatologists and pediatric surgeons
nerves coursing through the tissue (Fig. 7-147B). Enhancement after are usually prepared to deal with the airway compromise that is the
administration of paramagnetic contrast is variable, although at least a most common complication (up to 50%) of this tumor. As less than
portion of the tumor usually enhances (Fig. 7-147C). 5% of these tumors are malignant (735), early surgery to decompress

Barkovich_Chap07.indd 781 5/6/2011 9:17:59 PM


782 Pediatric Neuroimaging

FIG. 7-147. Plexiform neurofibroma. A. Axial T1-weighted image


shows a heterogeneous mass (arrows) in the posterior right neck.
B. Axial fat-suppressed T2-weighted image better shows the het-
erogeneity of the mass (arrows) and the hyperintense swirls of
thickened, curvilinear nerves that compose the tumor. C. Post-
contrast fat-suppressed T1-weighted image shows heterogeneous
enhancement of the tumor (arrows).

the airway results in a good prognosis for about 90% of affected neo- fatty and nonfatty, calcified and noncalcified), and bone (Fig. 7-148).
nates (737). Enhancement is heterogeneous and depends upon the composition of
Imaging is useful more for assessing the anatomic extent of the the tumor. The radiologic diagnosis is easy to establish in these cases.
tumor than for establishing a diagnosis. CT may be useful for identi- However, some teratomas are less mature and, as a result, more homo-
fication of fat or calcium, which increases confidence in the diagno- geneous. The differential diagnosis is broader in these patients. In
sis of teratoma. However, MRI is the most useful technique, allowing either case, it is important to look for airway compromise, involvement
imaging in multiple planes and best differentiating of tumor from of the pharynx and tongue, distortion of adjacent osseous structures
surrounding soft tissues. Well-differentiated teratomas are extremely (especially the mandible and hard palate), and extension into the skull
heterogeneous, with cystic components, soft tissue components (both base or even the middle cranial fossa.

Barkovich_Chap07.indd 782 5/6/2011 9:18:00 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 783

FIG. 7-148. Facial teratoma. A. Sagittal T1-weighted image shows a very heterogeneous mass (white arrows) arising from the facial bones. Note the
marked heterogeneity of the tumor, with areas of hyperintensity (presumably fat, black arrows), hypointensity, and isointensity compared to muscle.
B. Fat-suppressed postcontrast T1-weighted image shows suppression (black arrows) of the hyperintense areas, establishing that it is fat. The remainder of
the mass shows mild or moderate, heterogeneous enhancement.

The differential diagnosis varies with the appearance of the tumor the sternocleidomastoid muscle. If contrast is administered, variable
and the precise location. For predominantly cystic masses, the main dif- enhancement is seen. On MRI, the mass is isointense to normal muscle
ferential considerations are lymphatic malformation (cystic hygroma) on T1-weighted images, slightly hyperintense on T2-weighted images,
and meningocele. For solid, homogeneous lesions, the differential and shows similar enhancement to normal skeletal muscle (Fig. 7-149)
diagnosis includes dermoid/epidermoid, neurofibroma, hamartoma, (741). Occasionally, enhancement is heterogeneous; this should not
and rhabdomyosarcoma. For heterogeneous solid lesions, the differen- dissuade one from making the diagnosis of fibromatosis colli if all
tial includes craniopharyngioma, dermoid, encephalocele (is the brain other features are consistent with it.
exiting through the skull defect or being pushed away by an extracra-
nial mass), and sarcoma. If the lesion is largely fatty, dermoids and Rhabdomyosarcomas/Rhabdomyomas
lipomas are considerations, although large lipomas and large dermoids Rhabdomyosarcomas are the third most common primary child-
are uncommon in neonates. hood malignancy of the head and neck, following intracranial brain
tumors and retinoblastomas in frequency. They comprise 4% to 8% of
Cervical Lymphadenitis all malignant tumors in children less than 15-years-old; 43% of rhab-
Most patients with cervical lymphadenitis are not imaged, medical domyosarcomas arise before age 5 years, and 78% before age 12 years
therapy being the treatment of choice. Imaging is indicated if there is (742). Thirty-six percent of rhabdomyosarcomas arise in the head and
clinical suspicion of suppuration. Either sonography or CT can give a neck, with the orbit and nasopharynx/masticator space most com-
quick answer, ultrasound showing decreased central echogenicity and monly involved, followed by the paranasal sinuses and middle ear.
CT showing central hypodensity if central necrosis is present. When Intracranial involvement usually results from extension of extracranial
mass effect and surrounding inflammatory changes are minimal, tumor into the cranial vault through fissures and foramina. However,
atypical mycobacterial infections should be considered, particularly if very rarely, primary intracranial rhabdomyosarcomas do occur; they
the nodal masses are located in the parotid or submandibular regions cannot be differentiated radiologically from other primary intra-axial
(738). brain tumors (743–745). Histologically, the embryonal and alveo-
lar rhabdomyosarcomas are most common. Although most cases of
Fibromatosis Colli rhabdomyosarcoma appear to be sporadic in nature, some young chil-
Fibromatosis colli is a benign condition that is discovered primarily in dren (age, <3 years) have identifiable constitutional mutations of the
neonates and young infants. The infants typically come to attention p53 tumor suppressor gene and a possible hereditary predisposition to
between ages 2 and 4 weeks because of torticollis or a mass in the neck. cancer (746). Molecular criteria for rhabdomyosarcomas have recently
The mass is typically firm, nontender, and cannot be separated from been described. Approximately 60% of alveolar tumors have a FOXO1
the sternocleidomastoid muscle. There may be a history of breech pre- to PAX3 or PAX7 translocation (747); those lacking this characteris-
sentation or forceps delivery, which raises the question of trauma and tic have clinical features more similar to embryonal tumors. This may
hematoma as the cause of the mass (739,740). In the vast majority of have prognostic and therapeutic significance (746).
children, the mass spontaneously regresses during the first year of life. Rhabdomyomas are much less common than rhabdomyosarco-
The diagnosis of fibromatosis colli can be suggested by sonogra- mas, comprising only 2% of all tumors of striated muscle in the pediat-
phy, CT, or MRI. Sonography is usually diagnostic, showing focal or ric age group (748). The fetal type of rhabdomyoma is, by far, the most
diffuse enlargement of the sternocleidomastoid muscle with variable common type seen in children. These tumors consist of haphazardly
echogenicity. Often a hypoechoic rim (thought to represent com- arranged bundles of immature skeletal muscle fibers intermingled with
pressed normal muscle) is present. CT shows a focal enlargement of undifferentiated spindle-shaped but benign-appearing cells (749).

Barkovich_Chap07.indd 783 5/6/2011 9:18:01 PM


784 Pediatric Neuroimaging

FIG. 7-149. Fibromatosis colli in a 12-day-old infant. A. Axial


T1-weighted image shows a soft tissue intensity mass (arrows)
within the left sternocleidomastoid muscle. B. Axial T2-weighted
image shows that the mass (arrows) is heterogeneously hyperin-
tense compared with muscle. C. Postcontrast T1-weighted image
shows mild uniform enhancement of the mass approximately
equal to that of surrounding muscle.

Their most common locations are the head and neck, including the parameningeal group, consisting of tumors originating in the middle
oral cavity, retroauricular area, and orbit (748,750). Rhabdomyomas ear, nasopharynx/masticator space, paranasal sinuses, and nasal cavity,
are well-circumscribed, homogenous masses without necrosis or hem- is of particular importance from the neuroimaging perspective. Tumors
orrhage. Adjacent bones are remodeled but not invaded or destroyed. in these locations have the poorest prognosis because they commonly
MRI reveals low signal on both T1- and T2-weighted images; admin- invade the skull or spread intracranially via skull base foramina. Orbital
istration of intravenous contrast results in uniform enhancement rhabdomyosarcomas are most often extraconal in location, but may
(750,751). be both extraconal and intraconal; they most commonly develop in
Clinically, it is useful to classify rhabdomyosarcomas of the head the superonasal quadrant of the orbit. They most commonly spread
and neck by site of origin (752). Three major groups are identified: (a) by local invasion to the paranasal sinuses (20%), but can also spread
parameningeal, (b) orbital, and (c) other head and neck locations. The intracranially, typically growing through the orbital fissures into the

Barkovich_Chap07.indd 784 5/6/2011 9:18:02 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 785

FIG. 7-150. Rhabdomyosarcoma. A. Axial


contrast-enhanced CT through the mid-
dle of the orbits shows a mass near the
apex of the right orbit and extending
into the posterior ethmoidal air cells. The
mass extends through the superior orbital
fissure into the cavernous sinus (arrows).
B. Image 5 mm higher than A shows the
mass filling and expanding the right cav-
ernous sinus (arrows) and widening the
superior orbital fissure (open arrows).

cavernous sinuses and middle cranial fossa (753). Lymphatic spread has occurred is usually expanded. The extradural mass adjacent to
and hematogenous spread is rare other than in advanced disease. the expanded foramen usually demonstrates a wide base against the
On CT, rhabdomyosarcomas typically appear as irregular, ovoid, inner table of the skull; meningeal enhancement indicates a poor
well-circumscribed, homogeneous masses that are usually isodense prognosis (755). Usually, an extracranial or petrosal mass is also vis-
with muscle prior to contrast enhancement; they uniformly enhance ible (743–745,756). The MR findings are usually those of a mass that
after the infusion of contrast (Fig. 7-150). If calcification is present, it is isointense to muscle on T1-weighted images and hyperintense to
is typically due to destruction of adjacent bone (754). If intracranial muscle (hypointense to brain) on T2-weighted images (Figs. 7-151
extension occurs, the foramen or fissure through which the extension to 7-153). The tumors are often heterogeneous on T2-weighted images

FIG. 7-151. Nasopharyn-


geal rhabdomyosarcoma. A.
Sagittal T1-weighted image
shows the mass (arrows) in
the parapharyngeal region
that is isointense to muscle.
B. Axial T2-weighted image
shows the heterogeneous
mass (arrows) to be hyper-
intense compared to skel-
etal muscle.

Barkovich_Chap07.indd 785 5/6/2011 9:18:03 PM


786 Pediatric Neuroimaging

FIG. 7-151. (Continued) C. Postcontrast T1-weighted image shows homogeneous enhance-


ment (arrows) of the tumor.

FIG. 7-152. Nasopharyngeal rhabdomyosarcoma


with intracranial invasion. A. Sagittal T1-weighted
image shows a mass (arrows) growing upward from
the nasal cavity through the sphenoid bone and into
the inferior anterior cranial fossa. B. Axial T2-weighted
image shows the mass to be slightly hyperintense com-
pared with surrounding muscle. C. Postcontrast axial
T1-weighted image shows moderate homogeneous
enhancement of the mass (arrows).

Barkovich_Chap07.indd 786 5/6/2011 9:18:04 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 787

FIG. 7-153. Infratemporal rhabdomyosarcoma. A. Axial


T1-weighted image shows a soft tissue intensity mass (arrows)
in the left infratemporal fossa. B. Coronal T2-weighted
image with fat suppression shows the mass (arrows) is
slightly heterogeneous and is hyperintense compared with
muscle. C. Postcontrast T1-weighted image shows that the
mass (small arrows) enhances equally to muscle. No inva-
sion though the foramen ovale (large arrow) is seen.

due to presence of necrosis and hemorrhage. When tumors invade rhabdomyosarcoma can be suggested from imaging studies because it
the paranasal sinuses, MRI helps to differentiate tumor from retained is the most common primary extracranial tumor that invades the cra-
secretions, the latter being much more T2-hyperintense. Extension nial vault in childhood. Major differential diagnoses depend on the age
of tumor through a foramen or fissure is more easily visualized with of the patient and the location of the tumor; they include lymphoma,
MRI than with CT due to the lack of beam hardening artifact from the carcinoma (in teenagers), sarcomas, and juvenile nasal angiofibroma
skull base. However, erosion or infiltration of the skull base without (in teenage boys).
intracranial extension is difficult to appreciate on MRI and is better
evaluated with CT. Administration of paramagnetic contrast results in Nasopharyngeal Carcinoma
heterogeneous tumor enhancement, which sometimes gives a pattern Although nasopharyngeal carcinoma is rare in children with an inci-
of multiple mildly enhancing nodules separated by enhancing stroma; dence of less than 1% of all childhood cancers, carcinoma of the
this has been called the “botryoid sign” because of the resemblance to a nasopharynx has a predilection for adolescents and teenagers (759).
bunch of grapes (757). This sign may help in specificity of MR diagno- Indeed, although rhabdomyosarcoma and lymphoma are more com-
sis. Contrast enhancement also aids in the identification and localiza- mon causes of nasopharyngeal masses in younger children, nasopha-
tion of tumor that extends through the skull base foramina into the ryngeal carcinoma is more common in adolescents (759). These
extradural space (743,758). Contrast-enhanced MRI of the neck, there- tumors are typically of the nonkeratinizing variant and are associ-
fore, should always include fat suppression (Figs. 7-151, 7-153, and ated with Epstein-Barr virus (760). Affected children typically pres-
7-154), to aid in differentiation of enhancing tumor from fat. The neck ent with a neck mass, nasal obstruction, nasal bleeding and discharge,
should be imaged to screen for metastatic cervical lymphadenopathy. or fever of unknown origin. Unfortunately, these tumors are often
Although the CT and MR findings are not specific, the diagnosis of locally advanced by the time of diagnosis because the presentation is

Barkovich_Chap07.indd 787 5/6/2011 9:18:05 PM


788 Pediatric Neuroimaging

FIG. 7-154. Rhabdomyosarcoma with


intracranial invasion. Value of fat
suppression. A. Noncontrast axial
T1-weighted image shows a nasopha-
ryngeal mass (arrows) compressing and
laterally displacing the parapharyngeal
fat stripe. B. Postcontrast fat-suppressed
T1-weighted image 1 cm inferior to (A)
shows the heterogeneously enhanc-
ing mass (arrows) distinctly separate
from infratemporal and marrow fat.
C and D. Postcontrast coronal fat-
suppressed T1-weighted images show
enhancing nasopharyngeal tumor (white
arrows) growing intracranially through
a large hole (curved black arrows) in the
skull base, presumably the expanded
foramen ovale.

nonspecific and the clinical suspicion for tumor is usually low (761). invasion is recognized by loss of the normal high signal intensity of
Only after no response is seen to several courses of antibiotics is biopsy marrow fat on T1-weighted images and by development of high signal
obtained and diagnosis made. Cervical lymphadenopathy, a common intensity within the marrow space on T2-weighted images (762). CT
cause of presentation of nasopharyngeal carcinoma in adults, is so may show erosion of the cortical bone or widening of the petroclival
common in children that it does not cause sufficient alarm among par- fissure. Erosion of the surrounding soft tissue structures is most easily
ents of physicians for further investigation. recognized by effacement of the fat planes in the parapharyngeal space
The main differentiation in diagnosing nasopharyngeal carcinoma that separate the pharyngeal mucosa from surrounding muscles.
from the benign adenoidal masses that are so common in childhood
and adolescence is asymmetry. Benign adenoidal tissue is nearly always Juvenile Angiofibromas
symmetric, whereas nasopharyngeal carcinoma is almost always asym- Juvenile angiofibromas are highly vascular, locally invasive, histo-
metric (762). Another feature distinguishing carcinoma is local spread logically benign tumors that arise in the sphenopalatine foramen,
of the mass, typically posteriorly through the pharyngobasilar fascia nasopharynx, or posterior nasal cavity of adolescent boys. Local spread
towards the clivus or laterally into the parapharyngeal space. Clival into the sphenoid sinus, pterygopalatine fossa, infratemporal fossa,

Barkovich_Chap07.indd 788 5/6/2011 9:18:06 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 789

FIG. 7-155. A. Axial contrast enhanced CT image shows a right nasal mass (m) that extends posteriorly into and destroys the right pterygoid process (black
arrowheads) and extends laterally (black arrow) through the sphenopalatine foramen into the pterygopalatine fossa. B. Parasagittal reformation shows the
enhancing mass (arrows) in the pterygopalatine fossa.

orbit, and middle cranial fossa is often seen by the time of presentation. the nasopharynx into the sphenoid sinus is common. Homogeneous
Affected patients typically present with nasal obstruction and recur- enhancement is seen after intravenous infusion of iodinated contrast
rent epistaxis. In advanced cases, proptosis or cranial neuropathies may (Fig. 7-155) (743,763,764). MRI shows a lobulated mass that is hypoin-
develop (743,763,764). These tumors have an increased frequency in tense to isointense compared with muscle on T1-weighted images;
patients with familial adenomatous polyposis (765). although most are isointense to slightly hyperintense compared with
CT typically shows a sharply marginated, homogeneous soft tissue muscle on T2-weighted images (Figs. 7-156 to 7-158), the lesions are
mass that arises in the posterolateral wall of the nasal cavity, spreads occasionally very hyperintense. Marked, fairly uniform enhancement
anteriorly into the ipsilateral nasal cavity and the nasopharynx and is seen after intravenous administration of paramagnetic contrast
laterally into the pterygopalatine fossa, causing anterior bowing of the (Figs. 7-156 to 7-158). Punctate and curvilinear regions of low signal
posterior wall of the maxillary sinus. Expansion through the roof of intensity can be seen on both T1- and T2-weighted images, representing

FIG. 7-156. Small juvenile nasal angiofibroma. A. Axial T1-weighted image shows a muscle-intensity mass (large white arrows) arising in the posterior
nasal cavity and extending laterally to invade the pterygoid process (asterisk). Note that the posterior wall of the left maxillary sinus (small white arrows) is
anteriorly displaced. B. Fat-suppressed axial T2-weighted image shows the nasal mass (large white arrows) growing laterally (small white arrow) into the left
pterygoid process and pterygopalatine fossa.

Barkovich_Chap07.indd 789 5/6/2011 9:18:08 PM


790 Pediatric Neuroimaging

FIG. 7-156. (Continued) C. Coronal fat-suppressed T1-weighted image shows the brightly
enhancing mass extending from the nasal cavity through the sphenopalatine foramen
(at the large arrow) into the pterygomaxillary fissure and pterygopalatine fossa (small
arrows).

FIG. 7-157. Classic juvenile nasal angiofibroma. A. Axial


T1-weighted image shows a large mass arising in the posterior
nasal cavity and extending laterally through the left spheno-
palatine foramen (large arrow) into the pterygopalatine fossa.
Note that the normal fat in the pterygopalatine fossa seen on the
right (small arrows) is replaced by tumor on the left. B. Axial
T2-weighted image with fat suppression shows a mass that is
slightly hyperintense to skeletal muscle that is centered in the
posterior nasal cavity. The mass extends posteriorly (small white
arrows) into the sphenoid sinus and sphenoid bone, laterally
through the left sphenopalatine foramen into the pterygopalatine
fossa (small white arrowheads), and anteriorly (large arrowheads)
into the nasal cavity. Small hypointensities represent voids from
flow in blood vessels within the tumor. The large white arrows
point to retained secretions in the right ear due to compression
of the Eustachian tube. C. Postcontrast T1-weighted image with
fat suppression shows diffuse, somewhat heterogeneous enhance-
ment. Arrows show extension of tumor into the skull base.

Barkovich_Chap07.indd 790 5/6/2011 9:18:09 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 791

FIG. 7-158. Juvenile nasal angiofibroma


with skull base invasion. A. Axial T2-weighted
image with fat suppression shows a mass that
is slightly hyperintense to skeletal muscle
that is centered in the posterior nasal cavity.
The mass extends posteriorly (black arrows)
into the clivus, and bilaterally through the
sphenopalatine foramen into the pterygo-
palatine fossae (small white arrows). Small
hypointensities represent voids from flow
in blood vessels within the tumor. The large
white arrow points to retained secretions in
the right maxillary sinus. Retained fluid is
present in the right ear due to compression
of the Eustachian tube. B. Postcontrast coro-
nal T1-weighted image with fat saturation
shows intense, fairly uniform enhancement.
Black arrows show extension of tumor into
the skull base.

the prominent tumor vessels that are characteristic of these tumors; these characteristics have been present in the same patient in different
however, tumor vessels are not always seen by MRI and their absence in areas of tumor. MRI shows a soft tissue mass that is isointense to mus-
a mass otherwise typical for juvenile angiofibroma should not negate cle on T1-weighted images and isointense to hyperintense to muscle
the diagnosis (743,764). The key findings in making this diagnosis are on T2-weighted images. T1 shortening from melanin is not reported in
the age and sex of the patient (teenage boys are by far the group most most cases. Rarely, melanotic neuroectodermal tumors of infancy can
commonly affected) and the location in the posterior nasal cavity with originate intracranially, presumably from mesodermal elements within
extension through the sphenopalatine foramen into the pterygopala- the stroma of the choroid plexus. These tumors are indistinguishable
tine fossa. Treatment of choice is by intravascular techniques; details from the more common neoplasms that arise in those locations by
of intravascular diagnosis and therapy are discussed in Chapter 12. radiological criteria (766,767).

Melanotic Neuroectodermal Tumors of Infancy Neuroblastoma


Melanotic neuroectodermal tumors of infancy are tumors of mesoder- Neuroblastoma of the neck is discussed in Chapter 10 in the section of
mal origin that typically involve the skull, dura, or face. They often “Extramedullary Tumors.”
occur adjacent to cranial sutures and may be initially mistaken for der-
moids. Patients typically present with a mass and discoloration of adja- Fibrous Dysplasia of the Skull Base
cent skin. The tumors secrete high levels of vanillylmandelic acid and Fibrous dysplasia is mentioned in this section only because it can mimic
homovanillic acid in the urine, which can be used to make a preopera- skull base and head-and-neck tumors, such as rhabdomyosarcomas
tive diagnosis and to identify tumor recurrence at an early stage. Local and juvenile nasal angiofibromas on MRI (Fig. 7-159). Affected bone
resection is often curative (766,767). CT shows a soft tissue mass that is expanded and will enhance dramatically. A noncontrast CT scan
usually arises in the bones of the face. The bone can show expansion, (Fig. 7-160) with bone algorithm will show the classic “ground glass”
hyperostosis, erosion, or osteogenesis; in some reported cases, all of appearance of fibrous dysplasia, confirming the proper diagnosis.

FIG. 7-159. Fibrous dysplasia of the orbitosphenoid region. A. Sagittal T1-weighted image shows expanded, hypointense, unpneumatized sphenoid bone.
B. Coronal T2-weighted image shows the sphenoid bone (arrows) to be enlarged and markedly hypointense. Compare with Figure 7-158, but note that
there is no nasopharyngeal soft tissue mass in this case. C. After administration of paramagnetic contrast, coronal T1-weighted image shows the expanded
sphenoid bone enhancing dramatically, as is typical for fibrous dysplasia.

Barkovich_Chap07.indd 791 5/6/2011 9:18:10 PM


792 Pediatric Neuroimaging

FIG. 7-160. CT of fibrous dysplasia. A. Axial bone algorithm


CT image of temporal bone shows a diffusely expanded bone,
with attenuation similar to diploic space, lower than that of cor-
tical bone. This appearance is called a “ground glass” appear-
ance. B and C. Different patient. Axial bone algorithm images
through the orbit and supraorbital regions show expanded bone
(arrows) in the lateral wall of the orbit (B) and the supraorbital
portion of the frontal bone (C). Both areas have a “ground glass
appearance.”

References 7. Farwell JR, Dohrmann GJ, Flannery JT. Central nervous system tumors in
children. Cancer 1977;40:3123–3132.
1. Dirks PB. Brain tumour stem cells: the undercurrents of human brain cancer 8. Amador LV. Brain Tumors in the Young. Springfield: Charles C. Thomas,
and their relationship to neural stem cells. Phil Trans Roy Society B: Biol Sci 1983.
2008;363:139–152. 9. Rosemberg S, Fujiwara D. Epidemiology of pediatric tumors of the nervous
2. Tadmor R, Harwood-Nash DC, Scotti G, et al. Intracranial neoplasms in system according to the WHO 2000 classification: a report of 1,195 cases
children: the effect of computed tomography on age distribution. Radiology from a single institution. Childs Nerv Syst 2005;21:940–944.
1982;145:371–373. 10. Fitz CR. Neoplastic diseases. In: Gonzales CF, Grossman CB, Masdeu JC,
3. Young JL, Heise HW, Silverberg E, et al. Cancer Incidence, Survival and Mor- eds. Head and Spine Imaging. New York: Wiley and Sons, 1985:385–410.
tality for Children Under 15 Years of Age. New York: American Cancer Society, 11. Naidich TP, Zimmerman RA. Primary brain tumors in children. Semin
1978. Roentgenol 1984;19:100–114.
4. Merchant TE, Pollack IF, Loeffler JS. Brain tumors across the age spectrum: 12. Hooper R. Intracranial tumours in childhood. Childs Brain 1975;1:
biology, therapy, and late effects. Semin Radiat Oncol 2010;20:58–66. 136–140.
5. Lannering B, Sandström PE, Holm S, et al. Classification, incidence and sur- 13. Radkowski MA, Naidich TP, Tomita T, Byrd SE, McLone DG. Neonatal brain
vival analyses of children with CNS tumours diagnosed in Sweden 1984– tumors: CT and MR findings. J Comput Assist Tomogr 1988;12:10–20.
2005. Acta Pædiatrica 2009;98:1620–1627. 14. Tadmor R, Harwood-Nash DCF, Savoiardo M. Brain tumors in the first two
6. Gjerris F, Agerlin N, Børgeson WE, et al. Epidemiology and prognosis in chil- years of life: CT diagnosis. Am J Neuroradiol 1980;1:411–417.
dren treated for intracranial tumors in Denmark 1960–1984. Childs Nerv Syst 15. Tomita T, McLone DG. Brain tumors during the first twenty-four months
1998;14:302–311. of life. Neurosurgery 1985;17:913–919.

Barkovich_Chap07.indd 792 5/6/2011 9:18:12 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 793

16. Nomura S, Nishizaki T, Yamashita K, Ito H. Pediatric brain tumors in a 40. Gerstner ER, Sorensen AG, Jain RK, Batchelor TT. Advances in neuroimag-
10-year period from 1986 to 1995 in Yamaguchi prefecture: epidemiol- ing techniques for the evaluation of tumor growth, vascular permeability,
ogy and comparison with adult brain tumors. Pediatr Neurosurg 1998;28: and angiogenesis in gliomas. Curr Opin Neurol 2008;21:728–735.
130–134. 41. Law M, Oh S, Babb JS, et al. Low-grade gliomas: dynamic susceptibility-
17. Kaderali Z, Lamberti-Pasculli M, Rutka J. The changing epidemiology of weighted contrast-enhanced perfusion MR imaging, Äîprediction of
paediatric brain tumours: a review from the Hospital for Sick Children. patient clinical response. Radiology 2006;238:658–667.
Childs Nerv Syst 2009;25:787–793. 42. Barrett T, Brechbiel M, Bernardo M, Choyke PL. MRI of tumor angiogen-
18. CBTRUS. Statistical report: primary brain tumors in the United states, esis. J Magn Reson Imaging 2007;26:235–249.
2000–04. In: Central Brain Tumor Registry of the United States, Hinsdale, 43. Mills SJ, Soh C, O’Connor JPB, et al. Enhancing fraction in glioma and
IL. 2008. its relationship to the tumoral vascular microenvironment: a dynamic
19. Fangusaro J. Pediatric high grade gliomas and diffuse intrinsic pontine contrast-enhanced MR imaging study. Am J Neuroradiol 2010;31:726–731.
gliomas. J Child Neurol 2009;24:1409–1417. 44. Panigrahy A, Blüml S. Neuroimaging of pediatric brain tumors: from
20. Rineer J, Schreiber D, Choi K, Rotman M. Characterization and outcomes basic to advanced magnetic resonance imaging (MRI). J Child Neurol
of infratentorial malignant glioma: a population-based study using the 2009;24:1343–1365.
surveillance epidemiology and end-results database. Radiother Oncol 45. Panigrahy A, Krieger MD, Gonzalez-Gomez I, et al. Quantitative short echo
2010;95:321–326. time 1h-mr spectroscopy of untreated pediatric brain tumors: preoperative
21. Armstrong GT, Liu Q, Yasui Y, et al. Long-term outcomes among adult sur- diagnosis and characterization. Am J Neuroradiol 2006;27:560–572.
vivors of childhood central nervous system malignancies in the childhood 46. Panigrahy A, Nelson M, Blüml S. Magnetic resonance spectroscopy in
cancer survivor study. J Natl Cancer Inst 2009;101:946–958. pediatric neuroradiology: clinical and research applications. Pediatr Radiol
22. Huber J, Bradley K, Spiegler B, Dennis M. Long-term effects of transient 2010;40:3–30.
cerebellar mutism after cerebellar astrocytoma or medulloblastoma tumor 47. Tzika AA, Vigneron DB, Dunn RS, Nelson SJ, Ball WSJ. Intracranial tumors
resection in childhood. Childs Nerv Syst 2006;22:132–138. in children: small single-voxel proton MR spectroscopy using short- and
23. Palmer SL. Neurodevelopmental impact on children treated for medullo- long-echo sequences. Neuroradiology 1996;38:254–263.
blastoma: a review and proposed conceptual model. Dev Disabil Res Rev 48. Warren KE. NMR spectroscopy and pediatric brain tumors. Oncologist
2008;14:203–210. 2004;9:312–318.
24. Roncadin C, Dennis M, Greenberg M, Spiegler B. Adverse medical events 49. Desprechins B, Stadnik T, Koerts G, Shabana W, Breucq C, Osteaux M.
associated with childhood cerebellar astrocytomas and medulloblastomas: Use of diffusion-weighted MR imaging in differential diagnosis between
natural history and relation to very long-term neurobehavioral outcome. intracerebral necrotic tumors and cerebral abscesses. Am J Neuroradiol
Childs Nerv Syst 2008;24:995–1002. 1999;20:1252–1257.
25. Turner CD, Rey-Casserly C, Liptak CC, Chordas C. Late Effects of therapy 50. Guo A, Provenzale J, Cruz L, Jr, Petrella J. Cerebral abcesses: investiga-
for pediatric brain tumor survivors. J Child Neurol 2009;24:1455–1463. tion using apparent diffusion coefficient maps. Neuroradiology 2001;43:
26. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a 370–374.
hierarchy that originates from a primitive hematopoietic cell. Nat Med 51. Grand S, Passaro G, Ziegler A, et al. Necrotic tumor versus brain abscess:
1997;3:730–737. importance of amino acids detected at 1H MR spectroscopy — initial
27. Vescovi AL, Galli R, Reynolds BA. Brain tumour stem cells. Nat Rev Cancer results. Radiology 1999;213:785–793.
2006;6:425–436. 52. Burtscher I, Holtas S. In vivo proton MR spectroscopy of untreated and
28. Hadjipanayis CG, Van Meir EG. Brain cancer propagating cells: biology, treated brain abscesses. Am J Neuroradiol 1999;20:1049–1053.
genetics and targeted therapies. Trends Molec Med 2009;15:519–530. 53. Dev R, Gupta R, Poptani H, Roy R, Sharma S, Husain M. Role of in vivo
29. Clark PA, Treisman DM, Ebben J, Kuo JS. Developmental signaling path- proton magnetic resonance spectroscopy in the diagnosis and managment
ways in brain tumor-derived stem-like cells. Dev Dyn 2007;236:3297– of brain abscesses. Neurosurgery 1998;42:37–42.
3308. 54. Cha S, Knopp E, Johson G, Wetzel S, Litt A, Zagzag D. Intracranial mass
30. Jandial R, Levy M, Snyder E. Brain tumor stem cell and the tumor microen- lesions: dynamic contrast-enhanced susceptibility-weighted echo-planar
vironment. Neurosurg Focus 2008;24:E27. perfusion MR imaging. Radiology 2002;223:11–29.
31. Gilbertson RJ, Gutmann DH. Tumorigenesis in the brain: location, loca- 55. Law M, Meltzer D, Cha S. Spectroscopic magnetic resonance imaging of a
tion, location. Cancer Res 2007;67:5579–5582. tumefactive demyelinating lesion. Neuroradiology 2002;44:986–989.
32. Sharma MK, Mansur DB, Reifenberger G, et al. Distinct genetic signatures 56. Saindane A, Cha S, Law M, Xue X, Knopp E, Zagzag D. Proton MR
among pilocytic astrocytomas relate to their brain region origin. Cancer spectroscop of tumefactive demyelinating lesions. Am J Neuroradiol
Res 2007;67:890–900. 2002;23:1378–1386.
33. Taylor MD, Poppleton H, Fuller C, et al. Radial glia cells are candidate stem 57. Cianfoni A, Niku S, Imbesi SG. Metabolite findings in tumefactive demy-
cells of ependymoma. Cancer Cell 2005;8:323–335. elinating lesions utilizing short echo time proton magnetic resonance spec-
34. Medina LS, Pinter JD, Zurakowski D, Davis RG, Kuban K, Barnes PD. Chil- troscopy. Am J Neuroradiol 2007;28:272–277.
dren with headache: clinical predictors of surgical space-occupying lesions 58. Cha S, Pierce S, Knopp E, et al. Dynamic contrast-enhanced T2*-weighted
and the role of neuroimaging. Radiology 1997;202:819–824. MR imaging of tumefactive demyelinating lesions. Am J Neuroradiol
35. Kramer E, Rafto S, Packer R, Zimmerman RA. Comparison of myelography 2001;22:1109–1116.
with CT follow-up versus gadolinium MRI for subarachnoid metastatic 59. Giussani C, Poliakov A, Ferri RT, et al. DTI fiber tracking to differenti-
disease in children. Neurology 1991;41:46–50. ate demyelinating diseases from diffuse brain stem glioma. NeuroImage
36. Blews DE, Wang H, Kumar AJ, Robb PA, Phillips PC, Bryan RN. Intra- 2010;52:217–223.
dural spinal metastases in pediatric patients with primary intracranial 60. Pillai JJ, Zaca D, Choudhri A. Clinical impact of integrated physiologic
neoplasms: Gd-DTPA enhanced MR vs CT myelography. J Comput Assist brain tumor imaging. Technol Cancer Res Treat 2010;9:359–380.
Tomogr 1990;14:730–735. 61. Nilsson D, Starck G, Ljungberg M, et al. Intersubject variability in the ante-
37. Barkovich AJ. Neuroimaging of pediatric brain tumors. In: Berger MS, ed. rior extent of the optic radiation assessed by tractography. Epilepsy Res
Pediatric Neuro-Oncology, vol 3. Philadelphia: Saunders, 1992:739–770. 2007;77:11–16.
38. Cha S. Update on brain tumor imaging: from anatomy to physiology. Am J 62. Helton KJ, Phillips NS, Khan RB, et al. Diffusion tensor imaging of
Neuroradiol 2006;27:475–487. tract involvement in children with pontine tumors. Am J Neuroradiol
39. Inoue T, Ogasawara K, Beppu T, Ogawa A, Kabasawa H. Diffusion tensor 2006;27:786–793.
imaging for preoperative evaluation of tumor grade in gliomas. Clin Neurol 63. Lui Y, Law M, Chacko-Mathew J, et al. Brainstem corticospinal tract dif-
Neurosurg 2005;107:174–180. fusion tensor imaging in patients with primary posterior fossa neoplasms

Barkovich_Chap07.indd 793 5/6/2011 9:18:12 PM


794 Pediatric Neuroimaging

stratified by tumor type: a study of association with motor weakness and 88. Stanley P, Senac MO, Jr, Segall HD. Intraspinal seeding from intracranial
outcome. Neurosurgery 2007;61:1199–1208. tumors in children. Am J Roentgenol 1985;144:157–161.
64. Nilsson D, Rutka J, Snead O, Raybaud C, Widjaja E. Preserved structural 89. Packer RJ, Siegel KR, Sutton LN, et al. Leptomeningeal dissemination
integrity of white matter adjacent to low-grade tumors. Childs Nerv Syst of primary central nervous system tumors of childhood. Ann Neurol
2008;24:313–320. 1985;18:218–221.
65. Salmela MB, Cauley KA, Andrews T, Gonyea JV, Tarasiewicz I, Filippi CG. 90. Tomita T, McLone DG. Spontaneous seeding of medulloblastoma; results
Magnetic resonance diffusion tensor imaging of the optic nerves to guide of CSF cytology and arachnoid biopsy from the cisterna magna. Neurosur-
treatment of pediatric suprasellar tumors. Pediatr Neurosurg 2009;45: gery 1983;12:265–267.
467–471. 91. Brat DJ, Parisi JE, Kleinschmidt-DeMasters BK, et al. Surgical neuropa-
66. Smith J, Cha S, Mayo M, et al. Serial diffusion-weighted magnetic reso- thology update: a review of changes introduced by the WHO classification
nance imaging in cases of glioma: distinguishing tumor recurrence from of tumours of the central nervous system, 4th ed. Arch Pathol Lab Med
postresection injury. J Neurosurg 2005;103:428–438. 2008;132:993–1007.
67. Elster AD, Dipersio DA. Cranial postoperative site: assessment with con- 92. Giangaspero F, Perilongo G, Fondelli M, et al. Medulloblastoma with
trast-enhanced MR imaging. Radiology 1990;174:93–98. extensive nodularity: a variant with favorable prognosis. J Neurosurg 1999;
68. Hudgins PA, Davis PC, Hoffman JC, Jr. Gadopentetate dimeglumine 91:971–977.
enhanced MR imaging in children following surgery for brain tumor: spec- 93. Suresh TN, Santosh V, Yasha TC, et al. Medulloblastoma with extensive
trum of meningeal findings. Am J Neuroradiol 1991;12:301–308. nodularity: a variant occurring in the very young—clinicopathological and
69. Sundgren PC. MR spectroscopy in radiation injury. Am J Neuroradiol immunohistochemical study of four cases. Childs Nerv Syst 2004;20:55–60.
2009;30:1469–1476. 94. Farwell JR, Dohrmann GJ, Flannery JT. Medulloblastoma in childhood: an
70. Sundgren PC, Fan X, Weybright P, et al. Differentiation of recurrent brain epidemiological study. J Neurosurg 1984;61:657–664.
tumor versus radiation injury using diffusion tensor imaging in patients 95. Al-Mefty O, Jinkins JR, El-Senoussi M, et al. Medulloblastomas: a review
with new contrast-enhancing lesions. Magn Reson Imaging 2006;24:1131– of modern management with a report on 75 cases. Surg Neurol 1985;24:
1142. 606–624.
71. Gaensler EH, Dillon WP, Edwards MSB, Larson DA, Rosenau W, Wilson 96. Koeller K, Rushing E. From the archives of the AFIP: medulloblastoma:
CB. Radiation-induced telangiectasia in the brain similates cryptic vascular a comprehensive review with radiologic-pathologic correlation. Radio-
malformations at MR imaging. Radiology 1994;193:629–636. graphics 2003;23:1613–1637.
72. Bailey P, Cushing H. Medulloblastoma cerebelli. A common type of 97. Malheiros S, Carrete H, Jr, Stavale J, et al. MRI of medulloblastoma in
midcerebellar glioma of childhood. Arch Neurol Psychiatry 1925;14: adults. Neuroradiology 2003;45:463–467.
192–224. 98. Cervoni L, Caruso R, Celli P, Trillo G, Gagliaridi FM. Medulloblastoma
73. Louis D, Ohgaki H, Wiestler O, et al. The 2007 WHO classification of dell’adulto. Revisione di nove casi studiati con la RM. Rivista di Neurora-
tumours of the central nervous system. Acta Neuropathologica 2007;114: diologia 1996;9:91–94.
97–109. 99. Sandhu A, Kendall B. Computed tomography in management of medullo-
74. Gilbertson RJ, Ellison DW. The origins of medulloblastoma subtypes. Annu blastomas. Neuroradiology 1987;29:444–452.
Rev Pathol Mech Dis 2008;3:341–365. 100. Nelson MD, Diebler C, Forbes WSC. Pediatric medulloblastoma: atypi-
75. Rubenstein LJ. Tumors of the Central Nervous System. Atlas of Tumor Pathol- cal CT features at presentation in the SIOP II trial. Neuroradiology 1991;
ogy, 2nd series, Fasicle 6. Washington, D.C.: Armed Forces Institute of 33:140–142.
Pathology, 1972. 101. Stavrou T, Dubovsky E, Reaman G, Goldstein A, Vezina G. Intracranial
76. Russell DS, Rubenstein LJ. Pathology of Tumors of the Nervous System. calcifications in childhood medulloblastoma: relation to nevoid basal cell
Baltimore: Williams & Wilkins, 1989. carcinoma syndrome. Am J Neuroradiol 2000;21:790–794.
77. Rickert CH, Paulus W. Epidemiology of central nervous system tumors in 102. Rumboldt Z, Camacho DLA, Lake D, Welsh CT, Castillo M. Apparent
childhood and adolescence based on the new WHO classification. Childs diffusion coefficients for differentiation of cerebellar tumors in children.
Nerv Syst 2001;17:503–511. Am J Neuroradiol 2006;27:1362–1369.
78. Roberts RO, Lynch CF. Medulloblastoma: a population-based study of 103. Auer DP, Elbel GK, Jones RA. MR - Diffusion bei Hirntumoren. Klin Neu-
532 cases. J Neuropathol Exp Neurol 1991;50:134–144. roradiol 1998;8:137–142.
79. Maleci A, Cervoni L, Delfini R. Medulloblastoma in children and in adults: 104. Klisch J, Husstedt H, Hennings S, Velthoven V, Pagenstecher A, Schu-
a comparative study. Acta Neurochir 1992;119:62–67. macher M. Supratentorial primitive neuroectodermal tumors: diffusion
80. Farwell J, Flannery JT. Adult occurrence of medulloblastoma. Acta Neuro- weighted MRI. Neuroradiology 2000;42:393–398.
chir (Wien) 1987;86:1–5. 105. Wilke M, Eidenscink A, Muller-Weihrich S, Auer D. MR diffusion imaging
81. Albright L. Posterior fossa tumors. In: Berger MS, ed. Pediatric Neurooncol- and 1H spectroscopy in a child with medulloblastoma. A case report. Acta
ogy, vol 3. Philadelphia: Saunders, 1992:881–891. Radiol 2001;42:39–42.
82. Alston R, Newton R, Kelsey A, et al. Childhood medulloblastoma in north- 106. Meyers SP, Kemp SS, Tarr RW. MR imaging features of medulloblastomas.
west England 1954 to 1997: incidence and survival. Dev Med Child Neurol Am J Roentgenol 1992;158:859–865.
2003;45:308–314. 107. Harris L, Davies N, MacPherson L, et al. The use of short-echo-time 1H
83. David KD, Casey ATH, Hayward RD, Harkness WFJ, Phipps K, Wade MRS for childhood cerebellar tumours prior to histopathological diagno-
AM. Medulloblastoma: is the 5-year survival rate improving? A review of sis. Pediatr Radiol 2007;37:1101–1109.
80 cases from a single institution. J Neurosurg 1997;86:13–21. 108. Girard N, Wang ZJ, Erbetta A, et al. Prognostic value of proton MR
84. Perek D, Perek-Polnik M, Drogosiewicz M, Dembowska-Baginska B, Rosz- spectroscopy of cerebral hemisphere tumors in children. Neuroradiology
kowski M, Barszcz S. Risk factors of recurrence in 157 MB/PNET patients 1998;40:121–125.
treated in one institution. Childs Nerv Syst 1998;14:582–586. 109. Tzika AA, Vajapeyam S, Barnes PD. Multivoxel proton MR spectroscopy
85. Packer R, Rood B, MacDonald T. Medulloblastoma: present concepts of and hemodynamic MR imaging of childhood brain tumors: Preliminary
stratification into risk groups. Pediatr Neurosurg 2003;39:60–67. observations. Am J Neuroradiol 1997;18:203–218.
86. Park TS, Hoffman HJ, Hendrick EB, et al. Medulloblastoma: clinical pre- 110. Moreno-Torres A, Martinez-Pérez I, Baquero M, et al. Taurine detection
sentation and management; experience at the Hospital for Sick Children. by proton magnetic resonance spectroscopy in medulloblastoma: contri-
Toronto, 1950–1980. J Neurosurg 1983;58:543–552. bution to noninvasive differential diagnosis with cerebellar astrocytoma.
87. George RE, Laurent JP, McCluggage CW, Cheek WR. Spinal metastasis in Neurosurgery 2004;55:824–829.
primitive neuroectodermal tumors (medulloblastoma) of the posterior 111. Freilich RJ, Krol G, DeAngelis LM. Neuroimaging and cerebrospinal
fossa: evaluation with CT myelography and correlation with patient age fluid cytology in the diagnosis of leptomeningeal metastasis. Ann Neurol
and tumor differentiation. Pediatr Neurosci 1986;12:157–160. 1995;38:51–57.

Barkovich_Chap07.indd 794 5/6/2011 9:18:13 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 795

112. Shaw DW, Weinberger E, Brewer DK, Geyer JR, Berger MS, Blaser SI. Spi- 136. Gjerris F, Klinken L. Long-term prognosis in children with benign cerebellar
nal subdural enhancment after suboccipital craniectomy. Am J Neurora- astrocytoma. J Neurosurg 1978;49:179–184.
diol 1996;17:1373–1377. 137. Villarejo F, Belinchon de Diego JM, Gomez de la Riva A. Prognosis of cer-
113. Wiener MD, Boyko OB, Friedman HS, Hockenberger B, Oakes WJ. False ebellar astrocytomas in children. Childs Nerv Syst 2008;24:203–210.
positive spinal MR findings for subarachnoid spread of primary CNS 138. Koeller K, Rushing E. From the archives of the AFIP: pilocytic astrocytoma:
tumor in postoperative pediatric patients. Am J Neuroradiol 1990;11: radiologic-pathologic correlation. Radiographics 2004;24:1693–1708.
1100–1103. 139. Pencalet P, Maixner W, Sainte-Rose C, et al. Benign cerebellar astrocy-
114. Meyers SP, Wildenhain SL, Chang J-K, et al. Postoperative evaluation for tomas in children. J Neurosurg 1999;90:265–273.
disseminated medulloblastoma involving the spine: contrast-enhanced 140. Somaza S, Kondziolka D, Lunsford LD, Flickinger JC, Bissonette D,
MR findings, CSF cytologic analysis, timing of disease occurrence, and Albright AL. Early outcomes after stereotactic radiosurgery for growing
patient outcomes. Am J Neuroradiol 2000;21:1757–1765. pilocytic astrocytomas in children. Pediatr Neurosurg 1996;25:109–115.
115. Tarbell NJ, Loeffler JS, Silver B, et al. The change in patterns of relapse in 141. Ringertz N, Nordenstam HJ. Cerebellar astrocytoma. Neuropathol Exp
medulloblastoma. Cancer 1991;68:1600–1604. Neurol 1951;10:343–367.
116. Eberhart C, Cohen K, Tihan T, Goldthwaite P, Burger P. Medulloblastomas 142. Gol A, McKissock W. The cerebellar astrocytomas: a report on 98 verified
with systemic metastases: evaluation of tumor histopathology and clinical cases. J Neurosurg 1959;16:287–296.
behavior in 23 patients. J Pediatr Hematol Oncol 2003;25:198–203. 143. Burger P, Scheithauer B, Paulus W, Szymas J, Giannini C, Kleihues P. Pilo-
117. Biggs PJ, Garen PD, Powers JM, et al. Malignant rhabdoid tumor of the cytic astrocytoma. In: Kleihues P, Cavenee W, eds. Pathology and Genetics
central nervous system. Hum Pathol 1987;18:332–337. of Tumors of the Nervous System. Lyon: IARC, 2000:45–51.
118. Rorke LB, Packer RJ, Biegel JA. Central nervous system atypical teratoid/ 144. White J, Piepgras D, Scheithauer B, Parisi J. Rate of spontaneous hemor-
rhabdoid tumors of infancy and childhood: definition of an entity. J Neu- rhage in histologically proven cases of pilocytic astrocytoma. J Neurosurg
rosurg 1996;85:56–65. 2008;108:223–226.
119. Bambakidis N, Robinson S, Cohen M, Cohen A. Atypical teratoid/ 145. Lee C, Huh J, Sim K-B, Kim Y. Cerebellar pilocytic astrocytoma present-
rhabdoid tumors of the central nervous system: clinical, radiographic and ing with intratumor bleeding, subarachnoid hemorrhage, and subdural
pathologic features. Pediatr Neurosurg 2002;37:64–70. hematoma. Childs Nerv Syst 2009;25:125–128.
120. Parmar H, Hawkins C, Bouffet E, Rutka J, Shroff M. Imaging findings in 146. Sato K, Rorke LB. Vascular bundles and wickerworks in childhood brain
primary intracranial atypical teratoid/rhabdoid tumors. Pediatr Radiol tumors. Pediatr Neurosci 1989;15:105–110.
2006;36:126. 147. Linscott LL, Osborn AG, Blaser S, et al. Pilomyxoid astrocytoma: expand-
121. Meyers SP, Khademian ZP, Biegel JA, Chuang SH, Korones DN, Zim- ing the imaging spectrum. Am J Neuroradiol. 2008;29:1872–1877.
merman RA. Primary intracranial atypical teratoid/rhabdoid tumors of 148. Lee YY, van Tassel P, Bruner JM, Moser RP, Share JC. Juvenile pilocytic
infancy and childhood: MRI features and patient outcomes. Am J Neuro- astrocytomas: CT and MR characteristics. Am J Roentgenol 1989;152:1263–
radiol 2006;27:962–971. 1270.
122. Reinhardt D, Behnke-Mursch J, Weiss E, et al. Rhabdoid tumors of the 149. Zimmerman RA, Bilaniuk LT, Bruno L, et al. Computed tomography of
central nervous system. Childs Nerv Syst 2000;16:228–234. cerebellar astrocytoma. Am J Roentgenol 1978;130:929–933.
123. Bing F, Nugues F, Grand S, Bessou P, Salon C. Primary intracranial 150. Harris LM, Davies NP, MacPherson L, et al. Magnetic resonance spec-
extra-axial and supratentorial atypical rhabdoid tumor. Pediatr Neurol troscopy in the assessment of pilocytic astrocytomas. Europ J Cancer
2009;41:453–456. 2008;44:2640–2647.
124. Biswas A, Goyal S, Puri T, et al. Atypical teratoid rhabdoid tumor of 151. Allen JC,Siffert J,Hukin J.Clinical manifestations of childhood ependymoma:
the brain: case series and review of literature. Childs Nerv Syst 2009;25: a multitude of syndromes. Pediatr Neurosurg 1998;28:49–55.
1495–1500. 152. Centano RS, Lee AA, Winter J, Barba D. Supratentorial ependymo-
125. Warmuth-Metz M, Bison B, Dannemann-Stern E, Kortmann RD, Rut- mas: neuroimaging and clinicopathological correlation. J Neurosurg
kowski S, Pietsch T. CT and MR imaging in atypical teratoid/rhabdoid 1986;64:209–215.
tumors of the central nervous system. Neuroradiology 2008;50:447–452. 153. Pierre-Kahn A, Hirsch JF, Roux FX, et al. Intra-cranial ependymomas
126. Fenton L, Foreman N. Atypical teratoid/rhabdoid tumor of the central in childhood: survival and functional results of 47 cases. Childs Brain
nervous system in children: an atypical series and review. Pediatr Radiol 1983;10:145–156.
2003;33:554–558. 154. Swartz JD, Zimmerman RA, Bilaniuk LT. Computed tomography of
127. Tekautz TM, Fuller CE, Blaney S, et al. Atypical teratoid/rhabdoid tumors intracranial ependymomas. Radiology 1982;143:97–101.
(ATRT): Improved survival in children 3 years of age and older with radia- 155. Lehman NL. Patterns of brain infiltration and secondary structure for-
tion therapy and high-dose alkylator-based chemotherapy. J Clin Oncol mation in supratentorial ependymal tumors. J Neuropathol Exp Neurol
2005;23:1491–1499. 2008;67:900–910.
128. De Mot P, Demaerel P, Wilms G, Van Gool S, Sciot R. Imaging features of 156. Lantos P, Louis D, Rosenblum M, Kleihus P. Tumours of the nervous
multifocal primary rhabdoid tumour of the central nervous system with system – ependymoblastomas. In: Love S, Louis D, Ellison D, eds. Green-
meningeal and spinal metastases. Pediatr Radiol 2003;33:275–277. field’s Neuropathology. London-New York-Delhi: Arnold, 2002:824–832;
129. Hanna SL, Langston JW, Parham DM, Douglass EC. Primary malignant 875–877.
rhabdoid tumor of the brain: Clinical, imaging, and pathologic findings. 157. Kurt E, Zheng PP, Hop WCJ, et al. Identification of relevant prognostic
Am J Neuroradiol 1993;14:107–115. histopathologic features in 69 intracranial ependymomas, excluding
130. Arslanoglu A, Aygun N, Tekhtani D, et al. Imaging findings of CNS atypical myxopapillary ependymomas and subependymomas. Cancer 2006;106:
teratoid/rhabdoid tumors. Am J Neuroradiol 2004;25:476–480. 388–395.
131. Yoon C, Chuang S, Jay V. Primary malignant rhabdoid tumor of the brain: 158. U-King-Im J, Taylor M, Raybaud C. Posterior fossa ependymomas: new
CT and MR findings. Yonsei Med J 2000;41:8–16. radiological classification with surgical correlation. Childs Nerv Syst
132. Lee YK, Choi CG, Lee JH. Atypical teratoid/rhabdoid tumor of the 2010:1–8.
cerebellum: report of two infantile cases. Am J Neuroradiol 2004;25: 159. Ikezaki K, Matsushima T, Inoue T, Yokoyama N, Kanedo Y, Fukui M. Cor-
481–483. relation of microanatomical localization with postoperative survival in
133. Kuroiwa T, Numaguchi Y, Rothman M, et al. Posterior fossa glioblastoma posterior fossa ependymomas. Neurosurgery 1993;32:38–44.
multiforme: MR findings. Am J Neuroradiol 1995;16:583–589. 160. Nagib MG, O’Fallon MT. Posterior fossa lateral ependymoma in child-
134. Bristot R, Raco A, Vangelista T, Delfini R. Malignant cerebellar astrocy- hood. Pediatr Neurosurg 1996;24:299–305.
tomas in childhood. Child’s Nerv Syst 1998;14:532–536. 161. Rezai AR, Woo HH, Lee M, Cohen H, Zagzag D, Epstein FJ. Dissemi-
135. Viano J, Herrera E, Suarez J. Cerebellar astrocytomas: a 24-year experi- nated ependymomas of the central nervous system. J Neurosurg 1996;85:
ence. Childs Nerv Syst 2001;17:607–610. 618–624.

Barkovich_Chap07.indd 795 5/6/2011 9:18:13 PM


796 Pediatric Neuroimaging

162. Salazar M. A better understanding of CNS seeding and a brighter outlook 185. Epstein F, Wisoff J. Intra-axial tumors of the cervico-medullary junction.
for postoperatively irradiated patients with ependymoma. Int J Radiat J Neurosurg 1987;67:483–487.
Oncol Biol Phys 1983;9:1231–1235. 186. Khatib Z, Heideman R, Kovnar E, et al. Predominance of pilocytic histology
163. Wang Z, Sutton LN, Cnaan A, et al. Proton MR spectroscopy of pediatric in dorsally exophytic brain stem tumors. Pediatr Neurosurg 1994;20:2–10.
cerebellar tumors. Am J Neuroradiol 1995;16:1821–1833. 187. Mandell LR, Kadota R, Freeman C, et al. There is no role for hyperfrac-
164. Hwang J-H, Egnaczyk GF, Ballard E, Dunn RS, Holland SK, Ball WS, Jr. tionated radiotherapy in the management of children with newly diag-
Proton MR spectroscopic characteristics of pediatric pilocytic astrocy- nosed diffuse intrinsic brainstem tumors: results of a pediatric oncology
tomas. Am J Neuroradiol 1998;19:535–540. group phase III trial comparing conventional vs. hyperfractionated radio-
165. Krouwer HGJ, Kim GA, Radn SD, et al. Single voxel proton MR spectros- therapy. Int J Radiat Oncol Biol Phys 1999;43:959–964.
copy of nonneoplastic brain lesions suggestive of a neoplasm. Am J Neuro- 188. Hoffman HJ. Dorsally exophytic brain stem tumors and midbrain tumors.
radiol 1998;19:1695–1703. Pediatr Neurosurg 1996;24:256–262.
166. Packer RJ, Nicholson HS, Vezina LG, Johnson DL. Brainstem Gliomas. In: 189. Mantravardi RVP, Phatak R, Bellur S, et al. Brainstem glioma: an autopsy
Berger MS, ed. Pediatric Neuro-oncology, vol 3. Philadelphia: Saunders, study of 25 cases. Cancer 1982;49:1294–1296.
1992:863–879. 190. Smith RR, Zimmerman RA, Packer RJ, et al. Pediatric brainstem glioma;
167. Walker D, Punt J, Sokal M. Clinical management of brain stem glioma. post-radiation clinical and radiographic follow-up. Neuroradiology
Arch Dis Child 1999;80:558–564. 1990;32:265–271.
168. Grigsby PW, Thomas PR, Schwartz HG, Fineberg BB. Multivariant analy- 191. May PL, Blaser SI, Hoffman HJ, Humphreys RP, Harwood-Nash DC.
sis of prognostic factors in pediatric and adult thalamic and brainstem Benign intrinsic tectal “tumors” in children. J Neurosurg 1991;74:
tumors. Int J Radiat Oncol 1989;16:649–655. 867–871.
169. Queiroz LS, de Cruz Neto J, Lopes de Faria J. Glioblastoma multiforme of 192. Robertson PL, Muraszko KM, Brunberg JA, Axtell RA, Dauser RC, Turrisi
the medulla oblangata. Acta Neuropathol (Berl) 1974;29:355–360. AT. Pediatric midbrain tumors: a benign subgroup of brainstem gliomas.
170. Yen P, Chou A, Chen C, Jung S, Chuang H, Scott R. Primary medulla oblon- Pediatr Neurosurg 1995;22:65–73.
gata germinoma: a case report and review of the literature. J Neurooncol 193. Daglioglu E, Cataltepe O, Akalan N. Tectal gliomas in children: the impli-
2003;62:339–342. cations for natural history and management strategy. Pediatr Neurosurg
171. Kurtkaya-Yapicier O, Elmaci I, Boran B, Kilic T, Sav A, Pamir M. Dysem- 2003;38:223–231.
bryoplastic neuroepithelial tumor of the midbrain tectum: a case report. 194. Hamilton MG, Lauryssen C, Hagen N. Focal midbrain glioma: long term
Brain Tumor Pathol 2002;19:97–100. survival in a cohort of 16 patients and the implications for management.
172. Lagares A, Gomez P, Lobato R, Ricoy J, Ramos A, de la Lama A. Ganglio- Can J Neurol Sci 1996;23:204–207.
glioma of the brainstem: report of three cases and review of the literature. 195. Pollack IF, Pang D, Albright AL. The long term outcome in children
Surg Neurol 2001;56:315–322. with late onset aquedectal stenosis resulting from benign intrinsic tectal
173. Zagzag D, Miller D, Knopp E, et al. Primitive neuroectodermal tumors tumors. J Neurosurg 1994;80:681–688.
of the brainstem: investigation of seven cases. Pediatrics 2000;106: 196. Tomita T, Cortes R. Astrocytomas of the cerebral penuncle in children:
1045–1053. surgical experience in seven patients. Childs Nerv Syst 2002;18:225–230.
174. Weaver K, Armao D, Wiley J, Ewend M. Histiocytic lesion mimicking 197. Bowers DC, Georgiades C, Aronson LJ, et al. Tectal gliomas: natural
intrinsic brainstem neoplasm. Case report. J Neurosurg 1999;91: history of an indolent lesion in pediatric patients. Pediatr Neurosurg
1037–1040. 2000;32:24–29.
175. Grois N, Barkovich AJ, Rosenau W, Ablin AR. Central nervous system 198. Pollack IF, Shultz B, Mulvihill JJ. The management of brainstem gliomas
disease associated with Langerhans’ cell histiocytosis. Am J Ped Hematol/ in patients with neurofibromatosis 1. Neurology 1996;46:1652–1660.
Oncol 1993;15:245–254. 199. Bilaniuk LT, Molloy PT, Zimmerman RA, et al. Neurofibromatosis type 1:
176. Fisher P, Breiter S, Carson B, et al. A clinicopathologic reappraisal of brain brain stem tumours. Neuroradiology 1997;39:642–653.
stem tumor classification. Identification of pilocystic astrocytoma and 200. Milstein JM, Geyer JR, Berger MS, Bleyer WA. Favorable prognosis for
fibrillary astrocytoma as distinct entities. Cancer 2000;89:1569–1576. brainstem gliomas in neurofibromatosis. J Neurooncol 1989;7:367–371.
177. Jallo G, Biser-Rohrbaugh A, Freed D. Brainstem gliomas. Childs Nerv Syst 201. Molloy PT, Bilaniuk LT, Vaughan SN, et al. Brainstem tumors in patients
2003;20:143–153. with neurofibromatosis type 1: a distinct clinical entity. Neurology
178. Albright AL, Packer RJ, Zimmerman RA, Rorke LB, Boytt J, Hammond 1995;45:1897–1902.
GD. Magnetic resonance scans should replace biopsies for the diagnosis 202. Raffel C, McComb JG, Bodner S, Gilles FE. Benign brainstem lesions in
of diffuse brain stem gliomas: a report from the Children’s Cancer Group. pediatric patients with neurofibromatosis: case reports. Neurosurgery
Neurosurgery 1993;33:1026–1030. 1989;25:959–964.
179. Fischbein NJ, Prados MD, Wara W, Russo C, Edwards MSB, Barkovich AJ. 203. Broniscer A, Gajjar A, Bhargava R, et al. Brain stem involvement in chil-
Radiologic classification of brain stem tumors: correlation of magnetic dren with neurofibromatosis type 1: role of magnetic resonance imaging
resonance imaging appearance with clinical outcome. Pediatr Neurosurg and spectroscopy in the distinction from diffuse pontine glioma. Neuro-
1996;24:9–23. surgery 1997;40:331–338.
180. Barkovich AJ, Krischer J, Kun LE, et al. Brain stem gliomas: a classification 204. Baum P, Barkovich AJ, Koch T, Berg BO. Deep gray matter involvement
system based on magnetic resonance imaging. Pediatr Neurosci 1990–91;16: in children with acute disseminated encephalomyelitis. Am J Neuroradiol
73–83. 1994;15:1275–1283.
181. Kornreich L, Schwartz M, Karmazyn B, et al. Role of MRI in the manage- 205. Lowichik A, Rollins N, Delgado R, Visvesvara G, Burns D. Leptomyxid
ment of children with diffuse pontine tumors: a study of 15 patients and amebic meningoencephalitis mimicking brain stem glioma. Am J Neuro-
review of the literature. Pediatr Radiol 2005;35:872–879. radiol 1995;16:926–929.
182. Pollack IF, Hoffman HJ, Humphreys RP, Becker LE. The long term out- 206. Zimmerman RA. Neuroimaging of pediatric brain stem diseases other
come after surgical treatment of dorsally exophytic brain stem gliomas. than brain stem glioma. Pediatr Neurosurg 1996;25:83–92.
J Neurosurg 1993;78:859–863. 207. al-Deeb SM, Yaqub BA, Sharif HS, Motaery KR. Neurotuberculosis: a
183. Stroink AR, Hoffman HJ, Hendrick EB, Humphreys RP, Davidson G. review. Clin Neurol Neurosurg 1992;94:S30–S33.
Transependymal benign dorsally exophytic brain stem gliomas in child- 208. Gupta R, Vatsal D, Husain H, et al. Differentiation of tuberculous from
hood: diagnosis and treatment recommendations. Neurosurgery 1987;20: pyogenic brain abscesses with in vivo proton MR spectroscopy and mag-
439–444. netization transfer MR imaging. Am J Neuroradiol 2001;22:1503–1509.
184. Edwards MSB, Wara WM, Barkovich AJ. Focal brain-stem astrocytomas 209. Akutsu H, Matsumura A, Isobe T, et al. Chronological change of brain
causing symptoms of involvement of the facial nucleus: long term survival abscess in (1)H magnetic resonance spectroscopy. Neuroradiology
in six pediatric cases. J Neurosurg 1994;80:20–25. 2002;44:574–578.

Barkovich_Chap07.indd 796 5/6/2011 9:18:13 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 797

210. Wanebo J, Lonser R, Glenn G, Oldfield E. The natural history of heman- 237. Fujimaki T, Matsutani M, Funada N, et al. CT and MRI features of intrac-
gioblastomas of the central nervous system in patients with von Hippel- ranial germ cell tumors. J Neurooncol 1994;19:217–226.
Landau disease. J Neurosurg 2003;98:82–94. 238. Petzold A, Thom M, Powell M, Plant GT. Relapsing intracranial Rosai-
211. Lee DR, Sanches J, Mark AS, Dillon WP, Norman D, Newton TH. Posterior Dorfman disease. J Neurol Neurosurg Psychiatry 2001;71:538–541.
fossa hemangioblastomas: MR imaging. Radiology 1989;171:463–468. 239. Wright RA, Hermann RC, Parisi JE. Neurological manifestations of
212. Sato Y, Waziri M, Smith W, et al. Hippel-Lindau disease: MR imaging. Erdheim-Chester disease. J Neurol Neurosurg Psychiatry 1999;66:72–75.
Radiology 1988;166:241–246. 240. Zak IT, Altinok D, Neilsen SSF, Kish KK. Xanthoma dissminatum of the
213. Ho VB, Smirniotopoulos JG, Murphy FM, Rushing EJ. Radiologic- central nervous system and cranium. Am J Neuroradiol 2006;27:919–921.
pathologic correlation: hemangioblastoma. Am J Neuroradiol 1992; 241. Demaerel P, Van Gool S. Paediatric neuroradiological aspects of Langer-
13:1343–1352. hans cell histiocytosis. Neuroradiology 2008;50:85–92.
214. Allcutt DA, Hoffman HJ, Isla A, et al. Acoustic schwannomas in children. 242. Kilborn TN, The J, Goodman TR. Paediatric manifestations of Langerhans
Neurosurgery 1991;29:14–19. cell histiocytosis: a review of the clinical and radiological findings. Clin
215. Pinto RS, Kricheff II. Neuroradiology of intracranial neuromas. Semin Radiol 2003;58:269–278.
Roentgenol 1984;14:44–52. 243. Hayward J, Packer R, Finley J. Central nervous system and Langerhans’ cell
216. Latack JT, Gabrielson TD, Knake JE, et al. Facial nerve neuromas: radio- histiocytosis. Med Pediatr Oncol 1990;18:324–328.
logic evaluation. Radiology 1983;149:731–739. 244. Rosenfield NS, Abrahams J, Komp D. Brain MR in patients with Langer-
217. Kingsley DPE, Brooks GB, Leung AWL, Johnson MA. Acoustic neuromas: hans’ cell histiocytosis: findings and enhancement with Gd-DTPA. Pediatr
evaluation by MR imaging. Am J Neuroradiol 1985;6:1–5. Radiol 1990;20:433–436.
218. Gentry LR, Jacoby CG, Turski PA, et al. Cerebellopontine angle - petromastoid 245. Ragland RL, Moss DS, Duffis AW, et al. CT and MR findings in diffuse
mass lesions: comparative study of diagnosis with MR imaging and CT. cerebral histiocytosis. Am J Neuroradiol 1991;12:525–526.
Radiology 1987;162:513–520. 246. Prayer D, Grois N, Prosch H, Gadner H, Barkovich A. MR imaging presen-
219. Curati WL, Graif M, Kingsley DPE, et al. Acoustic neuromas: Gd-DTPA tation of intracranial disease associated with Langerhans cell histiocytosis.
enhancement in MR imaging. Radiology 1986;158:447–451. Am J Neuroradiol 2004;25:880–891.
220. Shah MV, Haines SJ. Pediatric skull, skull base, and meningeal tumors. 247. Thorphy MJ, Crosley CJ. Multifocal eosinophilic granuloma and the cer-
In: Berger MS, ed. Pediatric Neuro-oncology, vol 4. Philadelphia: Saunders, ebellum. Ann Neurol 1980;8:454–457.
1992:893–924. 248. Barthez M, Araujo E, Donadieu J. Langerhans cell histiocytosis and the
221. Maravilla KR. Intraventricular fat-fluid level secondary to rupture of an central nervous system in childhood: evolution and prognostic factors.
intracranial dermoid cyst. Am J Roentgenol 1977;128:500–501. Results of a collaborative study. J Child Neurol 2000;15:150–156.
222. Martinez-Lage J, Ramos J, Puche A, Poza M. Extradural dermoid tumours 249. Poe L, Dubowy R, Hochhauser L, et al. Demyelinating and gliotic cer-
of the posterior fossa. Arch Dis Child 1997;77:427–430. ebellar lesions in Langerhans cell histiocytosis. AJNR: Am J Neuroradiol
223. Zimmerman RA, Bilaniuk LT, Dolinskas C. Cranial CT of epidermoid 1994;15:1921–1928.
and congenital fatty tumors of maldevelopment origin. J Comput Tomogr 250. Grois N, Prayer D, Prosch H, Lassmann H. Neuropathology of CNS dis-
1979;3:40–47. ease in Langerhans cell histiocytosis. Brain Pathol 2005;128:829–838.
224. Tsuruda JS, Chew WM, Moseley ME, Norman D. Diffusion-weighted MR 251. Prosch H, Grois N, Wnorowski M, Steiner M, Prayer D. Long-term MR
imaging of the brain: value of differentiating between extraaxial cysts and imaging course of neurodegenerative Langerhans cell histiocytosis. Am J
epidermoid tumors. Am J Neuroradiol 1990;11:925–931. Neuroradiol 2007;28:1022–1028.
225. Chen S, Ikawa F, Kurisu K, Arita K, Takaba J, Kanou Y. Quantitative MR 252. Steiner M, Prayer D, Asenbaum S, et al. Modern imaging methods for the
evaluation of intracranial epidermoid tumors by fast fluid-attenuated assessmento of Langerhans’ cell histiocytosis-associated neurodegenera-
inversion recovery imaging and echo-planar diffusion-weighted imaging. tive syndrome: case report. J Child Neurol 2005;20:253–257.
Am J Neuroradiol 2001;22:1089–1096. 253. Wold LE, Laws ER. Cranial chordomas in children and young adults.
226. Ikushima I, Korogi Y, Hirai T, et al. MR of epidermoids with a variety of J Neurosurg 1983;59:1043–1047.
pulse sequences. Am J Neuroradiol 1997;18:1359–1363. 254. Matsumoto J, Towbin RB, Ball WS, Jr. Cranial chordomas in infancy and
227. Chew WM, Rowley HA, Barkovich AJ. Magnetization transfer contrast childhood. A report of two cases and review of the literature. Pediatr
imaging in pediatric patients. Radiology 1992;185(P):281. Radiol 1989;20:28–32.
228. Horowitz BL, Chari MV, James R, Bryan RN. MR of intracranial epider- 255. Handa J, Suzuki F, Nioka H, Koyama T. Clivus chordoma in childhood.
moid tumors: correlation of in vivo imaging with in vitro C13 spectros- Surg Neurol 1987;28:58–62.
copy. Am J Neuroradiol 1990;11:299–302. 256. Meyer JE, Oot RF, Lindfors KK. CT appearance of clival chordomas.
229. Barkovich AJ, Vandermarck P, Edwards MSB, Cogen PH. Congenital J Comput Assist Tomogr 1986;10:34–38.
nasal masses: CT and MR imaging features in 16 cases. Am J Neuroradiol 257. Sze G, Uichanco LS, Brant-Zawadzki MN, et al. Chordomas: MR imaging.
1991;12:105–116. Radiology 1988;166:187–191.
230. Henkelman MH, Watts JF, Kucharczyk W. High signal intensity in MR 258. Huvos AG, Marcove RC. Chondrosarcoma in the young: a clinicopatho-
images of calcified brain tissue. Radiology 1991;179:199–206. logic analysis of 79 patients younger than 21 years of age. Am J Surg Pathol
231. Bejjani gK, Wright CD, Schessel D, Sekhar LN. Endodermal cysts of the 1987;11:930–942.
posterior fossa. J Neurosurg 1998;89:326–335. 259. Dahlin CC, Unni KK. Bone Tumors, 4th ed. Springield, IL: Charles C.
232. Harris CP, Dias MS, Brockmeyer DL, Townsend JJ, Willis BK, Apfelbaum Thomas, 1986.
RI. Neurenteric cysts of the posterior fossa: recognition, management, and 260. Voorhies SP, Sundaresan N. Tumors of the skull. In: Wilkins RH, Ren-
embryogenesis. Neurosurgery 1991;29:893–897. gachary SS, eds. Neurosurgery. New York: McGraw-Hill, 1984:984–1009.
233. Preece MT, Osborn AG, Chin SS, Smirniotopoulos J. Intracranial neur- 261. Meyers SP, Hirsch WL, Jr, Curtin HD, Barnes L, Sekhar LN, Sen
enteric cysts: imaging and pathology spectrum. Am J Neuroradiol C. Chondrosarcomas of the skull base: MR imaging features. Radiology
2006;27:1211–1216. 1992;184:103–108.
234. Kim C, Wang K, Choe G, et al. Neurenteric cyst: its various presentations. 262. Shaw PJ, Eden T. Neuroblastoma with intracranial involvement: an ENSG
Childs Nerv Syst 1999;15:333–341. study. Med Pediatr Oncol 1992;20:149–155.
235. Shin J, Byun B, Kim D, Choi D. Neurenteric cyst in the cerebellopontine 263. Applegate GR, Hirsch WL, Applegate LJ, Curtin HD. Variability in the
angle with xanthogranulomatous changes: serial MR findings with patho- enhancement of the normal central skull base in children. Neuroradiology
logic correlation. Am J Neuroradiol 2002;23:663–665. 1992;34:217–221.
236. Brooks BS, Duvall ER, El Gammal T. Neuroimaging features of neuren- 264. Hadfield MG, Luo VY, Williams RL, Ward JD, Russo CP. Ewing sar-
teric cysts: analysis of nine cases and review of the literature. Am J Neuro- coma of the skull in an infant: case report and review. Pediatr Neurosurg
radiol 1993;14:735–746. 1996;25:100–104.

Barkovich_Chap07.indd 797 5/6/2011 9:18:13 PM


798 Pediatric Neuroimaging

265. Chateil JF, Dousset V, Meyer P, et al. Cranial aneurysmal bone cysts pre- 291. Goh S, Butler W, Thiele EA. Subependymal giant cell tumors in tuberous
senting with raised intracranial pressure: report of two cases. Neuroradiol- sclerosis complex. Neurology 2004;63:1457–1461.
ogy 1997;39:490–494. 292. Braffman BH, Bilaniuk LT, Naidich TP, et al. MR imaging of tuberous
266. Arthur RJ, Brunelle F. Computerized tomography in the evaluation of sclerosis: insights into the pathogenesis of this phakomatosis, comments
expansile lesions arising from the skull vault in childhood. A report of 5 on the role of gadopentate dimeglumine, and literature review. Radiology
cases. Pediatr Radiol 1988;18:294–301. 1992;183:227–238.
267. Bilge T, Coban O, Ozden B, Turantan I, Turker K, Bahar S. Aneurysmal 293. Franz DN, Leonard J, Tudor C, et al. Rapamycin causes regression of astro-
bone cysts of the occipital bone. Surg Neurol 1988;20:227–230. cytomas in tuberous sclerosis complex. Ann Neurol 2006;59:490–498.
268. Beltran J, Simon D, Levy M, Herman L, Weis L, Mueller C. Aneurysmal 294. Gardeur D, Palmieri A, Mashaly R. Cranial computed tomography in the
bone cysts: MR imaging at 1.5T. Radiology 1986;158:689–690. phakomatoses. Neuroradiology 1983;25:293–304.
269. Blandino A, Longo M, Luciani A, et al. RM in un caso di tumore a cellule 295. Martin N, de Brouker T, Cambier J, Marsault C, Nahum H. MRI evalua-
giganti del tavolato cranico. Rivista di Neuroradiologia 1996;9:339–343. tion of tuberous sclerosis. Neuroradiology 1987;29:437–443.
270. Desai KI, Nadkarni TD, Goel A, Muzumdar DP, Naresh KN, Nair CN. 296. Martin N, Debussche C, De Broucker T, Mompoint D, Marsault C, Nahum
Primary Ewing’s sarcoma of the cranium. Neurosurgery 2000;46:62–68; H. Gadolinium-DTPA enhanced MR imaging in tuberous sclerosis. Neu-
discussion 68–69. roradiology 1990;31:492–497.
271. Li W-U, Brock P, Saunders DE. Imaging characteristics of primary cranial 297. Altman NR, Purser RK, Post MJD. Tuberous sclerosis: characteristics at
Ewing sarcoma. Pediatr Radiol 2005;35:612–618. CT and MR imaging. Radiology 1988;167:527–532.
272. Palma L, Guidetti B. Cystic pilocytic astrocytomas of the cerebral hemi- 298. Kepes J, Rubinstein L, Eng L. Pleomorphic xanthoastrocytoma: a distinc-
spheres: surgical experience with 51 cases and long term results. J Neuro- tive meningocerebral glioma of young subjects with relatively favorable
surg 1985;62:811–815. prognosis. A study of 12 cases. Cancer 1979;44:1839–1852.
273. Mercuri S, Russo A, Palma L. Hemispheric supratentorial astrocytomas in 299. Kepes J, Louis D, Giannini C, Paulus W. Pleomorphic Xanthoastrocytoma.
children. Long-term results in 29 cases. J Neurosurg 1981;55:170–173. In: Kleihues P, Cavenee W, eds. Pathology and Genetics of Tumours of the
274. Berger MS, Keles GE, Geyer JR. Cerebral hemispheric tumors of child- Nervous System. Lyon, France: IARC, 2000:52–54.
hood. In: Berger MS, ed. Pediatric Neuro-oncology, vol 3. Philadelphia: 300. Giannini C, Scheithauer B, Burger P, et al. Pleomorphic xanthoastrocy-
Saunders, 1992:839–852. toma: what do we really know about it? Cancer 1999;85:2033–2045.
275. Martinez-Lage J, Perez-Espejo M, Esteban J, Poza M. Thalamic tumors: 301. Koeller K, Henry J. From the archives of the AFIP: superficial gliomas:
clinical presentation. Childs Nerv Syst 2002;18:405–411. radiologic-pathologic correlation. Armed Forces Institute of Pathology.
276. Shady J, Black PMcL, Kupsky W, et al. Seizures in children with supraten- Radiographics 2001;21:1533–1556.
torial astroglial neoplasms. Pediatr Neurosurg 1994;21:23–30. 302. Lipper M, Eberhard D, Phillips C, Verzina L, Cail W. Pleomorphic xan-
277. Artico M, Cervoni L, Celli P, Salvati M, Palma L. Supratentorial glioblas- thoastrocytoma, a distinctive astroglial tumor: neuroradiologic and
toma in children: a series of 27 surgically treated cases. Childs Nerv Syst pathologic features. Am J Neuroradiol 1993;14:1397–1404.
1993;9:7–9. 303. Pahapill P, Ramsay D, Del Maestro R. Pleomorphic xanthoastrocytoma:
278. Dropcho EJ, Wisoff JH, Walker RW, Allen JC. Supratentorial malig- case report and analysis of the literature concerning the efficacy of resec-
nant gliomas in childhood: a review of fifty cases. Ann Neurol 1987;22: tion and the significance of necrosis. Neurosurgery 1996;38:822–828.
355–364. 304. Johnson J, Jr, Hariharan S, Berman J, et al. Clinical outcome of pediat-
279. Tomita T, McLone DG, Naidich TP. Mural tumors with cysts in the cere- ric gangliogliomas: ninety-nine cases over 20 years. Pediatr Neurosurg
bral hemispheres of children. Neurosurgery 1986;19:998–1005. 1997;27:203–207.
280. Mamelak AN, Prados MD, Obana WG, Cogen PH, Edwards MSB. Treat- 305. Nelson M. Neuronal and mixed neuronal-glial tumors. In: Kleihues P,
ment options and prognosis of multicentric juvenile pilocytic astrocy- Cavenee W, eds. Pathology and Genetics of Tumors of the Nervous System.
toma. J Neurosurg 1994;81:24–30. Lyon, France: IARC, 2000.
281. Gajjar A, Bhargava R, Jenkins JJ, et al. Low-grade astrocytoma with 306. Mickle JP. Ganglioglioma in children: a review of 32 cases. Pediatr Neuro-
neuraxis dissemination at diagnosis. J Neurosurg 1995;83:67–71. surg 1992;18:310–314.
282. Joyce P, Bentson J, Takahashi M, et al. The accuracy of predicting his- 307. Chintagumpala MM, Armstrong D, Miki S, et al. Mixed neuronal-glial
tologic grades of supratentorial astrocytomas on the basis of comput- tumors (gangliogliomas) in children. Pediatr Neurosurg 1996;24:306–313.
erized tomography and cerebral angiography. Neuroradiology 1978;16: 308. Sutton LN, Packer RJ, Rorke LB, et al. Cerebral gangliogliomas during
346–348. childhood. Neurosurgery 1983;13:124–128.
283. Drayer BP, Johnson BP, Bird CR, Flom RA, Hodak JA. Magnetic resonance 309. Johansson JH, Rekate HL, Roessmann U. Gangliomas: pathological and
imaging and glioma. BNI Quarterly 1987;3:44–55. clinical correlation. J Neurosurg 1981;54:58–63.
284. Tien RD, Felsberg GJ, Friedman H, Brown M, MacFall J. MR imaging of 310. Garrido E, Becker LF, Hoffman HJ, et al. Gangliomas in children: a clini-
high-grade cerebral gliomas: value of diffusion-weighted echoplanar pulse copathological study. Childs Brain 1978;4:339–346.
sequences. Am J Roentgenol 1994;162:671–677. 311. Tampieri D, Moumdjian R, Melanson D, Ethier R. Intracerebral ganglio-
285. Bagley LJ, Grossman RI, Judy KD, et al. Gliomas: correlation of magnetic gliomas in patients with partial complex seizures: CT and MR imaging
susceptibility artifact with histologic grade. Radiology 1997;202:511–516. findings. Am J Neuroradiol 1991;12:749–755.
286. Gauvain K, McKinstry R, Mukherjee P, et al. Evaluating pediatric brain 312. Blumcke I, Wiestler O. Gangliogliomas: an intriguing tumor entirely asso-
tumor cellularity with diffusion-tensor imaging. Am J Neuroradiol ciated with focal epilepsies. J Neuropathol Exp Neurol 2002;61:575–584.
2001;177:449–454. 313. Otsubo H, Hoffman HJ, Humphreys RP, et al. Evaluation, surgical
287. Chang Y-W, Yoon H-K, Shin H-J, Roh HG, Cho JM. MR imaging of glio- approach and outcome of seizure patients with gangliogliomas. Pediatr
blastoma in children: usefulness of diffusion/perfusion-weighted MRI Neurosurg 1990–91;16:208–212.
and MR spectroscopy. Pediatr Radiol 2003;33:836–842. 314. Smith NM, Carli MM, Hanieh A, Clark B, Bourne AJ, Byard RW. Ganglio-
288. Law M, Young RJ, Babb JS, et al. Gliomas: Predicting time to progres- gliomas in childhood. Childs Nerv Syst 1992;8:258–262.
sion or survival with cerebral blood volume measurements at dynamic 315. Demierre B, Stichnoth FA, Hori A, Spoerri O. Intracerebral ganglioma.
susceptibility-weighted contrast-enhanced perfusion MR imaging. Radi- J Neurosurg 1986;65:177–182.
ology 2008;247:490–498. 316. Liu GT, Galetta SL, Rorke LB, et al. Gangliogliomas involving the optic
289. Reagan TJ. Neuropathology. In: Gomez MR, ed. Tuberous Sclerosis, 2nd ed. chiasm. Neurology 1996;46:1669–1673.
New York: Raven, 1988:63–74. 317. Rushing E, Rorke L, Sutton L. Problems in the nosology of desmoplastic
290. Shepherd CW, Scheitharu B, Gomez MR, et al. Brain tumours in tuberous tumors of childhood. Pediatr Neurosurg 1993;19:57–62.
sclerosis. A clinicopathological study of the Mayo Clinic experience. Ann 318. VandenBerg SR. Desmoplastic infantile ganglioglioma and desmoplastic
NY Acad Sci 1991;615:378–379. cerebral astrocytoma of infancy. Brain Pathol 1993;3:275–281.

Barkovich_Chap07.indd 798 5/6/2011 9:18:13 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 799

319. VandenBerg SR, May EE, Rubinstein LB, et al. Desmoplastic supratentorial 344. Palma L, Celli P, Mariottini A, Zalaffi A, Schettini G. The importance of
neuroepithelial tumors of infancy with divergent differentiation potential surgery in supratentorial ependymomas. Long-term survival in a series of
(“desmoplastic infantile gagnliogliomas”). J Neurosurg 1987;66:58–71. 23 cases. Childs Nerv Syst 2000;16:170–175.
320. Taratuto A, Monges J, Lylyk P, Leiguarda R. Superficial cerebral astro- 345. Armington WG, Osborn AG, Cubberly DA, et al. Supratentorial ependymo-
cytoma attached to dura. Report of six cases in infants. Cancer 1984;54: mas: CT appearance. Radiology 1985;157:367–372.
2505–2512. 346. Yuh E, Barkovich A, Gupta N. Imaging of ependymomas: MRI and CT.
321. Mallucci C, Lellouch-tubiana A, Salazar C, et al. The management of Childs Nerv Syst 2009;25:1203–1213.
desmoplastic neuroepithelial tumors of childhood. Childs Nerv Syst 347. Hart MD, Earle KM. Primitive neuroectodermal tumors of the brain in
2000;16:8–14. children. Cancer 1973;3232:890–897.
322. Alexiou GA, Stefanaki K, Sfakianos G, Prodromou N. Desmoplastic Infan- 348. Horton BC, Rubinstein LJ. Primary cerebral neuroblastoma: a clinico-
tile Ganglioglioma. Pediatr Neurosurg 2008;44:422–425. pathological study of 35 cases. Brain 1976;99:735–736.
323. Tamburrini G, Colosimo C, Jr, Giangaspero F, Riccardi R, Di Rocco C. 349. Ashwal S, Hinshaw DB, Jr, Bedros A. CNS primitive neuroectodermal
Desmoplastic infantile ganglioglioma. Childs Nerv Syst 2003;19:292–297. tumors of childhood. Med Pediatr Oncol 1984;12:180–188.
324. Paulus W, Schlote W, Perentes E, Jacobi G, Warmuth-Metz M, Roggendorf 350. Kingsley DPE, Harwood-Nash DCF. Radiological features of the neuroec-
W. Desmoplastic supratentorial neuroepithelial tumors of infancy. Histo- todermal tumors of childhood. Neuroradiology 1984;26:463–467.
pathology 1992;21:43–49. 351. Kosnik EJ, Bocsel CP, Bay J, et al. Primitive neuroectodermal tumors of the
325. Rypens F, Esteban M, Lellouch-Tubiana A, et al. Desmoplastic supratento- central nervous system in children. J Neurosurg 1978;48:741–746.
rial neuroepithelial tumours of childhood: imaging in 5 patients. Neuro- 352. Altman N, Fitz CR, Chuang SH, Harwood-Nash DC, Cotter C, Armstrong
radiology 1996;38 (Suppl 1):S165–S168. D. Radiologic characteristics of primitive neuroectodermal tumors in
326. Sperner J, Gottschalk J, Neumann K, Schorner W, Lanksch WR, Scheffner children. Am J Neuroradiol 1985;6:15–18.
D. Clinical, radiological and histological findings in desmoplastic infantile 353. Chambers EF, Turski PA, Sobel D. Radiologic characteristics of primary
ganglioglioma. Childs Nerv Syst 1994;10:458–462. cerebral neuroblastomas. Radiology 1981;139:101–104.
327. Ganesan K, Desai S, Udwadia-Hegde A. Non-infantile variant of desmo- 354. Erdem E, Zimmerman R, Haselgrove J, Bilaniuk L, Hunter J. Diffusion-
plastic ganglioglioma: a report of 2 cases. Pediatr Radiol 2006;36:541–545. weighted imaging and fluid attenuated inversion recovery imaging in
328. Tenreiro-Picon OR, Kamath SV, Knorr JR, Ragland RL, Smith TW, Lau the evaluation of primitive neuroectodermal tumors. Neuroradiology
KY. Desmoplastic infantile ganglioglioma: CT and MRI features. Pediatr 2001;43:927–933.
Radiol 1995;25:540–543. 355. Law M, Kazmi K, Wetzel S, et al. Dynamic susceptibilty contrast-enhanced
329. Duffner PK, Burger PC, Cohen ME, et al. Desmoplastic infantile ganglio- perfusion and conventional MR imaging findings for adult patients
gliomas: an approach to therapy. Neurosurgery 1994;34:583–589. with cerebral primitive neuroectodermal tumors. Am J Neuroradiol
330. Daumas-Duport C, Scheithauer BW, Chodkiewicz J-P, Laws ER, Jr, 2004;25:997–1005.
Vedrenne C. Dysembryoplastic neuroepithelial tumor: a surgically curable 356. Davis P, Wichman RD, Takei Y, Hoffman JC, Jr. Primary cerebral neuroblas-
tumor of young patients with intractable partial seizures. Neurosurgery toma: CT and MR findings in 12 cases. Am J Neuroradiol 1990;11:115–120.
1988;23:545–556. 357. Molloy PT, Yachnis AT, Rorke LB, et al. Central nervous stystem medulloep-
331. Sharma M, Jain D, Gupta A, et al. Dysembryoplastic neuroepithelial tumor: ithelioma: a series of eight cases including two arising in the pons. J Neuro-
a clinicopathological study of 32 cases. Neurosurg Rev 2009;32:161–170. surg 1996;84:430–436.
332. Campos A, Clusmann H, von Lehe M, et al. Simple and complex dysem- 358. Pang LM, Roebuck DJ, Ng HK, Chan YL. Sellar and suprasellar medulloep-
bryoplastic neuroepithelial tumors (DNT) variants: clinical profile, MRI, ithelioma. Pediatr Radiol 2001;31:594–596.
and histopathology. Neuroradiology 2009;51:433–443. 359. Briner J, Bannwart F, Kleihues P, et al. Malignant small cell tumor of the
333. Pasquier B, Peoc’H M, Fabre-Bocquentin B, et al. Surgical pathology of brain with intermidiate filaments: a case of a primary cerebral rhabdoid
drug-resistant partial epilepsy. A 10-year experience with a series of 327 tumor. Pediatr Pathol 1985;3:117–118.
consecutive resections. Epileptic Disord 2002;4:99–119. 360. Parwani AV, Stelow EB, Pambuccian SE, Burger PC, Ali SZ. Atypical tera-
334. Daumas-Duport C. Dysembryoplastic neuroepithelial tumors in epilepsy toid/rhabdoid tumor of the brain. Cancer Cytopathol 2005;105:65–70.
surgery. In: Guerrini R, ed. Dysplasias of Cerebral Cortex and Epilepsy. 361. Evans A, Ganatra R, Morris SJ. Imaging features of primary malignant
Philadelphia: Lippencott-Raven, 1996:71–80. rhabdoid tumour of the brain. Pediatr Radiol 2001;31:631–633.
335. Cervera-Pierot P, Varlet P, Chodkiewicz J-P, Daumas-Duport C. Dysem- 362. Bonnin JM, Rubinstein LJ. Astroblastomas: a pathological study of 23
bryoplastic neuroepithelial tumors located in the caudate nucleus area: tumors, with a postoperative follow-up in 13 patients. Neurosurgery 1989;
report of four cases. Neurosurgery 1997;40:1065–1070. 25:6–13.
336. Koeller KK, Dillon WP. Dysembryoplastic neuroepithelial tumors: MR 363. Brat D, Hirose Y, Cohen KJ, Feuerstein BG, Burger P. Astroblastoma: clini-
appearance. Am J Neuroradiol 1992;13:1319–1325. copathologic features and chrmomsomal abnormaltities defined by com-
337. Kuroiwa T, Bergey GK, Rothman MI, et al. Radiologic appearance of the parative genomic hybridization. Brain Pathol 2000;10:342–352.
dysembryoplastic neuroepithelial tumor. Radiology 1995;197:233–238. 364. Salvati M, D’Elia A, Brogna C, et al. Cerebral astroblastoma: analysis of
338. Raymond AA, Halpin SF, Alsanjari N, et al. Dysembryoplastic neuroepi- six cases and critical review of treatment options. J Neuro-Oncol 2009;93:
thelial tumor: features in 16 patients. Brain 1994;117:461–475. 369–378.
339. Fernandez C, Girard N, Paz Paredes A, Bouvier-Labit C, Lena G, 365. Unal E, Koksal Y, Vajtai I, Toy H, Kocaogullar Y, Paksoy Y. Astroblastoma in
Figarella-Branger D. The usefulness of MR imaging in the diagnosis of a child. Childs Nerv Syst 2008;24:165–168.
dysembroplastic neuroepithelial tumor in children: a study of 14 cases. Am 366. Port JD, Brat D, Burger P, Pomper MG. Astroblastoma: radiologic-
J Neuroradiol 2003;24:829–834. pathologic corrleation and distinction from ependymoma. Am J Neurora-
340. Bulakbasi N, Kocaoglu M, Sanal TH, Tayfun C. Dysembryoplastic neu- diol 2002;23:243–247.
roepithelial tumors: proton MR spectroscopy, diffusion, and perfusion 367. Razak N, Baumgartner J, Bruner J. Pediatric oligodendrogliomas. Pediatr
characteristics. Neuroradiology 2007;49:805–812. Neurosurg 1998;28:121–129.
341. Kaplan A, Lawson M, Spataro J, et al. Positron emission tomography 368. Kouwenhoven MCM, Kros JM, French PJ, et al. 1p/19q loss within oligo-
using [18F] fluorodeoxyglucose and [11c] 1-methionine to metabolically dendroglioma is predictive for response to first line temozolomide but not
characterize dysembryoplastic neuroepithelial tumors. J Child Neurol to salvage treatment. Europ J Cancer 2006;42:2499–2503.
1999;14:673–677. 369. Smith JS, Perry A, Borell TJ, et al. Alterations of chromosome arms 1p and
342. Roncali F, Consales A, Fioravanti A, Cenacchi G. Supratentorial cortical 19q as predictors of survival in oligodendrogliomas, astrocytomas, and
ependymoma: report of three cases. Neurosurgery 2005;57:E192. mixed oligoastrocytomas. J Clin Oncol 2000;18:636–645.
343. Sato Y, Ochiai H, Yamakawa Y, Nabeshima K, Asada Y, Hayashi T. Brain 370. Rizk T, Mottoloese C, Bouffet E, et al. Cerebral oligodendrogliomas in
surface ependymoma. Neuropathology 2000;20:315–318. children: an analysis of 15 cases. Childs Nerv Syst 1996;12:527–529.

Barkovich_Chap07.indd 799 5/6/2011 9:18:13 PM


800 Pediatric Neuroimaging

371. Shaw EG, Scheithauer BW, O’Fallon JR, Tazelaar HD, Davis DH. Oligo- 396. Nalesnik MA. Clinicopathologic characteristics of post-transplant lym-
dendrogliomas: the mayo clinic experience. J Neurosurg 1992;76:428–434. phoproliferative disorders. Recent Results Cancer Res 2002;159:9–18.
372. Cha S, Tihan T, Crawford F, et al. Differentiation of low-grade oligodendro- 397. Shroff R, Rees L. The post-transplant lymphoproliferative disorder: a lit-
gliomas from low-grade astrocytomas by using quantitative blood-volume erature review. Pediatr Nephrol 2004;19:369–377.
measurements derived from dynamic susceptibility contrast-enhanced 398. Brennan KC, Lowe LH, Yeaney GA. Pediatric central nervous system
MR imaging. Am J Neuroradiol 2005;26:266–273. posttransplant lymphoproliferative disorder. Am J Neuroradiol 2005;26:
373. Jenkinson MD, Smith TS, Joyce KA, et al. Cerebral blood volume, geno- 1695–1697.
type and chemosensitivity in oligodendroglial tumours. Neuroradiology 399. Castellano-Sanchez AA, Li S, Qian J, Lagoo A, Weir E, Brat D. Primary
2006;48:703–713. central nervous system posttransplant lymphoproliferative disorders. Am
374. Xu M, See SJ, Ng WH, et al. Comparison of magnetic resonance spectros- J Clinc Pathol 2004;121:246–253.
copy and perfusion-weighted imaging in presurgical grading of oligoden- 400. Kobayashi T, Kageyama N, Kida Y, Yoshida J, Shibuya N, Okamura K. Uni-
droglial tumors. Neurosurgery 2005;56:919–926. lateral germinomas involving the basal ganglia and thalamus. J Neurosurg
375. Ball RY, Treip CS. Intracranial extracerebral neuroglial hamartoma. Acta 1981;55:55–62.
Neuropathol (Berl) 1984;65:172–176. 401. Soejima T, Takeshita I, Yamamoto H, Tlukamoto Y, Fukui M, Matsuoka
376. Gyure KA, Morrison AL, Jones RV. Intracranial extracerebral neuroglial S. CT of germinomas in basal ganglia and thalamus. Neuroradiology
heterotopia: a case report and review of the literature. Ann Diagn Pathol 1987;29:366–370.
1999;3:182–186. 402. Komatsu Y, Narushima K, Kobayashi E, et al. CT and MR of germinoma in
377. Muzumdar D, Michaud J, Ventureyra ECG. Anterior cranial base glioneu- the basal ganglia. Am J Neuroradiol 1989;10:S9–S11.
ronal heterotopia. Childs Nerv Syst 2005:46. 403. Higano S, Takahashi S, Ishii K, Matsumoto K, Ikeda H, Sakamoto K. Ger-
378. Takhtani D, Melhem ER, Carson BS. Heterotopic cerebellum presenting as minoma originating in the basal ganglia and thalamus: MR and CT evalu-
a suprasellar mass with associated nasopharyngeal teratoma. Am J Neuro- ation. Am J Neuroradiol 1994;15:1435–1441.
radiol 2000;21:1119–1121. 404. Moon WK, Chang KH, Kim I-O, et al. Germinomas of the basal ganglia
379. Diehl B, Prayson R, Najm I, Ruggieri P. Hamartomas and epilepsy: clinical and thalamus: MR findings and a comparison between MR and CT. Am J
and imaging characteristics. Seizure 2003;12:307–311. Roentgenol 1994;162:1413–1417.
380. Nishio S, Mizuno J, Barrow DL, Takei Y, O’Brien MS. Intracranial extrac- 405. Okamoto K, Ito J, Ishikawa K, et al. Atrophy of the basal ganglia as the
erebral glioneural heterotopia. Childs Nerv Syst 1988;4:244–248. initial diagnostic sign of germinoma in the basal ganglia. Neuroradiology
381. Wolf H, Campos M, Zentner J, et al. Surgical pathology of temporal lobe 2002;44:389–394.
epilepsy. Experience with 216 cases. J Neuropathol Exp Neurol 1993;52: 406. Fujimaki T. Central nervous system germ cell tumors: classification,
499–506. clinical features, and treatment with a historical overview. J Child Neurol
382. Wolf H, Zentner J, Hufnagel A, et al. Surgical pathology of chronic epilep- 2009;24:1439–1445.
tic seizure disorders: experience with 63 specimens from extratemporal 407. Xu E, Wang X, Hao Z, Chen Z, Lu X. Germinoma in the basal ganglia with
corticectomies, lobectomies and functional hemispherectomies. Acta Neu- an abnormal karyotype: case report and review of the literature. Childs
ropathol (Berl) 1993;86:466–472. Nerv Syst 2010;26:707–712.
383. Zentner J, Hufnagel A, Wolf H, et al. Surgical treatment of temporal lobe 408. Jooma R, Kendall BE. Diagnosis and management of pineal tumors. J Neu-
epilepsy: clinical, radiological, and histopathological findings in 178 rosurg 1983;58:654–665.
patients. J Neurol Neurosurg Psychiatry 1995;58:666–673. 409. Casenave P, Eresue J, Guibert-Tranier F, Piton J, Caille JM. Tumors of the
384. Zentner J, Meyer B, Stangl A, Schramm J. Intrinsic tumors of the insula: a pineal region: diagnostic problems. J Neuroradiol 1984;11:47–63.
prospective surgical study of 30 patients. J Neurosurg 1996;85:263–271. 410. Rasalkar DD, Chu WCW, Cheng FWT, Paunipagar BK, Shing MK, Li
385. Roberts GA, Eldridge PR, Mackenzie JM. Case report: inflamma- CK. Atypical location of germinoma in basal ganglia in adolescents:
tory pseudotumour of the spine, with literature review. Br J Neurosurg radiological features and treatment outcomes. Br J Radiol 2010;83:
1997;11:570–572. 261–267.
386. Pettinato G, Manivel JC, DeRosa N, Dehner LP. Inflammatory myofi- 411. Liang L, Korogi Y, Sugahara T, et al. MRI of intracranial germ-cell tumors.
broblastic tumor (plasma cell granuloma). Clinicopathologic study of Neuroradiology 2002;44:382–388.
20 cases with immunohistochemical and ultrastructural observations. Am 412. Douglas-Akinwande AC, Ying J, Momin Z, Mourad A, Hattab EM. Diffu-
J Clin Pathol 1990;94:538–546. sion-weighted imaging characteristics of primary central nervous system
387. Bahadori M, Liebow AA. Plasma cell granulomas of the lung. Cancer germinoma with histopathologic correlation: a retrospective study. Aca-
1973;31:191–208. demic Radiol 2009;16:1356–1365.
388. Holodny AI, Kirsch CF, Hameed M, Sclar G. Tumefactive fibroinflamma- 413. Kim DI, Yoon PH, Ryu YH, Jeon P, J. HG. MRI of germinomas arising
tory lesion of the neck with progressive invasion of the meninges, skull from the basal ganglia and thalami. Neuroradiology 1998;40:507–511.
base, orbit, and brain. Am J Neuroradiol 2001;22:876–879. 414. Ozelame RV, Shroff M, Wood B, et al. Basal ganglia germinoma in children
389. McKinney AM, Short J, Lucato L, SantaCruz K, McKinney Z, Kim with associated ipsilateral cerebral and brain stem hemiatrophy. Pediatr
Y. Inflammatory myofibroblastic tumor of the orbit with associated Radiol 2006;36:325–330.
enhancement of the mininges and multiple cranial nerves. Am J Neurora- 415. Stemmer-Rachamimov A, Horgan M, Taratuto A, et al. Meningioangiom-
diol 2006;27:2217–2220. atosis is associated with neurofibromatosis 2 but not with somatic altera-
390. Tresser N, Rolf C, Cohen M. Plasma cell granulomas of the brain: pedi- tions of the NF2 gene. J Neuropathol Exp Neurol 1997;56:485–489.
atric case presentation and review of the literature. Childs Nerv Syst 416. Wiebe S, Munoz DG, Smith S, Lee DH. Meningioangiomatosis. A com-
1996;12:52–57. prehensive analysis of clinical and laboratory features. Brain 1999;122:
391. Sitton JE, Harkin JC, Gerber MA. Intracranial inflammatory pseudotu- 709–726.
mor. Clin Neuropathol 1992;11:36–40. 417. Aizpuru RN, Quencer RM, Norenberg M, Altman N, Smirniotopoulos M.
392. Hausler M, Schaade L, Ramaekers V, Doenges M, Heimann G, Sellhaus B. Meningioangiomatosis: clinical, radiologic, and histopathologic correla-
Inflammatory pseudotumors of the central nervous system: a report of tion. Radiology 1991;179:819–821.
3 cases and a literature review. Hum Pathol 2003;34:253–262. 418. Halper J, Scheithauer B, Okazaki H, et al. Meningio-angiomatosis: a report
393. Bramwit M, Kalina P, Rustia-Villa M. Inflammatory pseudotumor of the of six cases with special reference to the occurrence of neurofibrillary tan-
choroid plexus. Am J Neuroradiol 1997;18:1307–1309. gles. J Neuropath Exp Neurol 1986;45:426–446.
394. Harrington DS, Weisenberger DD, Purtilo DT. Malignant lymphoma in 419. Partington CR, Graves VB, Hegstrand LR. Meningioangiomatosis. Am J
the X-linked lymphoproliferative syndrome. Cancer 1987;59:1419–1429. Neuroradiol 1991;12:549–552.
395. Loren AW, Porter DL, Stadtmauer EA, Tsai DE. Post-transplant lymphop- 420. Kuzniecky R, Melanson D, Robitaille Y, Olivier A. Magnetic resonance
roliferative disorder: a review. Bone Marrow Transpl 2003;31:145–155. imaging of meningio-angiomatosis. Can J Neurol Sci 1988;15:161–164.

Barkovich_Chap07.indd 800 5/6/2011 9:18:13 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 801

421. Kim W, Kim I, Kim S, Cheon J, Yeon M. Meningioangiomatosis: MR 445. Kollias SS, Barkovich AJ, Edwards MSB. Magnetic resonance analysis of
imaging and pathological correlation in two cases. Pediatr Radiol suprasellar tumors of childhood. Pediatr Neurosurg 1991–1992;17:284–303.
2002;32:96–98. 446. Brown EW, Riccardi VM, Mawad M, et al. MR imaging of optic pathways
422. Mootha SL, Barkovich AJ, Grumbach MM, et al. Idiopathic hypothalamic in patients with neurofibromatosis. Am J Neuroradiol 1987;8:1031–1035.
diabetes insipidus, pituitary stalk thickening, and the occult intracra- 447. Arslanoglu A, Cirak B, Horska A, et al. MR imaging characteristics of pilo-
nial germinoma in children and adolescents. J Clin Endocrinol Metabol myxoid astrocytomas. Am J Neuroradiol 2003;24:1906–1908.
1997;82:1362–1367. 448. Kornreich L, Blaser S, Schwarz M, et al. Optic pathway glioma: correlation
423. Satogami N, Miki Y, Koyama T, Kataoka M, Togashi K. Normal pitu- of imaging findings with the presence of neurofibromatosis. Am J Neuro-
itary stalk: high-resolution MR imaging at 3T. Am J Neuroradiol 2010;31: radiol 2001;22:1963–1969.
355–359. 449. Sutton LN, Wang ZJ, Wehrli SL, et al. Proton spectroscopy of suprasellar
424. Maghnie M, Cosi G, Genovese E, et al. Central diabetes insipidus in chil- tumors in pediatric patients. Neurosurgery 1997;41:388–395.
dren and young adults. N Eng J Med 2000;343:998–1007. 450. Harrison MJ, Morgello S, Post KD. Epithelial cystic lesions of the sellar
425. Listernick R, Charrow J, Greenwald MJ, Mets M. Natural history of optic and parasellar region: a continuum of ectodermal derivatives? J Neurosurg
pathway tumors in children with neurofibromatosis type I: a longitudinal 1994;80:1018–1025.
study. J Pediatr 1994;125:63–66. 451. Erdheim J. Über Hypophysengangsgeschwulste und Hirncholesteatome.
426. Listernick R, Louis DN, Packer RJ, Gutmann DH. Optic pathway gliomas Sitzungsber Finn Akad Wiss 1904;113:537–726.
in children with neurofibromatosis 1: consensus statement from the NF1 452. Chen CJ. Suprasellar and infrasellar craniopharyngioma with a persistent
optic pathway glioma task force. Ann Neurol 1997;41:143–149. craniopharyngeal canal: case report and review of the literature. Neurora-
427. Takeuchi H, Kabuto M, Sato K, Kubota T. Chiasmal gliomas with spon- diology 2001;43:760–762.
taneous regression: proliferation and apoptosis. Childs Nerv Syst 1997;13: 453. Rutka JT, Hoffman HJ, Drake JM, Humphreys RP. Suprasellar and sellar
229–233. tumors in childhood and adolescence. In: Berger MS, ed. Pediatric Neuro-
428. Parsa CF, Hoyt CS, Lesser RL, et al. Spontaneous regression of optic oncology, vol 3. Philadelphia: Saunders, 1992:803–820.
gliomas: thirteen cases documented by serial neuroimaging. Arch Oph- 454. Hoffman HJ, da Silva M, Humphreys RP, et al. Craniopharyngioma in
thalmol 2001;119:516–529. children: fifteen years of management. J Neurosurg 1992;76:47–52.
429. Parazzini C, Triulzi F, Bianchini E, et al. Spontaneous involution of optic 455. De Vile CJ, Grant DB, Kendall BE, et al. Management of childhood cranio-
pathway lesions in neurofibromatosis type 1: serial contrast MR evalua- pharyngioma: can the morbidity of radical surgery be predicted? J Neuro-
tion. Am J Neuroradiol 1995;16:1711–1718. surg 1996;85:73–81.
430. Piccirilli M, Lenzi J, Delfinis C, Trasimeni G, Salvati M, Raco A. Spontane- 456. Sartoretti-Schefer S, Wichmann W, Aguzzi A, Valavanis A. MR differentia-
ous regression of optic pathways gliomas in three patients with neuro- tion of adamantinous and squamous-papillary craniopharyngiomas. Am
fibromatosis type I and critical review of the literature. Childs Nerv Syst J Neuroradiol 1997;18:77–87.
2006;22:1332-1337. 457. Miller DC. Pathology of craniopharyngiomas: clinical import of patho-
431. Pascual-Castroviejo I, Pascual-Pascual SI, Velázquez-Fragua R, Viaño J, logical findings. Pediatr Neurosurg 1994;21(Suppl):11–17.
García-Segura JM, Botella MP. Neurofibromatosis tipo 1 y gliomas de vÌas 458. Crotty TB, Scheithauer BW, Young WFJ, et al. Papillary craniopharyngioma:
Ûpticas. Una serie de 80 pacientes. Rev Neurol 2008;46:530–536. a clinicopathological study of 48 cases. J Neurosurg 1995;83:206–214.
432. Balcer L, Liu G, Heller F, et al. Visual loss in children with neurofibro- 459. Brunel H, Raybaud C, Peretti-Viton P, et al. Craniopharyngioma in chil-
matosis type 1 and optic pathway gliomas: relation to tumor location by dren: MRI study of 43 cases. Neurochirurgie 2002;48:309–318.
magnetic resonance imaging. Am J Ophthalmol 2001;131:442–445. 460. Kanungo N, Just N, Black M, Mohr G, Glikstein R, rochon L. Nasopha-
433. Medlock MD, Madsen JR, Barnes PD, Anthony DS, Cohen LE, Scott RM. ryngeal craniopharyngioma in an unusual location. Am J Neuroradiol
Optic chiasm astrocytomas of childhood. Pediatr Neurosurg 1997;27: 1995;16:1372–1374.
121–128. 461. Kawamata T, Kubo O, Kamikawa S, Hori T. Ectopic clival craniopharyn-
434. Russell A. A diencephalic syndrome of emaciation in infancy and child- gioma. Acta Neuropathol (Wien) 2002;144:1221–1224.
hood. Arch Dis Child 1951;26:274. 462. Pettorini B, Inzitari R, Massimi L, et al. The role of inflammation in the
435. Stern J, DiGiancinto GV, Housepian EM. Neurofibromatosis and genesis of the cystic component of craniopharyngiomas. Childs Nerv Syst
optic glioma. Clinical and morphological correlations. Neurosurgery 2010:1–6.
1979;4:524–527. 463. Fitz CR, Wortzman G, Harwood-Nash DC. CT in craniopharyngiomas.
436. Gallucci M, Catalucci A, Scheithauer B, Forbes G. Spontaneous involution Radiology 1978;127:687–691.
of pilocytic astrocytoma in a patient without neurofibromatosis type 1: 464. Ahmadi J, Destian S, Apuzzo MLJ, Segall HD, Zee C-S. Cystic fluid in
case report. Radiology 2000;214:223–226. craniopharyngiomas: MR imaging and quantative analysis. Radiology
437. Colosimo C, Cerase A, Maira G. Regression after biopsy of a pilocytic opti- 1992;182:783–785.
cochiasmatic astrocytoma in a young adult without neurofibromatosis. 465. Pusey E, Kortman KE, Flannigan BD, et al. MR of craniopharyngiomas:
Neuroradiology 2000;42:352–356. tumor delineation and characterization. Am J Neuroradiol 1987;8:439–445.
438. Ito S, Hoshino T, Shibuya M, Prados M, Edwards M, Davis R. Proliferative 466. Young SC, Zimmerman RA, Nowell MA, et al. Giant cystic craniopharyn-
characteristics of juvenile pilocytic astrocytoma determined by bromode- giomas. Neuroradiology 1987;29:468–473.
oxyuridine labeling. Neurosurgery 1992;31:413–419. 467. Kiran N, Suri A, Kasliwal M, Garg A, Ahmad F, Mahapatra A. Gross total
439. Alvord EC, Lufton S. Gliomas of the optic nerve or chiasm: outcome by excision of pediatric giant cystic craniopharyngioma with huge retro-
patient’s age, tumor site, and treatment. J Neurosurg 1988;68:85–98. clival extension to the level of foramen magnum by anterior trans petrous
440. Komotar R, Burger P, Carson B, et al. Pilocytic and pilomyxoid hypotha- approach: report of two cases and review of literature. Childs Nerv Syst
lamic/chiasmatic astrocytomas. Neurosurgery 2004;54:72–79. 2008;24:385–391.
441. Simon JH, Szumowski J, Totterman S, et al. Fat suppression MR imaging 468. Rush JL, Kusske JA, De Feo DR, et al. Intraventricular craniopharyngioma.
of the orbit. Am J Neuroradiol 1988;9:961–968. Neurology 1975;25:1094–1096.
442. Imes RK, Hoyt WF. MR imaging signs of optic nerve gliomas in neurofi- 469. Pascual JM, González-Llanos F, Barrios L, Roda JM. Intraventricular cranio-
bromatosis 1. Am J Ophthalmol 1991;111:729–734. pharyngiomas: topographical classification and surgical approach selection
443. Savoiardo M, Harwood-Nash DC, Tadmor R, Scotti G, Musgrave MA. based on an extensive overview. Acta Neurochirurgica 2004;146:785–802.
Gliomas of the intracranial anterior optic pathways in children. Radiology 470. Kornreich L, Horev G, Blaser S, Daneman D, Kauli R, Grunebaum M.
1981;138:601–610. Central precocious puberty: evaluation by neuroimaging. Pediatr Radiol
444. Fletcher WA, Imes RK, Hoyt WF. Chiasmal gliomas. Appearance and 1995;25:7–11.
long term changes demonstrated by computed tomography. J Neurosurg 471. Ng S, Kumar Y, Cody D, Smith C, Didi M. Cranial MRI scans are indicated
1986;65:154–159. in all girls with central precicious puberty. Arch Dis Child 2003;88:414–418.

Barkovich_Chap07.indd 801 5/6/2011 9:18:13 PM


802 Pediatric Neuroimaging

472. Mullatti N, Selway R, Nashef L, et al. The clnical spectrum of epilepsy 496. Boyko OB, Curnes JT, Oakes WJ, Burger PC. Hamartomas of the tuber
in children and adults with hypothalamic hamartoma. Epilepsia cinereum: CT, MR, and pathologic findings. Am J Neuroradiol 1991;12:
2003;44:1310–1319. 309–314.
473. Arzimanoglou AA, Hirsch E, Aicardi J. Hypothalamic hamartoma in epi- 497. Amstutz DR, Coons SW, Kerrigan JF, Rekate HL, Heiserman JE. Hypotha-
lepsy in children: illustrative cases of possible evolutions. Epileptic Disord lamic hamartomas: correlation of MR imaging and spectroscopic findings
2003;5:187–199. with tumor glial content. Am J Neuroradiol 2006;27:794–798.
474. Mahachoklertwattana P, Kaplan S, Grumbach MM. The luteinizing 498. Hahn FJ, Leibrock LG, Huseman CA, Makos MM. The MR appearance of
hormone-relaeasing hormon-secreting hypothalamic hamartoma is hypothalamic hamartomas. Neuroradiology 1988;30:65–68.
really a congenital malformation: natural history. J CLin Endocrin Metab 499. Markin RS, Leibrock LG, Huseman CA, McComb RD. Hypothalamic
1993;77:118–124. hamartoma: a report of 2 cases. Pediatr Neurosci 1987;13:19–26.
475. Freeman JL, Coleman LT, Wellard RM, et al. MR imaging and spectro- 500. Nishio S, Fujiwara S, Aiko Y, Takeshita I, Fukui M. Hypothalamic hamar-
scopic study of epileptogenic hypothalamic hamartomas: analysis of 72 toma: report of two cases. J Neurosurg 1989;70:640–645.
cases. Am J Neuroradiol 2004;25:450–462. 501. Martin D, Seeger U, Ranke M, Grodd W. MR imaging and spectroscopy of a
476. Andermann F, Arzimanoglou A, Berkovic SF. Hypothalamic hamartoma tuber cinereum hamartoma in a patient with growth hormone deficiency and
and epilepsy: the pathway of discovery. Epileptic Disord 2003;5:173–176. hypogonadotropic hypogonadism. Am J Neuroradiol 2003;24:1177–1180.
477. Striano S, Striano P, Cirillo S, et al. Small hypothalamic hamartomas and 502. Felig P, Baxter JD, Broadus AJ, Frohman LA. Endocrinology and metabo-
gelastic seizures. Epileptic Disord 2002;4:129–133. lism. In: Robertson GL, ed. Posterior Pituitary. New York: McGraw-Hill,
478. Clarren S, Alvord E, Jr, Hall J. Congenital hypothalamic hamartoblastoma, 1987:338–385.
hypopituitarism, imperforate anus, and postaxial polydactyly–a new 503. Dunger D, Broadbent V, Yeoman E. The frequency and natural history of
syndrome? Part II: Neuropathological considerations. Am J Med Genet diabetes insipidus in children with Langerhan-cell histiocytosis. N Eng J
1980;7:75–83. Med 1989;321:1157–1162.
479. Hall J, Pallister P, Clarren S, et al. Congenital hypothalamic hamartoblas- 504. Grois N, Fahrner B, Arceci R, et al. Central nervous system disease in
toma, hypopituitarism, imperforate anus and postaxial polydactyly–a new Langerhans cell histiocytosis. J Pediatr 2010;156:873–881.
syndrome? Part I: clinical, causal, and pathogenetic considerations. Am J 505. Kim E-Y, Choi J-U, Kim T-S, Kim D-I, Kim K-Y. Huge Langerhans cell
Med Genet 1980;7:47–74. histiocytosis granuloma of choroid plexus in a child with Hand-Schüller-
480. Diebler C, Ponsot G. Hamartomas of the tuber cinereum. Neuroradiology Christian disease. J Neurosurg 1995;83:1080–1084.
1983;25:93–101. 506. Gizewski ER, Forsting M. Histiocytosis mimicking a pineal tumor. Neuro-
481. Sato M, Ushio Y, Arita N, Mogami H. Hypothalamic hamartoma: report of radiology 2001;43:644–646.
two cases. Neurosurgery 1985;16:198–206. 507. Hamilton B, Connolly ES, Mitchell WT. Isolated intramedullary histiocy-
482. Albright AL, Lee PA. Neurosurgical treatment of hypothalamic hamar- tosis X of the cervical spinal cord. J Neurosurg 1995;83:716–718.
tomas causing precocious puberty. J Neurosurg 1993;78:77–82. 508. Manelfe C, Louvet JP. CT in diabetes insipidus. J Comput Assist Tomogr
483. Munari C, Kahane P, Francione S. Role of the hypothalamic hamar- 1979;3:309–316.
toma in the genesis of gelastic fits. Electroencephalogr Clin Neurophysiol 509. Tien R, Newton TH, McDermott MW, Dillon WP, Kucharczyk J. Thick-
1995;95:154–160. ened pituitary stalk on MR images in patients with diabetes insipidus and
484. Kuzniecky RI, Guthrie B, Mountz J. Intrinsic epileptogenesis of hypotha- Langerhans cell histiocytosis. Am J Neuroradiol 1990;11:703–708.
lamic hamartomas in gelastic epilepsy. Ann Neurol 1997;42:60–67. 510. Maghnie M, Arico M, Villa A, Genovese E, Beluffi G, Severi F. MR of the
485. Deonna T, Ziegler A-L. Hypothalamic hamartoma, precocious puberty, hypothalamic-pituitary axis in Langerhans cell histiocytosis. Am J Neuro-
and gelastic seizures: a special model of epileptic developmental disorder. radiol 1992;13:1365–1371.
Epileptic Disord 2000;2:33–37. 511. Haddad SF, Van Gilder JC, Menezes AH. Pediatric pituitary tumors. Neu-
486. Palmini A, Chandler C, Andermann F, et al. Resection of the lesion in rosurgery 1991;29:509–514.
patients with hypothalamic hamartomas and catastrophic epilepsy. Neu- 512. Laws ER, Sheithauer BW, Groover RV. Pituitary adenomas in childhood
rology 2002;58:1338–1347. and adolescence. Prog Exp Tumor Res 1987;30:359–361.
487. Polkey C. Resective surgery for hypothalamic hamartoma. Epileptic Disord 513. Katavetin P, Cheunsuchon P, Swearingen B, Hedley-Whyte ET, Misra M,
2003;5:281–286. Levitsky LL. Review: pituitary adenomas in children and adolescents.
488. Kuzniecky RI, Guthrie BL. Steriotactic surgical approach to hypothalamic J Pediatr Endocrinol Metab 2010;23:427–431.
hamartomas. Epileptic Disord 2003;5:275–280. 514. Abe T, Lüdecke DK, Saeger W. Clinically nonsecreting pituitary adenomas
489. Harvey AS, Freeman JL, Berkovic SF, Rosenfeld JV. Transcallosal resection in childhood and adolescence. Neurosurgery 1998;42:744–751.
of hypothalamic hamartomas in patients with intractable epilepsy. Epilep- 515. Fisher B, Gaspar L, Stitt L, Noone B. Pituitary adenoma in adolescents: a
tic Disord 2003;5:257–265. biologically more aggressive disease? Radiology 1994;192:869–872.
490. Delalande O, Fohlen M. Disconnecting surgical treatment of hypotha- 516. Richmond IL, Wilson CB. Pituitary adenomas in childhood and adoles-
lamic hamartoma in children and adults with refractory epilepsy and cence. J Neurosurg 1978;49:163–168.
proposal of a new classification. Neurologia medico-chirurgica 2003;43: 517. Bills DC, Meyer FB. A retrospective analysis of pituitary apoplexy. Neuro-
61–68. surgery 1993;33:602–609.
491. Frazier J, Goodwin C, Ahn E, Jallo G. A review on the management of 518. Shah S, Pereira J, Becker C, Aronin P. Pituitary apoplexy in adolescence:
epilepsy associated with hypothalamic hamartomas. Childs Nerv Syst case report. Pediatr Radiol 1995;25:S26–S27.
2009;25:423–432. 519. Newton DR, Dillon WP, Norman D, et al. Gd-DTPA-enhanced MR imag-
492. Guibaud L, Rode V, Saint-Pierre G, Procros J-P, Foray P, Tran-Minh V. ing of pituitary adenomas. Am J Neuroradiol 1989;10:949–954.
Giant hypothalamic hamartoma: an unusual neonatal tumor. Pediatr 520. Kucharczyk W, Davis DO, Kelly WM, Sze G, Norman D, Newton TH.
Radiol 1995;25:17–18. Pituitary adenomas: high resolution MR imaging at 1.5 T. Radiology
493. Valdueza J, Cristante L, Dammann O, et al. Hypothalamic hamartomas: 1986;161:761–765.
with special reference to gelastic epilepsy and surgery. Neurosurgery 521. Sakamoto Y, Takahashi M, Korogi Y, Bussaka H, Ushio Y. Normal and
1994;34:949–958. abnormal pituitary glands: gadopentate dimeglumine-enhanced MR
494. Debeneix C, Bourgeois M, Trivin C, Sainte-Rose C, Brauner R. Hypotha- imaging. Radiology 1991;178:441–445.
lamic hamartoma: comparison of clinical presentation and magnetic reso- 522. Bartynski W, Lin L. Dynamic and conventional spin-echo MR of pituitary
nance images. Horm Res 2001;56:12–18. microlesions. Am J Neuroradiol 1997;18:965–972.
495. Lin S-R, Bryson MM, Goblen RP, Fitz CR, Lee Y-Y. Radiologic findings 523. Kanou Y, Arita K, Kurisu K, Tomohide A, Iida K. Clinical implications
of hamartomas of the tuber cinereum and hypothalamus. Radiology of dynamic MRI for pituitary adenomas: clinical and histologic analysis.
1978;127:697–703. J Clin Neurosci 2002;9:659–663.

Barkovich_Chap07.indd 802 5/6/2011 9:18:14 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 803

524. Elster AK, Chen MYM, Williams DW, III, Key LL. Pituitary gland: 550. Tien RD, Barkovich AJ, Edwards MSB. MR imaging of pineal tumors. Am
MR imaging of physiologic hypertrophy in adolescence. Radiology J Neuroradiol 1990;11:557–565.
1990;174:681–685. 551. Kyritsis A. Management of primary intracranial germ cell tumors. J Neuro-
525. Young-Poussaint T, Barnes PD, Anthony CC, Spack N, Scott RM, Tarbell Oncol 2010;96:143–149.
NJ. Hemorrhagic pituitary adenomas of adolescence. Am J Neuroradiol 552. Baumgartner JE, Edwards MSB. Pineal tumors. In: Berger MS, ed. Pediat-
1996;17:1907–1912. ric Neuro-oncology, vol 3. Philadelphia: Saunders, 1992:853–862.
526. Voelker JL, Campbell RL, Muller J. Clinical, radiographic, and patho- 553. Edwards MSB, Hudgins RJ, Wilson CB, Levin VA, Wara WM. Pineal region
logic features of symptomatic Rathke’s cleft cysts. J Neurol Sci 1991;74: tumors in children. J Neurosurg 1988;68:689–697.
535–544. 554. Vaquero J, Ramiro J, Martinez R, Coca S, Bravo G. Clinicopathological
527. Takanashi J, Tada H, Barkovich AJ, Saeki N, Kohno Y. Pituitary cysts in experience with pineocytomas: report of five surgically treated cases.
childhood evaluated by MR imaging. Am J Neuroradiol 2005;26:2144– Neurosurgery 1990;27:612–618.
2147. 555. D’Andrea AD, Packer RJ, Rorke LB, et al. Pineocytomas of child-
528. Hirano A, Hirano M. Benign cysts in the central nervous system: neuro- hood. A reappraisal of natural history and response to therapy. Cancer
pathological observations of the cyst walls. Neuropathology 2004;24:1–7. 1987;59:1353–1357.
529. Okamoto S, Handa H, Yamashita J, Ishikawa M, Nagasawa S. CT in intra- 556. Cuccia V, Rodríguez F, Palma F, Zuccaro G. Pinealoblastomas in children.
and suprasellar epithelial cysts (symptomatic Rathke cleft cysts). Am J Childs Nerv Syst 2006;22:577–585.
Neuroradiol 1985;6:515–519. 557. Zimmerman RA, Bilaniuk LT, Wood JH, Bruce DA, Schut L. CT of
530. Kucharczyk W, Peck WW, Kelly WM, Norman D, Newton TH. Rathke pineal, parapineal, and histologically related tumors. Radiology 1980;137:
cleft cysts: CT, MR imaging, and pathologic features. Radiology 1987;165: 669–677.
491–496. 558. Nakamura M, Saeki N, Iwadate Y, Sunami K, Osato K, Yamaura A. Neuro-
531. Israel ZH, Yacoub M, Gomori JM, et al. Rathke’s cleft cyst abscess. Pediatr radiological characteristics of pineocytoma and pineoblastoma. Neurora-
Neurosurg 2000;33:159–161. diology 2000; 42:509–514.
532. Nakamura Y, Okada H, Wada Y, Kajiyama K, Koshiyama H. Lymphocytic 559. De Campo MP, Davis MD. The role of CT in the diagnosis and man-
hypophysitis: its expanding features. J Endocrinol Invest 2001;24:262–267. agement of childhood pineal region tumours. Australas Radiol 1991;35:
533. Kamel N, Ilgin S, Gullu S, Tonyukuk V, Deda H. Lymphocytic hypophysitis 336–339.
and infundibuloneurohypophysitis; clinical and pathological evaluations. 560. Chang T, Teng MM, Guo WY, Sheng WC. CT of pineal tumors and intrac-
Endocr J 1999;46:505–512. ranial germ-cell tumors. Am J Neuroradiol 1989;10:1039–1044.
534. Gellner V, Kurschel S, Scarpatetti M, Mokry M. Lymphocytic hypophysitis 561. Muller-Forell W, Schroth G, Egan PJ. MR imaging in tumors of the pineal
in the pediatric population. Childs Nerv Syst 2008;24:785–792. region. Neuroradiology 1988;30:224–231.
535. Imura H, Nakao K, Shimatsu A, et al. Lymphocytic infundibuloneu- 562. Zee CS, Segall H, Apuzzo M, et al. MR imaging of pineal region neoplasms.
rohypophysitis as a cause of central diabetes insipidus. N Eng J Med J Comput Assist Tomogr 1991;15:56–63.
1993;329:683–689. 563. Nakagawa H, Iwasaki S, Kichikawa K, et al. MR imaging of pineocytoma:
536. Honegger J, Fahlbusch R, Bornemann A, et al. Lymphocytic and gran- report of two cases. Am J Neuroradiol 1990;11:195–198.
ulomatous hypophysitis: experience with nine cases. Neurosurgery 564. Rippe D, Boyko OB, Friedman HS, et al. Gd-DTPA-enhanced MR imaging
1997;40:713–723. of leptomeningeal spread of primary intracranial CNS tumor in children.
537. Feigenbaum SI, Martin C, Wilson CB, Jaffe RB. Lymphocytic adenohy- Am J Neuroradiol 1990;11:329–332.
pophysitis: a pituitary mass lesion occurring in pregnancy. Am J Obstet 565. Zang X, Nilaver G, Stein BM, Fetell MR, Duffy PE. Immunocytochemis-
Gynecol 1991;164:1549–1555. try of pineal astrocytes: species differences and functional implications.
538. Tien R, Kucharczyk J, Kucharczyk W. MR imaging of the brain in patients J Neuropath Exp Neurol 1985;44:486–495.
with diabetes insipidus. Am J Neuroradiol 1991;12:533–542. 566. Boydston WR, Sanford RA, Muhlbauer MS, et al. Gliomas of the tectum
539. Hoffman HJ, Otsubo H, Hendrick EB, et al. Intracranial germ-cell tumors and periaqueductal region of the mesencephalon. Pediatr Neurosurg
in children. J Neurosurg 1991;74:545–551. 1991–1992;17:234–238.
540. Felix I, Becker LE. Intracranial germ cell tumors in children: an immu- 567. Megyeri L. Cystische Veränderungen des Corpus pinale. Frankfurt Z Pathol
nohistochemical and electron microscopic study. Pediatr Neurosci 1990– 1960;70:699–704.
1991;16:156–162. 568. Hasegawa A, Ohtsubo K, Mori W. Pineal gland in old age: quantitative
541. Matsutani M, Sano K, Takakura K, et al. Primary intracranial germ cell and qualitative morphological study of168 human autopsy cases. Brain
tumors: a clinical analysis of 153 histologically verified cases. J Neurosurg Res 1987;409:343–349.
1997;86:446–455. 569. Cooper ER. The human pineal gland and pineal cysts. J Anat 1932;67:28–46.
542. Lantos PL, Louis DN, Rosenblum MK. Germ cell tumors. In: Graham 570. Sawamura Y, Ikeda J, Ozawa M, Minoshima Y, SaitomH, Abe H. Magnetic
DI, Lantos PL, eds. Greenfield’s Neuropathology, 7th ed. London: Arnold, resonance images reveal a high incidence of asymptomatic pineal cysts in
2002:944–945. young women. Neurosurgery 1995;37:11–16.
543. Matsuoka S, Itoh M, Shimonome T, et al. Intramedullary spinal cord 571. Klein P, Rubenstein LJ. Benign symptomatic glial cysts of the pineal gland:
germinoma. Surg Neurol 1991;35:122–126. a report of seven cases and review of the literature. J Neurol Neurosurg
544. Yasue M, Tanaka H, Nakajima M, et al. Germ cell tumors of the basal Psychiatry 1989;52:991–995.
ganglia and thalamus. Pediatr Neurosurg 1993;19:121–126. 572. Mamourian AC, Towfighi J. Pineal cysts: MR imaging. Am J Neuroradiol
545. Miyauchi A, Matsumoto K, Kohmura E, Doi T, Hashimoto K, Kawano K. 1986;7:1081–1086.
Primary intramedullary spinal cord germinoma. J Neurosurg 1996;84: 573. Lee DH, Norman D, Newton TH. MR imaging of pineal cysts. J Comput
1060–1061. Assist Tomogr 1987;11:586–590.
546. Bailey S, Skinner R, Lucraft HH, Perry RH, Todd N, Pearson ADJ. Pineal 574. Golzarian J, Balériaux D, Bank WO, Matos C, Flament-Durand J. Pineal
tumours in the north of England 1968–1993. Arch Dis Child 1996;75: cyst: normal or pathological? Neuroradiology 1993;35:251–253.
181–185. 575. Pu Y, Mahankali S, Hou J, et al. High prevalence of pineal cysts in healthy
547. Shokry A, Janzer RC, von Hochstetter AR, et al. Primary intracranial adults demonstrated by high-resolution, noncontrast brain MR imaging.
germ cell tumors: a clinicopathological study of 14 cases. J. Neurosurg Am J Neuroradiol 2007;28:1706–1709.
1985;62:826–830. 576. Barboriak DP, Lee L, Provanzale JM. Serial MR imaging of pineal cysts. Am
548. Jennings MT, Gelman R, Hochberg F. Intracranial germ cell tumors: J Roentgenol 2001;176:737–743.
natural history and pathogenesis. J Neurosurg 1985;63:155–167. 577. Fleege M, Miller G, Fletcher G, Fain J, Scheithauer B. Benign glial cysts of
549. Kilgore DP, Strother CM, Starshak RJ, Haughton VM. Pineal germinoma: the pineal gland: unusual imaging characteristics with histologic correla-
MR imaging. Radiology 1986;158:435–438. tion. Am J Neuroradiol 1994;15:161–166.

Barkovich_Chap07.indd 803 5/6/2011 9:18:14 PM


804 Pediatric Neuroimaging

578. Mamourian AC, Yarnell T. Enhancement of pineal cysts on MR images. 605. Lund-Johansen M, Scheie D, Muller T, Lundar T, Helseth E. Neurosurgical
Am J Neuroradiol 1991;12:773–774. treatment of meningiomas in children and young adults. Childs Nerv Syst
579. Tomita T, McLone DG, Flannery AM. Choroid plexus papillomas of 2001;17:719–723.
neonates, infants, and children. Pediatr Neurosci 1988;14:23–30. 606. Hadfield MG, Quezado MM, Williams RL, Luo VY. Ewing’s family of
580. St. Clair SK, Humphreys RP, Pillay PK, Hoffman HJ, Blaser SI, Becker LE. tumors involving structures related to the central nervous system: a review.
Current management of choroid plexus carcinoma in children. Pediatr Pediatr Dev Pathol 2000;3:203–210.
Neurosurg 1991–1992;17:225–233. 607. Davidson GS, Hope JK. Meningeal tumors of childhood. Cancer
581. Ellenbogen RG, Winston KR, Kupsky WJ. Tumors of the choroid plexus in 1989;63:1205–1210.
children. Neurosurgery 1989;25:327–335. 608. Özhan S, Tali ET, Isik S, Saygili MR, Baykaner K. Haematoma-like primary
582. Boyd MD, Steinbok P. Choroid plexus tumors: problems in diagnosis and intracranial malignant fibrous histiocytoma in a 5-year-old girl. Neurora-
management. J Neurosurg 1987;66:800–805. diology 1999;41:523–525.
583. Berger C, Thiesse P, Lellouch-Tubiana A, Kalifa C, Pierre-Kahn A, 609. Sano K, Wakai S, Ochiai C. Characteristics of meningiomas in childhood.
Bouffet E. Choroid plexus carcinomas in childhood: clinical features and Child’s Brain 1981;8:98–106.
prognostic factors. Neurosurgery 1998;42:470–475. 610. Kinoshita Y, Kajiwara H, Yokota A, Koga Y. Proton magnetic resonance
584. Martin N, Pierot L, Sterkers O, Mompoint D, Nahum H. Primary choroid spectroscopy of brain tumors: an in vitro study. Neurosurgery 1994;35:
plexus palilloma of the cerebellopontine angle: MR imaging. Neuroradiol- 606–614.
ogy 1990;31:541–543. 611. Majos S, Alonso J, Aguilera C, et al. Utility of proton MR spectroscopy in
585. Coates TL, Hinshaw DB, Jr, Peckman N, et al. Pediatric choroid plexus the diagnosis of radiologically atypical intracranial meningiomas. Neuro-
neoplasms: MR, CT, and pathologic correlations. Radiology 1989;173: radiology 2003;45:129–136.
81–88. 612. Howe F, Opstad K. 1H MR spectroscopy of brain tumors and masses.
586. Kendall B, Rerder-Grosswasser I, Valentine A. Diagnosis of masses pre- NMR Biomed 2003;16:123–131.
senting within the ventricles on computed tomography. Neuroradiology 613. Hasegawa M, Kida S, Yamashima T, Yamashita J, Takakuwa S. Multicentric
1983;25:11–22. infantile myofibromatosis in the cranium: case report. Neurosurgery
587. Meyers SP, Khademian ZP, Chuang SH, Pollack IF, Korones DN, 1995;36:1200–1203.
Zimmerman RA. Choroid plexus carcinomas in children: MRI features 614. Laningham FH, Kun L, Reddick WE, Ogg RJ, Morris EB, Pui C-H. Child-
and patient outcomes. Neuroradiology 2004;46:770–780. hood central nervous system leukemia: historical perspectivies, cur-
588. Akil H, Coupe NJ, Singh J. Spinal deposits of a benign choroid plexus rent therapy, and acute neurological sequelae. Neuroradiology 2007;49:
papilloma. J Clin Neurosci 2008;15:708–712. 873–888.
589. Horska A, Ulug A, Melham E, et al. Proton magnetic resonance spec- 615. Kretchmer K, Gutjahr P, Kutzner J. CT studies before and after CNS treat-
troscopy of choroid plexus tumors in children. J Magn Reson Imaging ment for acute lymphocytic leukemia and malignant non-Hodgkins lym-
2001;14:78–82. phoma in childhood. Neuroradiology 1980;20:173–180.
590. Drake JM, Hedrick EB, Becker LE, et al. Intracranial meningiomas in 616. Vázquez E, Lacaya J, Castellote A, et al. Neuroimaging in pediatric leuke-
children. Pediatr Neurosci 1986;12:134–139. mia and lymphoma: differential diagnosis. Radiographics 2002;22:1411–
591. Hope JKA, Armstrong DA, Babyn PS, et al. Primary meningeal tumors in 1428.
children: correlation of clinical and CT findings with histologic type and 617. Chen C, Zimmerman R, Faro S, Bilaniuk L, Chou T, Molloy P. Childhood
prognosis. Am J Neuroradiol 1992;13:1353–1364. leukemia: central nervous system abnormalities during and after treat-
592. Perilongo G, Sutton L, Goldwein JW, et al. Childhood meningiomas: expe- ment. Am J Neuroradiol 1996;17:295–310.
rience in the modern era. Pediatr Neurosurg 1992;18:16–23. 618. Reddick WE, Laningham FH, Glass JO, Pui C-H. Quantitative morpho-
593. Amirjamshidi A, Mehrazin M, Abbassioun K. Meningiomas of the central logic evaluation of magnetic resonance imaging during and after treat-
nervous system occurring below the age of 17: report of 24 cases not asso- ment of childhood leukemia. Neuroradiology 2007;49:889-904.
ciated with neurofibromatosis and review of literature. Childs Nerv Syst 619. Chourmouzi D, Pistevou-Gompaki K, Plataniotis G, Skaragas G, Papa-
2000;16:406–416. dopoulos L, Drevelegas A. MRI findings of extramedullary haemopoiesis.
594. Al-Gahtany M, Shroft M, Bouffet E, et al. Primary central nervous sys- Eur Radiol 2001;11:1803–1806.
tem sarcomas in children: clinical, radiological, and pathological features. 620. Knoblich R. Extramedullary hematopoiesis presenting as intrathoracic
Childs Nerv Syst 2003;19:808–817. tumors. Report of a case in a patient with thalassemia minor. Cancer
595. Rutigliano M, Pollack I, Ahdab-Barmada M, Pang D, Albright AL. Intrac- 1960;13:462–468.
ranial infantile myofibromatosis. J Neurosurg 1994;81:539–543. 621. Lyall A. Massive extramedullary bone marrow formation in a case of
596. Pekala JS, Gururangan S, Provenzale J, Mukundan S, Jr. Central nervous pernicious anemia. J Pathol Bacteriol 1935;41:469–472.
system extraosseous Ewing sarcoma: radiologic manifestations of this 622. Haidar S, Ortiz-Neira C, Shroff M, Gilday D, Blaser S. Intracranial involve-
newly defined pathologic entity. Am J Neuroradiol 2006;27:580–583. ment in extramedullary hematopoiesis: case report and review of the lit-
597. Hari JK, Azzarelli B, Caldemeyer KS. Malignant fibrous histiocytoma in a erature. Pediatr Radiol 2005;35:630–634.
9-year-old girl. Pediatr Neurosurg 1996;24:160–166. 623. De Greeter F, Van Retenghem D. Scintigraphic diagnosis of intrathoracic
598. Reyes-Mugica M, Chou P, Gonzalez-Crussi F, Tomita T. Fibroma of extramedullary hematopoiesis in alcohol related macrocytosis. J Nucl Med
the meninges in a child: immunohistological and ultrastructural study. 1996;37:473–475.
J Neurosurg 1992;76:143–147. 624. Jooma R, Kendall BE. Intracranial tumors in the first year of life. Neurora-
599. Artico M, Ferrante L, Cervoni L, Collonese C, Fortuna A. Pediatric cystic diology 1982;23:267–274.
meningioma: report of three cases. Childs Nerv Syst 1995;11:137–140. 625. Geyer JR. Infant brain tumors. In: Berger MS, ed. Pediatric Neuro-oncol-
600. Reddy DR, Kolluri VRS, Rao KS, et al. Cystic meningiomas in children. ogy, vol 3. Philadelphia: Saunders, 1992:781–791.
Childs Nerv Syst 1986;2:317–319. 626. DiRocco C, Ceddia A, Iannelli A. Intracranial tumors in the first year of
601. Starshak RJ. Cystic meningiomas in children: a diagnostic challenge. life. A report on 51 cases. Acta Neurochir (Wien) 1993;123:14–24.
Pediatr Radiol 1996;26:711–714. 627. Brown K, Mapstone TB, Oakes WJ. A modern analysis of intracranial
602. Kim S, Wang K, Kim S, et al. Childhood meningioma: unusual location, tumors of infancy. Pediatr Neurosurg 1997;26:25–32.
atypical radiological findings, and favorable treatment outcome. Childs 628. Balestrini MR, Micheli R, Giordano L, Lasio G, Giombini S. Brain tumors
Nerv Syst 2001;17:656–662. with symptomatic onset in the first two years of life. Childs Nerv Syst
603. Samson Sujit Kumar G, Rajshekhar V. Deep sylvian meningioma: a case 1994;10:104–110.
report and review of literature. Child Nerv Syst 2009;25:129–132. 629. Mehrotra N, Shamji M, Vassilyadi M, Ventureyra E. Intracranial tumors
604. Finizio FS. Aspetti clinici e neuroradiologici dei meningiomi negli adoles- in first year of life: the CHEO experience. Childs Nerv Syst 2009;25:1563–
centi e nei giovani. Rivista di Neuroradiologia 1996;9:349–354. 1569.

Barkovich_Chap07.indd 804 5/6/2011 9:18:14 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 805

630. Severino M, Schwartz E, Thurnher M, Rydland J, Nikas I, Rossi A. Con- 656. Shields C, Meadows A, Shields J, Carvalho C, Smith A. Chemoreduction
genital tumors of the central nervous system. Neuroradiology 2010;52: for retinoblastoma may prevent intracranial neuroblastic malignancy
531–548. (trilateral retinoblastoma). Arch Ophthalmol 2001;119:1269–1272.
631. Serowka K, Chiu Y, Gonzalez I, et al. Central nervous system (CNS) tumors 657. Morgan G. Diffuse infiltrating retinoblastoma. Br J Ophthalmol 1971;55:
in the first six months of life: The Childrens Hospital Los Angeles Experi- 600–611.
ence, 1979 - 2005. Pediatr Hematol-Oncol 2010;27:90–102. 658. Nicholson DH, Norton EWD. Diffuse infiltrating retinoblastoma. Trans
632. Buetow P, Smirniotopoulos J, Done S. Congenital brain tumors: a review Am Ophthalmol Soc 1980;88:265–281.
of 45 cases. Am J Neuroradiol 1990;11:793–799. 659. Schofield PB. Diffuse infiltrating retinoblastoma. Br J Ophthalmol 1960;44:
633. Nakayama K, Nakamura Y. Localization of congenital glioblastomas in 35–39.
the Japanese: a case report and review of the literature. Childs Nerv Syst 660. Shields CL, Ghassemi F, Tuncer S, Thangappan A, Shields JA. Clinical
2002;18:149–152. spectrum of diffuse infiltrating retinoblastoma in 34 consecutive eyes.
634. Shamji MF, Vassilyadi M, Lam C, Montes JL, Farmer JP. Congenital tumors Ophthalmology 2008;115:2253–2258.
of the central nervous system: the MCH experience. Pediatr Neurosurg 661. Hedges TR, Pozzi-Mucelli R, Char DH, Newton TH. CT demonstration of
2009;45:368–374. ocular calcifications: correlations with clinical and pathological findings.
635. Isaacs H. Fetal brain tumors: a review of 154 cases. Am J Perinatol Neuroradiology 1982;23:15–21.
2009;26:453–466. 662. Brisse HJ, Guesmi M, Aerts I, et al. Relevance of CT and MRI in retinoblas-
636. Duffner P, Horowitz M, Krischer J, et al. The treatment of malignant brain toma for the diagnosis of postlaminar invasion with normal-sized optic
tumors in infants and very young children: an update of the Pediatric nerve: a retrospective study of 150 patients with histological comparison.
Oncology Group experience. Neuro-oncol 1999;1:152–161. Pediatr Radiol 2007;37:649–656.
637. Rilliet B, Vernet O. Gliomas in children: a review. Childs Nerv Syst 663. Bagley LJ, Hurst RW, Zimmerman RA, Shields JA, Shields CL, De Potter
2000;16:735–741. P. Imaging in the trilateral retinoblastoma syndrome. Neuroradiology
638. Rivera-Luna R, Medina-Sanson A, Leal-Leal C, et al. Brain tumors in 1996;38:166–170.
children under 1 year of age: emphasis on the relationship of prognostic 664. Saket RR, Mafee MF. Anterior-segment retinoblastoma mimicking
factors. Childs Nerv Syst 2003;19:311–314. pseudoinflammatory angle-closure glaucoma: review of the literature
639. Lafay-Cousin L, Strother D. Current treatment approaches for infants with and the important role of imaging. Am J Neuroradiol 2009;30:1607–
malignant central nervous system tumors. Oncologist 2009;14:433–444. 1609.
640. Pettorini BL, Park Y-S, Caldarelli M, Massimi L, Tamburrini G, Di Rocco 665. O’Brien JM. Retinoblastoma: clinical presentation and the role of neu-
C. Radiation-induced brain tumours after central nervous system irradia- roimaging. Am J Neuroradiol 2001;22:426–428.
tion in childhood: a review. Childs Nerv Syst 2008;24:793–805. 666. Coats G. Forms of retinal disease with massive exudation. R Lond Oph-
641. Young JL, Smith MA, Roffers SD, Liff JM, Bunin GR. Retinoblastoma. thalmol Hosp Res 1908;17:440–525.
In: Reis LAG, Smith MA, Gurney JG, eds. Cancer Incidence and Survival 667. Chang M, McLean IW, Merritt JC. Coats disease: a study of 62 histologi-
Among Children and Adolescents: United States SEER Program 1975–1995. cally confirmed cases. J Pediatr Ophthalmol Strabismus 1984;21:163–168.
Bethesda, MD: National Institutes of Health, 1999. 668. Smirniotopoulos JG, Bargallo N, Mafee M. Differential diagnosis of leu-
642. Price HI, Batnitzky S, Danzinger A, Karlin CA, Goldberg L. The neurora- kokoria: radiologic-pathologic correlation. Radiographics 1994;14:1059–
diology of retinoblastoma. RadioGraphics 1982;2:7–23. 1079.
643. Provenzale JM, Weber AL, Klintworth GK, McLendon RE. Radiologic- 669. Sherman JL, McLean IW, Brallier DR. Coats’ Disease: CT-pathologic cor-
pathologic correlation: bilateral retinoblastoma with coexistent pine- relation in two cases. Radiology 1983;146:77–78.
aloblastoma (trilateral retinoblastoma). Am J Neuroradiol 1995;16: 670. Chung EM, Specht CS, Schroeder JW. Pediatric orbit tumors and tumor-
157–165. like lesins: neuroepithelial lesions of the ocular globe and optic nerve.
644. Retinoblastoma TCftNRo. Survival rate and risk factors for patients with Radiographics 2007;27:1159–1186.
retinoblastoma in Japan. Jpn J Ophthalmol 1992;36:121–131. 671. Galluzzi P, Venturi C, Cerase A, et al. Coats disease: smaller volume of the
645. Woo KI, Harbour JW. Review of 676 second primary tumors in patients affected globe. Radiology 2001;221:64–69.
with retinoblastoma: association between age at onset and tumor type. 672. Edwards MG, Pordell GR. Ocular toxocariasis studied by CT scanning.
Arch Ophthalmol 2010;128:865–870. Radiology 1985;157:685–686.
646. Ju D, Moss M, Crikelair G. Effect of radiation on the development of facial 673. Mafee MF. Imaging of the globe. In: Valvassori GE, Mafee MF, Carter Bl,
structures in retinoblastoma cases. Am J Surg 1963;106:807–815. eds. Imaging of Head and Neck. New York: Thieme, 1995:216–246.
647. Guyuron B. The hourglass facial deformity. J Craniomaxillofac Surg 1990;18: 674. Mafee MF, Goldberg MF, Valvassori GE, Capek V. CT in the evaluation of
187–191. patients with persistent hyperplastic primary vitreous (PHPV). Radiology
648. Holladay DA, Holladay A, Montebello JF, Redmond KP. Clinical pre- 1982;145:713–717.
sentation, treatment, and outcome of trilateral retinoblastoma. Cancer 675. Galluzzi P, Hadijstilianou T, Venturi C. Leukocoria: clinical and neurora-
1991;67:710–715. diological findings. Riv Neuroradiol 2004;17:61–78.
649. Paulino A. Trilateral retinoblastoma: is the location of the intracranial 676. Mullaney PB, Jacquemin C, Abboud E, Karcioglu ZA. Tuberous sclerosis in
tumor important? Cancer 1999;86:135–141. infancy. J Pediatr Ophthalmol Strabismus 1997;34:372–375.
650. Jurkiewicz E, Paku&lstrok;a-KoÖ ciesza I, Rutynowska O, Nowak K. Trilat- 677. Shields J, Eagle RC, Shields C, Potter PD. Congenital neoplasms of the
eral retinoblastoma: an institutional experience and review of the litera- nonpigmented ciliary epithelium (medulloepithelioma). Opthalamology
ture. Childs Nerv Syst 2010;26:129–132. 1996;103:1998–2006.
651. De Potter P, Shields CL, Shields JA. Clinical variations of trilateral retino- 678. Graeb DA, Rootman J, Robertson WD, Lapointe JS, Nugent RA, Hay EJ.
blastoma. A report of thirteen cases. J pediatr Ophthalmol Strabismus 1994; Orbital lymphangiomas: clinical, radiologic and pathologic characteris-
31:26–31. tics. Radiology 1990;175:417–421.
652. Finelli SA, Shurin SB, Bardenstein DS. Trilateral retinoblastoma: two vari- 679. Hopper KD, Sherman JL, Boal DK, Eggli KD. CT and MR imaging of the
ations. Am J Neuroradiol 1995;16:166–170. pediatric orbit. Radiographics 1992;12:485–503.
653. Skulski M, Egelhoff JC, Kollias SS, Mazewski C, Ball WS, Jr. Trilateral 680. Lallemand DP, Brasch RC, Char DH, Norman D. Orbital tumors in chil-
retinoblastoma with suprasellar involvement. Neuroradiology 1997;39: dren. Radiology 1984;151:85–88.
41–43. 681. Nugent RA, Lapointe JS, Rootman J, Robertson WD, Graeb DA. Orbital
654. Provenzale JM, Gururangan S, Klintworth G. Trilateral retinoblastoma: dermoids: features on CT. Radiology 1987;165:475–478.
clinical and radiologic progression. Am J Roentgenol 2004;183:505–511. 682. Simmons JD, Lamasters D, Char DH. CT of ocular colobomas. Am J
655. Kingston JE, Plowman PN, Hungerford JL. Ectopic intracranial retino- Roentgenol 1983;141:1223–1226.
blastoma of childhood. Br J Ophthalmol 1985;69:242–248. 683. Rootman J. Diseases of the Orbit. New York: Lippincott, 1988.

Barkovich_Chap07.indd 805 5/6/2011 9:18:14 PM


806 Pediatric Neuroimaging

684. Bilaniuk LT, Zimmerman RA, Newton TH. Magnetic resonance imaging: 711. Rickles NH, Little JW. The histogenesis of the branchial cyst. Am J Pathol
orbital pathology. In: Newton TH, Bilaniuk LT, eds. Radiology of the Eye 1967;50:765–773.
and Orbit, vol 4. Kentfield, CA: Clavedel, 1990:5.1–5.84. 712. Shin J, Lee H, Kim S, et al. Parapharyngeal second branchial cyst manifesting
685. Enjolras O. Classification and management of the various superficial vas- as cranial nerve palsies: MR findings. Am J Neuroradiol 2001;22:510–512.
cular anomalies: hamangiomas and vascular malformations. J Dermatol 713. Miller MB, Rao VM, Tom BM. Cystic masses of the head and neck: pitfalls
1997;24:701–710. in CT and MR interpretation. Am J Roentgenol 1992;159:601–607.
686. Bilaniuk LT. Vascular lesions of the orbit in children. Neuroimaging Clin N 714. Harnsberger HR, Mancuso AA, Muraki AS, et al. Branchial cleft anomalies
Am 2005;15:107–120. and their mimics: CT evaluation. Radiology 1984;152:739–748.
687. Char DH, Unsöld R, Sobel DF, Salvolini U, Newton TH. Computed 715. Benson MT, Dalen K, Mancuso AA. Congenital anomalies of the bran-
tomography: ocular and orbital pathology. In: Newton TH, Bilaniuk LT, chial apparatus: embryology and pathologic anatomy. Radiographics
eds. Radiology of the Eye and Orbit, vol 4. Kentfield, CA: Clavedel, 1990: 1992;12:943–957.
9.1–9.64. 716. Ahuja A, King A, Metreweli C. Second branchial cleft cysts: variability of
688. Wright JE, Sullivan TJ, Garner A, Wulc AE, Moseley IF. Orbital venous sonographic appearances in adult cases. Am J Neuroradiol 2000;21:315–319.
anomalies. Ophthalmology 1997;104:905–913. 717. Robson C, Hazra R, Barnes P, Robertson R, Jones D, Husson R. Nontuber-
689. Mavrikakis I, Heran MKS, White V, Rootman J. The role of thrombosis as culous mycobacterial infection of the head and neck in immunocompe-
a mechanism of exacerbation in venous and combined venous lymphatic tent children: CT and MR findings. Am J Neuroradiol 1999;20:1829–1835.
vascular malformations of the orbit. Ophthalmology 2009;116:1216– 718. Guba AM, Adam AE, Jaques DA, et al. Cervical presentation of thymic
1224. cysts. Am J Surg 1978;136:430–436.
690. Bisdorff A, Mulliken JB, Carrico J, Robertson RL, Burrows P. Intracranial 719. Harnsberger HR. Handbook of Head and Neck Imaging, 2nd ed. St. Louis:
vascular anomalies in patients with perorbital lymphatic and lymphati- Mosby, 1995.
covenous malformations. Am J Neuroradiol 2007;28:335–341. 720. Konez O, Burrows P. Magnetic resonance of vascular anomalies. Magn
691. Katz SE, Rootman J, Vangveeravong S, Graeb D. Combined venous lym- Reson Imaging Clin N Am 2002;10:363–388.
phatic malformations of the orbit (so-called lymphangiomas). Associa- 721. Goyal M, Causer P, Armstrong D. Venous vascular malformations in pedi-
tion with noncontiguous intracranial vascular anomalies. Ophthalmology atric patients: comparison of results of alcohol sclerotherapy with pro-
1998;105:176–184. posed MR imaging classification. Radiology 2002;223:639–644.
692. Bilaniuk LT. Orbital vascular lesions: role of imaging. Radiol Clin N Am 722. Mulliken JB, Enjolras O. Congenital hemangiomas and infantile heman-
1999;37:169–183. gioma: missing links. J Am Acad Dermatol 2004;50:875.
693. Greene AK, Burrows PE, Smith L, Mulliken JB. Periorbital lymphatic mal- 723. North PE, Waner M, James CA, Mizeracki A, Frieden IJ, Mihm MC, Jr.
formation: clinical course and management in 42 patients. Plast Reconstr Congenital nonprogressive hemangioma: a distinct clinicopathologic
Surg 2005;115:22–30. entity unlike infantile hemangioma. Arch Dermatol 2001;137:1607–1620.
694. Bilaniuk L. Vascular lesions of the orbit in children. Neuroimaging Clin N 724. Boon LM, Enroljas O, Mulliken JB. Congenital hemangioma: evidence of
Am 2005;15:107–120. accelerated involution. J Pediatr 1996;128:329–335.
695. Shields JA, Shields CL, Scartozzi R. Survey of 1264 patients with orbital 725. Pascual-Castroviejo I, Viaño J, Moreno F, et al. Hemangiomas of the head,
tumors and simulating lesions: The 2002 Montgomery Lecture, part 1. neck, and chest with associated vascular and brain anomalies: a complex
Ophthalmology 2004;111:997–1008. neurocutaneous syndrome. Am J Neuroradiol 1996;17:461–471.
696. Mafee M, Jampil LM, Langer BG, Tso M. CT of optic nerve colobomas, 726. Frieden IJ, Reese V, Cohen D. PHACE syndrome. The association of poste-
morning glory anomalies, and colobomatous cyst. Radiol Clin North Am rior fossa brain malformations, hemangiomas, arterial anomalies, coarc-
1987;25:693–699. tation of the aorta and cardiac defects, and eye anomalies. Arch Dermatol
697. Bilaniuk LT, Farber M. Imaging of developmental anomalies of the eye 1996;132:307–311.
and orbit. Am J Neuroradiol 1992;13:793–803. 727. Metry D, Dowd C, Barkovich A, Frieden I. The many faces of PHACE
698. Jakobiec FA. Granulocytic sarcoma. Am J Neuroradiol 1991;12:263–264. syndrome. J Pediatr 2001;139:117–123.
699. Banna M, Aur R, Akkad S. Orbital granulocytic sarcoma. Am J Neuroradiol 728. Holt G. von Recklinghausen’s neurofibromatosis. Otolaryngol Clin North
1991;12:255–258. Am 1987;20:179–193.
700. Jacquemin C, Bosley T, Liu D, Svedberg G, Buhaliqa A. Reassessment of 729. Suh J, Abenoza P, Galloway H, Everson L, Griffiths H. Peripheral (extracra-
sphenoid dysplasia associated with neurofibromatosis type I. Am J Neuro- nial) nerve tumors: correlation of MR imaging and histologic findings.
radiol 2002;23:644–648. Radiology 1992;183:341–346.
701. Vazquez E, Enriquez G, Castellote A, et al. US, CT, and MR imaging of 730. Halliday A, Sobel R, Martuza R. Benign spinal nerve sheath tumors: their
neck lesions in children. Radiographics 1995;15:105–122. occurrence sporadically and in neurofibromatosis types 1 and 2. J Neuro-
702. Kamel HAM, Brennan PR, Farrell MA. Cervical epidural lipoblastoma- surg 1991;74:248–253.
tosis: changing MR appearance after chemotherapy. Am J Neuroradiol 731. Burk Jr. D, Brunberg J, Kanal E, Latchaw R, Wolf G. Spinal and paraspi-
1999;20:386–389. nal neurofibromatosis: surface coil MR imaging at 1.5 T1. Radiology
703. de Oliveira Ribeiro AC, Joshi VM, Funkhouser WK, Mukherji SK. Inflam- 1987;162:797–801.
matory myofibroblastic tumor involving the pterygopalatine fossa. Am J 732. Bhargava R, Parham D, Lasater O, Chari R, Chen G, Fletcher B. MR imag-
Neuroradiol 2001;22:518–520. ing differentiation of benign and malignant peripheral nerve sheath
704. Albright J, Pransky S. Nontuberculous mycobacterial infections of the tumors: use of the target sign. Pediatr Radiol 1997;27:124–129.
head and neck. Pediatr Clin North Am 2003;50:503–514. 733. Canale D, Bebin J. Von Recklinghausen disease of the nervous system. In:
705. Thomas JR. Thyroglossal-duct cysts. Ear Nose Throat J 1979;58:21–27. Vinken P, Bruyn G, eds. Handbook of Clinical Neurology: The Phakoma-
706. Solomon JR, Rangecroft L. Thyroglossal duct lesions in childhood. toses. Amsterdam: North Holland, 1972:132–162.
J Pediatr Surg 1984;19:555–561. 734. Smirniotopoulos J, Murphy F. The phakomatoses. Am J Neuroradiol
707. Pounds LA. Neck masses of congenital origin. Pediatr Clin North Am 1992;13:725–746.
1981;28:841–858. 735. Carr M, Thorner P, Phillips J. Congenital teratomas of the head and neck.
708. Vogl T, Steger W, Ihrler S, Ferrera P, Grevers G. Cystic masses in the floor J Otolaryngol 1997;26:246–252.
of the mouth: value of MR imaging in planning surgery. Am J Roentgenol 736. Azizkhan R, Haase G, Applebaum H, et al. Diagnosis, management, and
1993;161:183–186. outcome of cervicofacial teratomas in neonates: a Childrens Cancer Group
709. Koch B. Cystic malformations of the neck in children. Pediatr Radiol study. J Pediatr Surg 1995;30:312–316.
2005;35:463–477. 737. Elmasalme F, Giacomantonio M, Clarke K, Othman E, Matbouli S. Con-
710. Telander RL, Deane SA. Thyroglossal and branchial cleft cyst and sinuses. genital cervical teratoma in neonates. Case report and review. Eur J Pediatr
Surg Clin North Am 1977;57:779–791. Surg 2000;10:252–257.

Barkovich_Chap07.indd 806 5/6/2011 9:18:14 PM


Chapter 7 • Intracranial, Orbital, and Neck Masses of Childhood 807

738. Bagla S, Tunkel D, Kraut M. Nontuberculous mycobacterial lymphadenitis 753. Shields C, Shields J, Honavar SG, Demirci H. Primary ophthalmic rhab-
of the head and neck: radiologic observations and clinical context. Pediatr domyosarcoma in 33 patients. Trans Am Ophthalmol Soc 2001;99:133–142.
Radiol 2003;33:402–406. 754. Conneely MF, Mafee MF. Orbital rhabdomyosarcoma and simulating
739. Tom LW, Rossiter JL, Sutton LN, Davidson RS. Torticollis in Children. lesions. Neuroimaging Clin N Am 2005;15:121–136.
Otolaryngol Head Neck Surg 1991;105:1–9. 755. Feldman BA. Rhabdomyosarcoma of the head and neck. Laryngoscope
740. MacDonald D. Sternomastoid tumor and muscular tortocollis. J Bone 1982;92:424–440.
Joint Surg 1969;51B:432–437. 756. Ruymann R. Rhabdomyosarcomas in children and adolescents. Hematol
741. Crawford SC, Harnsberger HR, Johnson L, et al. Fibromatosis colli of Oncol Clin North Am 1987;1:622–654.
infancy: CT and sonographic findings. Am J Roentgenol 1988;151:1183– 757. Hagiwara A, Inoue Y, Nakayama T, et al. The “botryoid sign”: a character-
1186. istic feature of rhabodmyosarcomas in the head and neck. Neuroradiology
742. Som PH, Brandwein M. Rhabdomyosarcomas. In: Som PM, Curtin HD, 2001;43:331–335.
eds. Head and Neck Imaging, 3rd ed. St. Louis: Mosby, 1996:219–220. 758. Yousem DM, Lexa FJ, Bilaniuk LT, Zimmerman RA. Rhabdomyosarcomas
743. Ginsberg LE. Neoplastic diseases affecting the central skull base: CT and in the head and neck. Radiology 1990;177:683–686.
MR imaging. Am J Roentgenol 1992;159:581–589. 759. Ries LAG, Smith MA, Gurney JG, et al. Cancer Incidence and Survival
744. Latack JT, Hutchinson RJ, Heyn RM. Imaging of rhabdomyosarcomas of among Children and Adolescents: United States SEER Program 1975–1995.
the head and neck. Am J Neuroradiol 1987;8:353–359. Bethesda: National Cancer Institute, National Institutes of Health, 1999.
745. Scotti G, Harwood-Nash DC. CT of rhabdomyosarcomas of the skull base 760. Niedobitek G. Epstein-Barr virus infection in the pathogenesis of nasopha-
in children. J Comput Asst Tomogr 1982;6:33–39. ryngeal carcinoma. Mol Pathol 2000;53:248–254.
746. Robson C. Imaging of head and neck neoplasms in children. Pediatr Radiol 761. Ayan I, Kaytan E, Ayan N. Childhood nasopharyngeal carconoma: from
2010;40:499–509. biology to treatment. Lancet Oncol 2003;4:13–21.
747. Davicioni E, Anderson MJ, Finckenstein FG, et al. Molecular classifica- 762. Stambuk HE, Patel SG, Mosier KM, Wolden SL, Holodny AI. Nasopharyn-
tion of rhabdomyosarcoma–genotypic and phenotypic determinants of geal carcinoma: recognizing the radiographic features in children. Am J
diagnosis: a report from the Children’s Oncology Group. Am J Pathol Neuroradiol 2005;26:1575–1579.
2009;174:550–564. 763. Hasso AN, Vignaud J. Pathology of the paranasal sinuses, nasal cavity and
748. Coffin C, Dehner L. The soft tissues. In: Stocker J, Dehner L, eds. Pediatr facial bones. In: Newton TH, Hasso AN, Dillon WP, eds. Computed Tomogra-
Pathol Philadelphia: Lippincott, 1992:1091–1032. phy of the Head and Neck, vol 3. San Anselmo, CA: Calvedel, 1988:7.1–7.31.
749. Konrad E, Hubner G. Rhabdomyoma of the eyebrow region: a light- and 764. Lloyd GAS, Phelps PD. Juvenile angiofibroma: imaging by MR, CT and
electron microscopic study of a recurrent rhabdomyoma of fetal type. conventional techniques. Clin Otolaryngol 1986;11:247–259.
Graefes Arch Clin Exp Ophthalmol 1983;220:187–192. 765. Ponti G, Losi L, Pellacani G, et al. Wnt pathway, angiogenetic and hormonal
750. Myung J, Kim I, Chun J, et al. Rhabdomyoma of the orbit: a case report. markers in sporadic and familial adenomatous polyposis-associated juve-
Pediatr Radiol 2002;32:589–592. nile nasopharyngeal angiofibromas (JNA). Appl Immunohistochem Mol
751. Hatsukawa Y, Furukawa A, Kawamura H, Sugimoto S, Mushiake S, Morphol 2008;16:173–178.
Morimoto K. Rhabdomyoma of the orbit in a child. Am J Ophthalmol 766. Mirich DR, Blaser SI, Harwood-Nash DC, Armstrong DC, Becker LE, Pos-
1997;123:142–144. nick JC. Melanotic neuroectodermal tumor of infancy: clinical, radiologic,
752. Raney RB, Jr, Donaldson MH, Sutow WW, Lindberg RD, Maurer HM, Tefft and pathologic findings in five cases. Am J Neuroradiol 1991;12:689–697.
M. Special considerations related to primary site in rhabdomyosarcoma: 767. Pierre-Kahn A, Cinalli G, Lellouch-Tubiana A, et al. Melanotic neuroec-
experience of the Intergroup Rhavdomyosarcoma Study, 1972–1976. todermal tumor of the skull and meninges in infancy. Pediatr Neurosurg
NCI Monogr 1981;56:69–74. 1992;18:6–15.

Barkovich_Chap07.indd 807 5/6/2011 9:18:14 PM


CHAPTER

Hydrocephalus
8 A. RAYBAUD AND A. JAMES BARKOVICH

Embryology and physiology of CSF dynamics Specific categories of hydrocephalus


Choroid plexuses and CSF formation Hydrocephalus resulting from excessive formation of CSF
CSF circulation (choroid plexus papillomas)
CSF absorption Hydrocephalus secondary to intraventricular obstruction of
Mechanisms of hydrocephalus Hydrocephalus secondary to extraventricular obstruction of CSF
Classical model: obstructive hydrocephalus Benign enlargement of the subarachnoid spaces in infants
Greitz model: communicating hydrocephalus Definition
Classification of hydrocephalus Clinical manifestations
Obstructive hydrocephalus Imaging
Communicating hydrocephalus Imaging of treated hydrocephalus and resulting
Clinical aspects of hydrocephalus complications
Fetal hydrocephalus Radiologic assessment of third ventriculostomies
Postnatal hydrocephalus Shunt malfunctions
Radiologic diagnosis of hydrocephalus Shunt infections
The tools Subdural hematoma formation
Fetal diagnosis of hydrocephalus Slit ventricle syndrome
Postnatal diagnosis of hydrocephalus Imaging of intracranial hypotension
Other complications

ydrocephalus may be defined morphologically as a process in tations of hydrocephalus vary considerably. In this chapter, we discuss
H which the cerebrospinal fluid (CSF) compartment is actively
enlarged at the expense of the brain tissue. It is a common
many of these processes. In addition, we show that the degree of dila-
tation of the CSF pathways and the amount of damage to the brain
disorder in all age groups, especially so in children. The basic issue of depend upon the cause of hydrocephalus, its severity, and the age of the
hydrocephalus is relatively easy to understand. The ventricles progres- patient at the time when the hydrocephalus develops.
sively increase in size in conjunction with increasing head size and
progressive compression and, eventually, loss of brain tissue. Some
CSF must be diverted out of the head in order to preserve the brain.
Establishing the diagnosis also is relatively simple in most cases such EMBRYOLOGY AND PHYSIOLOGY
as increased ventricular size, effacement of the subarachnoid spaces,
macrocephaly, and typically an obstruction somewhere along the CSF
OF CSF DYNAMICS
pathway (e.g., from a posterior fossa tumor or aqueductal stenosis). Closure of the neural tube in man occurs by about 28 postconceptional
But some aspects of hydrocephalus are not so easy to understand. Is days (see Chapters 5 and 9). Certain portions of the central lumen of
the ventriculomegaly due to increased intraventricular pressure? If so, the neural tube then constrict, while others expand to form the basic
why do the ventricles enlarge in communicating hydrocephalus when pattern of the ventricular system such as enlargement of prosenceph-
the “block” to CSF flow is in the subarachnoid spaces? Brain tissue is alic (forebrain), mesencephalic (midbrain), and rhombencephalic
mainly water and, therefore, is not compressible other than some space (hindbrain) vesicles. Some midline dorsal meningeal mesenchyme
provided by the compression of the vascular bed (<4% of the brain tis- invaginates into the lumen of the fourth ventricle during the second
sue) and effacement of the pericerebral spaces. In chronic aqueductal gestational month followed by similar invaginations into the lateral
stenosis, it may take months or years for the ventricular size to increase. and third ventricles; these invaginations form the choroid plexuses
Therefore, the daily CSF production (estimated at 500 mL) must be of the ventricular system and secrete the early CSF (1,2). Initially, the
somehow resorbed by cerebral parenchyma; why do we see an increase plexuses are large relative to the size of the lateral ventricles, filling
in ventricular size? The best answer (for now) is that the morphological approximately 75% of the ventricular lumen during the third month of
pattern that we call hydrocephalus is probably the end result of many gestation. The size of the choroid plexus relative to the lateral ventricle
different processes; accordingly, the imaging and pathologic manifes- gradually diminishes as the brain and ventricular system grow.

808

Barkovich_Chap08.indd 808 5/6/2011 10:25:59 PM


Chapter 8 • Hydrocephalus 809

Choroid Plexuses and CSF Formation through the third ventricle, into the Sylvian aqueduct and the fourth
ventricle. From the fourth ventricle, CSF courses through the foramina
The choroid plexuses have several functions. They secrete multiple of Magendie and Luschka into the cisterna magna and the basilar cis-
proteins that are postulated to stimulate and modulate the growth of terns, respectively. It is estimated that 80% of flowing CSF then enters
progenitor cells in the ventricular zone of the germinal matrices dur- the cisternal system, flowing into the suprasellar cistern and cistern of
ing development (see Chapter 5) (3). They act on diverse chemosensi- the lamina terminalis, the ambient and superior cerebellar cisterns, and
tive areas of the ependyma throughout life (4); these small areas in the eventually flowing over the cerebral convexities. The remaining 20%
walls of the third and fourth ventricle are called the circumventricular of CSF undergoing bulk flow initially enters the spinal subarachnoid
organs; they contain specialized ependymal cells called tanycytes that space but is eventually recirculated into the cerebral subarachnoid
form exchange interfaces allowing the brain, mostly the hypothalamus, space (21), although some (10%–15%) is absorbed in the spine at the
to react in response to changing CSF composition. In certain hydro- level of the nerve root sheaths (22–24).
cephalic animal models, the progenitor cells in the ventricular walls are Although the concept of bulk flow in the ventricular system is
less sensitive to the proliferative effects of certain growth factors than generally accepted and serves as an adequate model for most cases of
controls. Thus, fetal hydrocephalus may adversely affect brain develop- hydrocephalus, it fails to explain some cases of progressive ventriculo-
ment (3). megaly (25). It has been demonstrated that the CSF “flow” through the
The extracerebral CSF spaces appear near the end of the embryonic subarachnoid spaces is quite slow (.35 mL/min) and that subarachnoid
period, resulting from the expansion of the meningeal extracellular CSF motion is nearly entirely pulsatile; that is, the water molecules
space as the gelatinous primitive mesenchyme that surrounds the brain pulsate back and forth, driven by expansion of the intracranial blood
(meninx primitiva) undergoes a systematic degeneration to become vessels as they fill with blood after each cardiac systole, but very few
the fluid-filled leptomeninges (5,6). The communication between ven- CSF water molecules actually follow the rules of bulk flow and actually
tricular and subarachnoid CSF results from thinning and opening of circulate through the subarachnoid space from the outlet foramina of
the inferior midline portion of the fourth ventricular roof to create the the fourth ventricle to the arachnoid granulations (26). Thus, it appears
foramen of Magendie during the 11th or 12th gestational week, fol- that the circulation of CSF results from the combination of two processes.
lowed by opening of the lateral foramina of Luschka (7,8). The function The secretion-absorption process (bulk flow) is slow and passive,
of CSF in the embryo is not completely understood. However, there is particularly in the subarachnoid spaces, and may be compared to the
essentially no intrinsic brain vasculature when the plexuses develop lymphatic circulation. The separate, pulsatile process is fast (~60–80
(9,10), and it is postulated that important chemicals enter the CSF from pulsations/min at rest) and is driven by pressure waves produced by
the blood vessels of the choroid plexuses and circumventricular organs; enlargement of the intracranial arteries as the blood flows into them
these chemicals may be vital in controlling the early development of the after each cardiac systole. These pulsations continuously mix the CSF.
nervous system along the entire length of the neural tube (11). As the cranial cavity is rigid and the brain tissue not compressible, the
In the adult, CSF is secreted at a rate of approximately 0.3 to pressure waves need to be dampened: the force transmitted through
0.4 mL/min, resulting in a daily secretion of approximately 500 mL the subarachnoid space via CSF is dampened by the expansion of the
(12). The choroid plexus produces an estimated 60% to 90% of CSF, highly elastic spinal thecal sac, while the force transmitted through the
with the other 10% to 40% most likely coming from the parenchyma vascular system is dampened by expansion of the venous sinuses. This
of the brain and spinal cord (13,14). CSF secretion by the choroid dual dampening process constitutes the compliance of the system. The
plexuses uses ATP hydrolysis to generate unidirectional flux of sodium, loss of thecal elasticity (or restriction of the access to the spinal suba-
chloride, and bicarbonate ions across the plexus epithelium, driving the rachnoid space) results in less attenuation of the CSF pressure wave;
movement of water by osmosis (4). The total volume of CSF was clas- this force then is fully exerted upon the brain tissue. If the venous pres-
sically assumed to be 40 to 60 mL in infants, 60 to 100 mL in children, sure is increased, the dampening of the force in the vascular system
and approximately 150 mL in adults (12). However, studies using volu- is lost and the force of the vascular pressure wave is exerted upon the
metric magnetic resonance imaging (MRI) to quantify the CSF volume parenchyma. In both instances, the pressure waves cause a progressive
have found higher values, with a volume of ∼150 mL in the intracranial loss of brain tissue. Another factor to consider is the anatomic organi-
subarachnoid spaces and 100 to 120 mL in the spinal subarachnoid zation: the arteries surround the brain, which surrounds the ventricles.
spaces (15). In addition, the interstitial fluid of the central nervous sys- Therefore, the force of each arterial pulsation is transmitted centrip-
tem is estimated to contain 100 to 300 mL of CSF (4). CSF and extra- etally via the parenchyma to the ventricle, pushing CSF toward the
cellular fluid in the brain and spinal cord are in continuity with each cisterna magna; it is only partly counter-balanced by the small pulsa-
other, and exchanges occur constantly across the pial membrane, the tions of the intraventricular choroid plexuses. Finally, the transmission
Virchow-Robin perivascular spaces, and the ependyma (16). of the pressure wave after each pulsation is extremely rapid across the
noncompressible CSF but is slower through the more elastic vascular
bed; as a consequence, the systolic pressure wave results in constriction
CSF Circulation
of the outlets of the bridging veins before pushing blood into the capil-
Classic teaching suggests that CSF flows from the cerebral ventricles lary bed. As a result, the vascular bed expands (through the so-called
through the ventricular foramina and into the subarachnoid spaces, “bagpipe” effect), increasing the perfusion pressure and, consequently,
ultimately being absorbed into the venous system (13,14); this cerebral perfusion (25).
movement of water molecules through the ventricles and cisterns is In summary, pressure waves from arterial pulsations are important
referred to as bulk flow; it results from a hydrostatic pressure gradi- for normal CSF flow and cerebral perfusion; however, these pressure
ent between the site of its formation (arterial) and its site of absorp- waves need to be dampened for the system to function properly. The
tion (venous) (17,18). Another mechanism, the coordinated beating of compliance of the system depends on the venous pressure and on the
cilia in ependymal cells, creates a current of CSF along the walls of the elasticity of the dura. If the venous pressure is elevated, or if the pressure
lateral ventricles that likely contributes to bulk flow (19,20). CSF leaves waves do not expand the elastic dura, the force of a pressure wave is not
the lateral ventricles via the foramina of Monro and then circulates dampened and it pulsates against the parenchyma at full strength. This

Barkovich_Chap08.indd 809 5/6/2011 10:25:59 PM


810 Pediatric Neuroimaging

causes compression of the cerebral tissue, alterations of axonal function


and of cerebral blood flow (CBF), and progressive brain injury.
MECHANISMS OF HYDROCEPHALUS
Neonates and young infants have open fontanelles and sutures, Classical Model: Obstructive Hydrocephalus
so both the cranial and the spinal dura contribute to this elasticity,
whereas in the older child and adult, the cranial dura is attached to a In this concept, hydrocephalus results from an imbalance between CSF
rigid calvarium, so only the spinal dura (long and surrounded by fat production and absorption. The rate of CSF production tends to be
and veins) contributes to elasticity. Thus, in older children, obstruction fairly constant within the brain and, with the exception of hydrocepha-
of the foramen magnum (which reduces the total elasticity of the CSF- lus secondary to choroid plexus papillomas, overproduction of CSF is
containing spaces) and any other process that reduces the dampening not a cause of hydrocephalus. (Some authors suggest that diffuse villous
of the pressure wave (arachnoid fibrosis and loculation, spinal cord hyperplasia is a very rare cause of hydrocephalus secondary to over-
tumor, suprasellar cyst, high venous pressure) favors the development production (43–45); others claim that these cases represent multifocal
of hydrocephalus by reducing the dampening efficacy of the system. choroid plexus papillomas (46); if choroid plexus hyperplasia exists, it
Any factor that increases the pressure wave (any pulsating mass in the is very rare and will not be discussed further in this text.) Therefore,
CSF spaces) may also compromise the equilibrium if the compliance hydrocephalus is almost always the result of impaired absorption of
reaches its limit, as was demonstrated experimentally (27). CSF. The diminished absorption may result from a blockage of CSF
flow within the ventricular system, the cisterna magna, the basilar cis-
terns, or the cerebral convexities. Alternatively, diminished absorption
CSF Absorption may result from blockage of the arachnoid villi or the lymphatic chan-
According to the most generally accepted model, CSF absorption nels associated with the cranial nerves, spinal nerves, and adventitia of
occurs mainly through the arachnoid villi, evaginations of the suba- the cerebral vessels. As the intracranial pressure increases, CSF may be
rachnoid space into the lumen of the dural and venous sinuses. This absorbed through the arachnoid membrane, the stroma of the choroid
model proposes that the villi behave as one-way valves that have an plexus, or may pass through the extracellular space of the cortical man-
opening pressure of between 20 and 50 mm of water; drainage is deter- tle to reach the medullary veins or the brain surface (transependymal
mined by hydrostatic pressure differences between CSF and the venous absorption of CSF) (13,36,47). Because these new absorption path-
sinuses (13,14,17). However, several authors have suggested that ways open at increased pressure, a new equilibrium can be established
approximately half of the CSF may drain via other routes. As the CSF between CSF production and absorption at the higher intracranial
space and extracellular spaces are in continuity via the ependymal and pressure. This situation is referred to as compensated hydrocephalus.
pial surfaces and the perivascular spaces, a significant part of the CSF Stenotic or obstructive changes in small blood vessels in the com-
is presumed to be absorbed by the intracerebral veins, possibly even pressed periventricular white matter of hydrocephalic patients cause
as the primary mode of absorption (28–31). The lymphatic system neuronal and astrocytic swelling in the deep gray matter and spongy,
probably plays a significant role, particularly through the cribriform atrophic changes in the white matter of the cerebral hemisphere
plate into the lymphatic vessels located in the nasal submucosa (32– (46,48–51). The blood flow and cerebral metabolic rate, as measured
34), in the perineural spaces (13,35–37), and through the optic nerves by uptake of 18F-fluorodeoxyglucose on positron emission tomogra-
into the orbital lymphatics (38). The anatomical organization of the phy, are reduced in the periventricular white matter in hydrocephalic
dural clefts described where cortical veins join the sagittal and cavern- infants (52). Blood flow is particularly affected in the distribution of
ous sinuses, suggests that they could be another pathway of egress of the anterior cerebral arteries, leading to ischemic injury of the basal
CSF into the dural sinuses (39,40). Still others contend that the brain, forebrain and medial cerebral hemispheres (53). Moreover, the cilia
rather than absorbing CSF, acts as a conduit for fluid to move from that normally cover the ependymal surface of the ventricular walls
the ventricles into the subarachnoid spaces or into the prelymphatic disappear in hydrocephalic states (54), ependymal cells in the wall of
channels of the blood vessels (41). CSF may nourish cerebral vessels the ventricle stretch and degenerate (46), the choroid plexuses become
through pathways in the adventitia that may be analogous to systemic fibrotic (48), and the septum pellucidum undergoes fenestration (48).
vasa vasorum. Another possible route of CSF absorption is through
the epithelium of the choroid plexus to the fenestrated capillaries (4)
Greitz Model: Communicating Hydrocephalus
and ultimately to the Galenic venous system. Oi and Di Rocco refer
to these routes as the “minor pathways” of CSF absorption and to the As mentioned above, an interesting new model of CSF dynamics and
classic route via the subarachnoid spaces and arachnoid granulations the development of hydrocephalus has been proposed in a series of
as the “major pathway” (42). Moreover, they and others suggest that articles by Greitz et al. (25,26,30,31,55,56). CSF pulsations have been
CSF absorption in neonates and infants is predominantly via the so-called observed from the early days of neuroradiology (57), and have been
minor pathways and that the major pathway only becomes an important explained by the transmission of pulsations from intracranial arteries.
route of CSF absorption during the middle or end of the first postnatal These arterial pulsations, occurring in a rigid skull that is filled almost
year (33,42). This proposal is supported by the observation that third entirely with incompressible water, transmit blood through capillary
ventriculostomy is less effective in neonates and young infants than beds and toward the venous sinuses while, at the same time, displacing
in older children and adults and becomes progressively more effective some CSF into the spinal subarachnoid spaces, which is encompassed
with maturation (42). Others suggest that the “minor pathways” are by more elastic, distensible dura that expands to absorb and dampen
recruited in neonates and infants only as auxiliary mechanisms when the pressure waves (see Section “CSF Circulation” above). The CSF dis-
intracranial pressures are high (arachnoid villi and granulations are placement is greatest at the craniovertebral junction, where it has been
not formed in neonates and are unlikely to be involved in CSF resorp- measured at 6 mm per stroke in normal individuals, and at 3 mm in
tion) (33,42). It is likely that many or all of the mechanisms described communicating hydrocephalus (15). If the spinal dural sac does not
above function in CSF absorption, some as primary pathways and oth- expand to dampen the CSF pressure wave (because, e.g., the foramen
ers as alternate pathways. It is also likely that all of these pathways are magnum is obstructed or the spinal meninges have lost their elasticity),
involved in the pathophysiology of hydrocephalus when drainage of or if the dampening through the vascular bed is prevented by high
CSF through them becomes impaired. venous pressure, or if the intraventricular choroid plexus pulsations

Barkovich_Chap08.indd 810 5/6/2011 10:25:59 PM


Chapter 8 • Hydrocephalus 811

are increased, the force of the pulsating arteries is exerted upon the by the traditional models. Examples of the utility of these concepts are
brain parenchyma. This model leads to three main conclusions: discussed in the section on “Normal Pressure Hydrocephalus.”
■ Even if nothing opposes the bulk flow of CSF, hydrocephalus may
develop because the craniospinal meningeal compliance is lost CLASSIFICATION OF HYDROCEPHALUS
(communicating hydrocephalus).
Although the pathogenesis of hydrocephalus is complex, it is useful
■ Even in cases with obstruction of the bulk flow pathway (obstructive
from a clinical and therapeutic point of view to differentiate between
hydrocephalus), part of the problem comes from the fact that the
obstructive and communicating hydrocephalus (Table 8-1).
obstruction restricts the compliance of the system by not allowing
the pressure waves to reach the spinal dura.
■ Even with no obstruction in the ventricles, increased pulsation Obstructive Hydrocephalus
waves within the ventricles (e.g., from choroid plexus papilloma or
Hyperacute obstructive hydrocephalus develops in hours from a sud-
choroid plexus hyperplasia) may overcome the compliance of the
den occlusion of the CSF pathways, with rapid increase of the intra-
system and generate hydrocephalus. (Such a situation has been cre-
ventricular pressure. This may occur after severe head trauma, shunt
ated experimentally (27).)
obstruction or, rarely, granulomatous meningitis. The brain paren-
Thus, normal volume and elasticity of the spinal dura are needed for chyma has no time to accommodate the increase in ventricular pressure
proper function of the system in older children and adults. As men- by loss of tissue, so the ventricles are rounded but minimally increased
tioned above, the area of the spinal meninges is small in relation to the in size, making morphological diagnosis difficult. As neither the paren-
volume of the cranium during the first months of life. From the point chyma nor the CSF is instantaneously compressible, the only part of
of view of compliance, however, the increased elasticity of the skull due the brain in which volume is lost is the vascular bed. This hypoperfu-
to open sutures and fontanelles compensates for small size of the spinal sion is worsened by the fact that any expansion of the brain, even slight,
canal. The situation in fetuses is not well understood, as the situation is may compress superficial draining veins against the calvarium, while
compounded by high pressure of the surrounding amniotic fluid and the deep veins are compressed by high intraventricular pressure, result-
by a blood circulatory system that is different from the postnatal one. ing in further increase of parenchymal and intracranial pressure. This
While the bulk flow cannot be detected by noninvasive means, vicious circle leads to absence of perfusion of the brain. Only tapping
MR phase-contrast imaging has the ability to evaluate the direction the ventricles allows relief of the ventricular tamponade.
and velocity of CSF flow. Quantitative data in healthy adult individu- Progressively acute obstructive hydrocephalus typically develops over
als have been measured (25). Assuming a CBF of 60 mL/100 g/min, weeks or months, usually from the growth of an intraventricular tumor.
and a heart rate of 60 beats/min, the volume of the arterial stroke is CSF outflow is progressively impeded, due to impaired bulk flow, so the
15 mL for a 1500 g brain. Of this, 90% flows directly through the vas- ventricles progressively enlarge. If the head enlarges as well, the cere-
cular bed to the veins, and the expansion of the intracranial arteries bral volume may be partly preserved; if not, the ventricular enlarge-
corresponds to 1.5 mL (the sum of the systolic stroke volume of CSF ment will be at least partially due to loss of brain tissue, with the degree
displaced at the foramen magnum—0.8 mL—and of blood displaced of tissue loss depending on the age of the patient and on the speed of
into the venous sinuses—0.7 mL). The systolic expansion of the brain onset of hydrocephalus. The increased ventricular pressure results in
capillaries is minimal (0.03 mL, only 2% of the arterial expansion) but compression of the subependymal veins; this, in turn, prevents the nor-
real; it is transmitted inwards to the ventricles, and in the aqueduct, mal absorption of interstitial fluid from the deep white matter, result-
the displaced volume of CSF is correspondingly 0.03 mL per beat (25). ing in development of periventricular interstitial edema and (often)
Given the small section of the aqueduct, this results in a measurable parenchymal injury, but does not reflect any increased absorption by
CSF systolic velocity peak. In another report, the MR-measured sys- the parenchyma (50). If not treated, progressively acute hydrocephalus
tolic peak aqueductal CSF velocity has been evaluated at 3.4 to 4.1 cm/s will cause brain herniation and/or circulatory arrest.
in normal volunteers, corresponding to a mean CSF flow of 0.02 to Chronic obstructive hydrocephalus can be a result of many causes,
0.03 mL/s (58). Using a similar MR phase-contrast imaging method such as chronic aqueductal stenosis or leptomeningitis. This chronic
in volunteers also, another group could measure the CSF production condition causes only mild clinical signs of increased intracranial
rate and found 0.305 mL/min ± 0.145 (equivalent to about 430 mL/d), pressure (i.e., headaches) and no periventricular interstitial edema,
which is in good agreement with the data obtained with more invasive as the secretion of CSF is presumably compensated by sufficient fluid
methods (58). Using these basic concepts, Greitz et al. have reconsid- absorption via capillaries. However, the ventricles and, typically, the
ered the way of approaching hydrocephalus and have devised a new skull slowly enlarge and the brain slowly loses volume over the years.
model that differs from the traditional one. These concepts may be use- An acute decompensation may occur, after trauma for example. The
ful in understanding cases of hydrocephalus that cannot be explained terms “arrested” and “compensated” hydrocephalus are used when no

TABLE 8-1 Classification of Obstructive Hydrocephalus


Timing Vasc. Bed Ventr. Size Interst. Edema Skull Outcome
Hyper-acute Sudden ↓ = No = Arrest
Progressive Weeks/months = ↑ Yes =/↑ Decompensates
Chronic Years = ↑ No ↑ Slow ↑
Arrested Years = ↑ No ↑/= Stable?

Barkovich_Chap08.indd 811 5/6/2011 10:25:59 PM


812 Pediatric Neuroimaging

progression can be detected clinically or by imaging over long periods and the associated malformation, or whether the hydrocephalus may
of time; this seems to be an uncommon occurence. merely be one component of a malformation syndrome.

Communicating Hydrocephalus Postnatal Hydrocephalus


The term communicating hydrocephalus describes cases in which no Prior to the age of 2 years, hydrocephalus is almost always accompa-
clear obstruction to CSF flow can be demonstrated; no morphologic nied by progressive head enlargement. The Chiari II malformation,
finding explains the development of progressive ventricular dilation with aqueductal stenosis, and aqueductal gliosis account for 80% of hydro-
clinical symptoms of hydrocephalus and good response to CSF diver- cephalus in this age group, and 60% of all hydrocephalus regardless
sion. Commonly assumed to be related to obstruction or fibrosis of the of age (48,71). Other relatively common causes of hydrocephalus in
arachnoid granulations, it is better explained by the Greitz model intro- infancy include intrauterine, perinatal, and neonatal infection, and
duced in Section “Greitz Model: Communicating Hydrocephalus” (25). perinatal/neonatal hemorrhage. Rare causes of hydrocephalus include
As explained earlier, free movement of the CSF is impaired by arachnoid other malformations, congenital midline tumors, choroid plexus pap-
pathology (e.g., fibrosis from a previous history of meningeal infection illomas, and vein of Galen malformations (48).
or bleed, or brain trauma) or impaired elasticity of the spinal dura. As In infants with hydrocephalus, the head tends to grow at an abnor-
a result, the propagation of pulse pressure waves through the CSF is mal rate, producing macrocephaly within the first few months of life.
impaired, and their force is exerted against the brain tissue. It is tempt- The forehead is disproportionately large (frontal bossing), the skull is
ing to extend this concept to other disorders such as segmental restric- thin, the sutures are separated, the anterior fontanelle is tense, and the
tion of the meningeal space (significant spinal degenerative changes in scalp veins are dilated. Ocular disturbances are frequent and include
adults, achondroplasia or mucopolysaccharidoses in children, spinal paralysis of upward gaze, abducens nerve paresis, nystagmus, ptosis, and
cord tumors) or the Chiari I deformity where the obstruction of the diminished pupillary light response. Spasticity of the lower extremities
foramen magnum may be considered a CSF pathway obstruction that is common, resulting from disproportionate stretching and distortion
causes a loss of compliance. This new concept brings into question the of the corticospinal axons that arise from the medial parts (leg area) of
use of the term “normal” pressure hydrocephalus, as many physiologi- the motor cortex. These axons have a longer distance to travel around,
cal studies have shown in adults that the amplitude of the arterial-CSF and are more directly exposed to pressure from, the dilated lateral ven-
pulsation pressure wave (if not the mean pressure) is characteristically tricle than the more lateral corticospinal and corticobulbar axons that
increased (59–62). supply the upper extremities and the face (48,72,73).
Children older than 2 years tend to present with neurologic symp-
toms resulting from increased intracranial pressure or with focal
CLINICAL ASPECTS OF HYDROCEPHALUS deficits referable to the primary lesion; these symptoms occur prior
The most important and most consistent clinical finding in pediatric to significant changes in the head size. The most common causes of
hydrocephalus is an excessive rate of head growth. A large head circum- hydrocephalus in this age group are posterior fossa neoplasms (see
ference in a child is not, in itself, of concern (it actually allows some Chapter 7) and obstruction of the aqueduct. Although each of the
preservation of brain volume). Serial head circumference measure- specific lesions that result in hydrocephalus have some special fea-
ments showing an excessive rate of head growth as compared with nor- tures, certain clinical characteristics are common to all hydrocephalic
mal standards, however, should raise clinical suspicion of developing patients. In most patients, the increased intracranial pressure results
hydrocephalus. Several factors influence the clinical course of hydro- in an early morning headache that improves after being upright for a
cephalus. The most important of these are the age of the patient at the while (allowing CSF to re-equilibrate). Papilledema and strabismus are
onset of hydrocephalus and the duration of the disease (younger age frequent. Pyramidal tract signs are more marked in the lower extremi-
and longer duration imply worse prognoses). The rate of increase of ties, as described above. Hypothalamic-pituitary dysfunction is prob-
intracranial pressure and the presence of associated structural lesions ably caused by compression of the hypothalamus, pituitary stalk, and
are also associated with poor prognosis (63,64). pituitary gland by the enlarged anterior recesses of the third ventricle;
affected patients may present with small stature, obesity, gigantism,
amenorrhea or menstrual irregularities, hypothyroidism, or diabetes
Fetal Hydrocephalus insipidus. Perceptual and motor deficits and visual spatial disorgani-
Ventriculomegaly is a common indication for fetal MRI or level 3 zation result from stretched axons of the parietal and occipital lobes
fetal sonograms (65–67). Fetuses with small heads are unlikely to have around the dilated posterior horns of lateral ventricles (48,72,73). Cog-
hydrocephalus. However, fetal hydrocephalus is not always associated nitive disorders may also result from compression of the hippocampi
with macrocephaly in utero and, thus, normocephalic fetuses may have and stretching of the fornices and hippocampal commissure.
hydrocephalus. Associated malformations of the central nervous system
(CNS) or non-CNS viscera are identified in 41% to 78% of fetuses or RADIOLOGIC DIAGNOSIS
neonates in whom hydrocephalus is detected (68,69). The most com-
mon of these are myelomeningoceles, aqueductal stenosis, and callosal
OF HYDROCEPHALUS
anomalies. Excluding myelomeningoceles, Garel et al. (69) found brain The Tools
abnormalities in 44% of fetuses with ventriculomegaly, with many
causes including callosal anomalies (9.5%), ischemic injury (7%), and Magnetic Resonance Imaging
posterior fossa anomalies (7%). If treated in utero (by cephalocentesis MRI is the best tool to investigate hydrocephalus, its causes, and its
or ventriculo-amniotic shunt), only 38% to 55% of survivors will have consequences. The imaging protocol must be adapted to the patient,
normal cognitive function (68,70) and average survival is a little more but thin section sagittal T1, T2FSE, and FIESTA/CISS sequences show
than 4 years. It is not known whether the associated brain damage is the midline, while coronal T2 best shows the morphology of the lat-
the result of the presence of hydrocephalus in the developing brain, eral ventricles (especially the temporal horns), and axial FLAIR shows
whether a prenatal injury may have caused both the hydrocephalus parenchymal lesions and interstitial edema (Fig. 8-1). Signal voids

Barkovich_Chap08.indd 812 5/6/2011 10:25:59 PM


Chapter 8 • Hydrocephalus 813

FIG. 8-1. MRI for hydrocephalus. Normal images. A. Midline sagittal T1, thin sec-
tion (1.5 mm) image shows the commissures, septum pellucidum and fornix. Note
the anterior recesses of the third ventricle (chiasmatic recess in front of the chiasm
and infundibular recess behind) and posterior recesses (pineal recess below the pineal
gland and suprapineal recess above it). Also well seen are the midbrain with tectal plate
and aqueduct, brainstem, fourth ventricle and vermis, cisterna magna, basilar, interpe-
duncular and suprasellar cisterns. B. Midline sagittal T2, thin (3.0 mm) image shows
the same anatomical features. Note in addition the flow void through the aqueduct.
C. Coronal T2-weighted image through the ventricular bodies and temporal horns and
anterior hippocampi. Note the morphology of the third and lateral ventricles, especially
temporal horns and hippocampal commissure. D. Axial FLAIR image is excellent for
evaluation of the parenchyma, especially the periventricular white matter. E. High defi-
nition CISS/FIESTA (1.0 mm or less) image is optimal for depiction of the third ventri-
cle and its recesses, the aqueduct and fourth ventricle. Note the exquisite demonstration
of the membrane of Liliequist (black arrows) extending from the dorsum sellae to the
mamillary bodies. This sequence is not appropriate for demonstrating the flow voids.

Barkovich_Chap08.indd 813 5/6/2011 10:25:59 PM


814 Pediatric Neuroimaging

due to rapid CSF flow are seen well on T2FSE sequences; these signal (As discussed in Chapter 1, a low dose CT technique, which does not
voids can be useful to demonstrate patency of interventricular connec- allow assessment of subtle parenchymal lesions, is safer for the pedi-
tions (foramina or aqueduct) (Fig. 8-1B) or a third ventriculostomy. atric patient who will likely have many imaging exams during their
Remember, however, that although absence of a signal void suggests lifetime.) “Fast” MR-scanning (using single-shot, ultrafast imaging
foraminal occlusion, foraminal narrowing without occlusion increases sequences such as those used in fetal imaging) has been suggested as a
the velocity and accentuates the signal void. High-definition steady- potentially useful sequence, as it alleviates the need for sedation/anes-
state (CISS/FIESTA) imaging better demonstrates morphology; steno- thesia and the repetitive exposure to significant ionizing radiation (75).
sis of the aqueduct, cysts and abnormal intraventricular or arachnoid Although these heavily weighted T2 sequences are adequate for assess-
membranes are seen much better than on conventional sequences ing the ventricular/cisternal size and morphology, they have poor sen-
(Figs. 8-1E and 8-2A). sitivity and specificity for parenchymal changes. For various reasons
Contrast administration is useful initially to characterize the cause (not easy to implement, limited diagnostic yield, lack of sensitivity/
of hydrocephalus (tumor tissue or infection) but is not useful for fol- specificity, limited access to MR, necessity to reset the magnetic pres-
lowing the hydrocephalus itself. Some have safely used intracisternal sure valve), it has failed to gain wide acceptance.
contrast (74), but it has failed to gain acceptance as the trend in brain Occasionally, hydrocephalus may be caused by, or associated with,
imaging now is toward minimal invasiveness. Magnetic susceptibility a spine lesion (tumor or malformation or syringohydromyelia) rather
T2* imaging may demonstrate blood and blood residue. Other MR than an intracranial one. The protocol for spinal MRI in these cases
sequences are not generally useful in the initial work-up. should include sagittal T2 sequences of the whole spine; if needed, axial
MR is not commonly used for follow-up imaging of hydrocepha- T2, and sagittal T1 and possibly contrast-enhanced T1 sequences may
lus, as the need for sedation in young children and the relatively lim- be acquired though any region of interest. Use of fat suppression allows
ited access to MR have resulted in CT being the usual study of choice. better recognition of soft tissue mass, fluid or blood.

FIG. 8-2. Evaluation of the CSF kinetics. A. Midline sagittal


T2-weighted image. Severe hydrocephalus with a prominent flow
void in the aqueduct. B. Cine phase-contrast imaging, systolic
phase (compare with the flow in the straight sinus). Superior
to inferior flow through the aqueduct, similar to the flow void
in (A), is shown by hyperintense signal. C. Cine phase-contrast
imaging, diastolic phase, shows inferior to superior flow as
hypointense signal.

Barkovich_Chap08.indd 814 5/6/2011 10:26:01 PM


Chapter 8 • Hydrocephalus 815

MR techniques are also able to provide physiologic data on brain individual patients is rarely useful; such measurements are only valid
metabolism (single- or multivoxel 1H MR spectroscopy) and brain when obtained from groups of subjects. Practically speaking, func-
perfusion (by either arterial spin-labeling or dynamic susceptibility- tional methods are not useful for individual diagnoses and will not be
weighted contrast perfusion imaging, see Chapter 1). Qualitative CSF discussed in this chapter.
flow is often demonstrated on routine MR sequences (76–78). On
routine spin-echo MR scans, dynamic CSF flow can be detected by CT Scanning
signal loss in areas of rapid or turbulent flow (76,77). By use of certain Together with MRI, CT is the most commonly used modality to evalu-
techniques, such as gradient echo and FLAIR, rapidly moving CSF can ate hydrocephalus. It is fast and simple and data is easily reformatted in
be made to appear bright while keeping the signal of stationary or multiple planes (Fig. 8-3). Periventricular interstitial edema is readily
slowly flowing CSF dark; the patency of the aqueduct can be assessed apparent on CT, and subarachnoid blood may appear better on CT
in this way (79). Special techniques that take advantage of the relative than on MR. Mass effects responsible for the hydrocephalus are easily
change in phase angle of moving spins can be used to quantify the recognized and contrast enhancement helps in the characterization of
flow of CSF through the foramina of Monro, aqueduct, foramen of the pathology. However, the diagnostic yield and the versatility of CT
Magendie, or brainstem cisterns (Fig. 8-2B and C) (80–82). However are much less that those of MR, and CT can expose children to signifi-
CSF motion is dependent on so many physiological factors—arterial cant doses of ionizing radiation (see Chapter 1). In our practices, initial
stroke (related to blood pressure, perfusion pressure), distal vascu- evaluation of the brain and CT is used for follow-up assessment only,
lar compliance (vascular status, pO2, and pCO2), meningeal compli- with implementation of a low-dose technique that limits evaluation
ance, CSF pressure, individual anatomy, etc.—that quantification in mostly to assessment of ventricular size (see Chapter 1).

FIG. 8-3. 3D-CT imaging. Nice demonstration of the ventricu-


lar and cisternal anatomy in the axial (A), sagittal (B), and coronal
(C) planes. Although not as sensitive and versatile as MR, modern
CT is a fast and efficient diagnostic tool, and it is commonly used
for screening or follow-up. Its main disadvantage is the radia-
tion dose delivered, especially concerning in children. In most
instances, a very low dose CT (see Chapter 1) is used to check
ventricular size.

Barkovich_Chap08.indd 815 5/6/2011 10:26:02 PM


816 Pediatric Neuroimaging

Ultrasonography in children with elevated intracranial pressure. The maximum change


Head ultrasound is the primary modality of choice for the screening of in resistive index was significantly higher in infants who subsequently
infants and fetuses suspected of having hydrocephalus. It is noninva- required shunting than in infants who did not (p = 0.001) (83). Thus,
sive, can be performed at the bedside, and gives a good evaluation of the need for shunt placement may be determined by assessment of
the ventricular size and morphology. It is also sensitive (if not specific) resistive indices with and without fontanelle compression.
to parenchymal abnormalities and, when Doppler is used, can give an
efficient functional assessment of the vasculature. Its use, however, is
Fetal Diagnosis of Hydrocephalus
limited to the first months of life when the fontanelles are open, and
it does not give nearly as comprehensive a presurgical assessment of The prenatal diagnosis of hydrocephalus is currently made almost
hydrocephalus as MRI. exclusively by obstetrical sonography. In fact, the fetal sonogram shows
Monitoring of intracranial pressure is an important factor in ventriculomegaly, not frank hydrocephalus, as many of the radiologic
the differentiation of hydrocephalus from atrophy. New innovative signs of hydrocephalus (see next section) cannot be accurately deter-
approaches with transfontanelle sonography may allow assessment of mined by fetal sonography. The diagnosis of ventriculomegaly is made
intracranial pressure noninvasively. Taylor and Madsen (83) studied the if the fetal sonogram shows that the atrium of the lateral ventricle is
hemodynamic response to fontanelle compression in infants with ventric- larger than 10 mm (66,84). Fetal MRI gives higher reproducibility and
ulomegaly. These authors used Doppler sonography to determine resis- provides better contrast, allowing a more precise measurement of the
tive indices in the anterior and middle cerebral arteries in premature ventricle (69) (Fig. 8-4). The measurement is best made on coronal
neonates who had suffered intracranial hemorrhage. Baseline resistive images parallel to the brainstem at the level of the choroid plexus (69)
indices without fontanelle compression did not correlate with intracra- (Fig. 8-4C). This measurement obviously does not determine whether
nial pressure. However, a statistically significant correlation was found the ventriculomegaly is the result of increased intraventricular pressure
between the changes in resistive index during fontanelle compression or destruction of periventricular brain tissue. Thus, some authors have

FIG. 8-4. Fetal ventriculomegaly, 26 weeks GA fetus. A. Sag-


ittal view shows normal midline structures. B and C. Axial
and coronal views show significant ventriculomegaly (14 and
17 mm largest atrial diameter; normal is <10 mm). Note that
the pericerebral spaces are still prominent, which is not con-
sistent with fetal hydrocephalus.

Barkovich_Chap08.indd 816 5/6/2011 10:26:03 PM


Chapter 8 • Hydrocephalus 817

proposed use of the ratio between the atrial diameter and the biparietal disappears in cases of destructive ventriculomegaly (91). Aqueductal
diameter (atrio-cerebral ratio); this value progressively decreases dur- occlusion or stenosis is suspected when the posterior fossa appears
ing gestation (13.6% at 22 weeks, 8% at 38 weeks) (85). Whatever the normal; this can usually be evaluated on sagittal views of the midbrain.
cause, the presence of fetal ventriculomegaly suggests that brain devel- One complication of hydrocephalus in the fetus is the pressure erosion
opment may be abnormal and is reason to do a more definitive fetal of the septum pellucidum, which may be impossible to differentiate
neuroimaging study, either a more sophisticated sonogram or a fetal from developmental absence of the septum. MRI is useful in exclud-
MR scan (86). The diameter of the atrium of the fetal lateral ventricle ing associated or causative malformations (holoprosencephaly, cal-
is of some prognostic significance. If the atrium measures larger than losal agenesis, CRASH syndrome, periventricular nodular heterotopia
10 mm, it is likely to be abnormal. Moreover, as the size of the ven- [92,93]) and in demonstrating the cause of hydrocephalus (aqueductal
tricle increases, the prognosis worsens (66,84). If the atrium measures stenosis, hindbrain malformations, hemorrhages, infection, vein of
12 mm or less, outcome is normal in 93% (87). If the atrium measures Galen malformation, torcular AV fistula, or rarely tumor).
12.1 to 15 mm, the child has a 21% to 25% chance of developmen- Therefore, while sonography will likely remain the initial screening
tal delay (66,87). If the atrium is larger than 15 mm, the chance of modality for the prenatal detection of ventriculomegaly, MRI of the
developmental delay increases to greater than 40% (84,87). However, fetus has been proved a useful adjunct to look for associated destructive
male fetuses have bigger ventricles than do female fetuses (88); indeed, lesions or parenchymal malformations that may be associated with the
some have suggested setting the upper limit of normal ventricular size ventricular enlargement (69,85,86,90,92–94).
in boys at 12 mm instead of 10 mm (89). Therefore, the prognosis for
female fetuses is worse than that for male fetuses with a similar degree Postnatal Diagnosis of Hydrocephalus
of ventriculomegaly (88). The sizes of the third and fourth ventricles
can also be measured in the fetus; typically, they are measured from The Ventricular System
transverse ultrasound images and from coronal (third ventricle) and The characteristic triad of hydrocephalus on imaging is the ventricu-
sagittal (fourth ventricle) MRI images. The diameter of the third ven- lar rounding and dilation, effacement of the pericerebral spaces over
tricle should be less than 3.5 mm on ultrasound and less than 4 mm on the convexity, and (at least in children) macrocephaly. Normal lateral
MR, while the fourth ventricle diameter should be below 4.8 mm on ventricles are typically narrow, encroached upon by the heads of the
sonography and below 7 mm on MRI (90). caudates, the thalami, the hippocampi, the calcarine sulci, and the
Differentiating between simple ventriculomegaly and hydrocepha- collateral sulci (Fig. 8-1). In hydrocephalus the ventricles are dilated
lus is not always easy. Hydrocephalic fetuses often have normal head (Fig. 8-6) and present a rounded appearance on the axial and mostly
size (Fig. 8-5A). However, the ventricular expansion effaces the typi- the coronal images, notably of the lateral angle (Fig. 8-6C–E). Several
cally prominent subarachnoid space that is normally observed over features allow the differentiation between hydrocephalus and ex vacuo
the cerebral convexity in fetuses (Fig. 8-5B, compare with Fig. 8-4B), ventriculomegaly in children (Table 8-2). For the frontal horns, bodies
especially before 32 weeks (this space progressively becomes filled by and atria, widening and rounding are not really specific as they can be
the growing brain in the last weeks of gestation). The ventricular mor- similar in atrophy. The appearance of the temporal horns, however,
phology is also useful; in hydrocephalus the ventricles expand sym- is quite specific: in atrophy, the roof and the floor of the horn remain
metrically and have a rounded configuration, especially the temporal roughly parallel, whereas in hydrocephalus, the horn is rounded with
horns. If the hydrocephalus is not too severe, the cellular layering in the the choroidal fissure becoming enlarged and the hippocampus being
cerebral mantle (ventricular zone, intermediate zone, subplate, cortex, compressed and displaced inferomedially (95) (Fig. 8-6C and D). The
described in Chapter 2) is usually preserved; this layering commonly temporal horns dilate less than the bodies of the lateral ventricles in

FIG. 8-5. Fetal hydrocephalus, twin pregnancy, 21 weeks GA fetuses. A. This image shows the two fetal heads, one which is normal,
and the other hydrocephalic. Note that the head diameter is similar in both, but the pericerebral spaces of the hydrocephalic fetus are
effaced and the ventricles enlarged. B. Axial image of the hydrocephalic brain shows the pericerebral effacement.

Barkovich_Chap08.indd 817 5/6/2011 10:26:04 PM


818 Pediatric Neuroimaging

FIG. 8-6. Triventricular hydrocephalus. Mass in the tectal plate with severe aqueductal
stenosis. A. Midline sagittal T2 image shows stretched corpus callosum and septum
pellucidum; the fornix (black arrows) appears separated from the splenium (compare
with Fig. 8-1A). Anterior third ventricular recesses are massively dilated, and the massa
intermedia effaced. A mass (m) compresses the aqueduct with dilation of its rostral
portion. Normal fourth ventricle with flow voids through its outlet (but not through
the aqueduct). The supratentorial cisterns are effaced. B. FIESTA thin imaging gives
precise depiction of the lesion of the dorsal midbrain and the location of narrowing of
the aqueduct. C. Anterior coronal T2 image shows dilated third and lateral ventricles.
Note the rounding of the temporal horns and the inferomedial compression of the hip-
pocampi. D. Posterior coronal T2 image show that the hippocampal commissure (black
arrows), normally transverse, is vertical and stretched by the enlarged ventricles (com-
pare with Fig. 8-1C). This explains why the posterior fornix seems separated from the
splenium on the midline sagittal cut in (A). E. Axial FLAIR image shows ventricular
dilation; note that the walls of the third ventricle at the level of the thalami are parallel.
The hyperintensity around the frontal and occipital horns is periventricular intersti-
tial edema, resulting from the increased intraventricular pressure that compresses the
subependymal veins, preventing fluid resorption and resulting in the accumulation of
interstitial fluid there.

Barkovich_Chap08.indd 818 5/6/2011 10:26:05 PM


Chapter 8 • Hydrocephalus 819

The ventricular index is the ratio of the ventricular diameter


TABLE 8-2 Imaging Characteristics of at the frontal horns to the diameter of the brain at the same level
(Fig. 8-8A). The ventricular angle (Fig. 8-8C) measures the diver-
Hydrocephalus (Given in gence of the frontal horns. The angle between the anterior or superior
Order of Utility) margins of the frontal horns at the level of the foramina of Monro is
diminished by concentric enlargement of the frontal horns (96). The
• Enlargement of the anterior or posterior recesses of the third diminution of the angle may be appreciated on either axial or coronal
ventricle images. The concentric enlargement of the frontal horns eventually
causes them to become rounded, producing an appearance of “Mickey
• Downward convexity of the floor of the third ventricle (results
Mouse ears” on axial scans. This concentric enlargement of the fron-
in decreased mamillopontine distance)
tal horns also causes enlargement of the so-called frontal horn radius
• Commensurate dilatation of the temporal horn with the lateral (Fig. 8-8D), which is determined by measuring the widest diameter
ventricles of the frontal horns taken at a 90° angle to the long axis of the frontal
• Narrowing of the ventricular angle horn (96). Ventricular rounding may be the single feature identified in
hyperacute hydrocephalus (within hours), when the volume of brain
• Widening of the frontal horn radius tissue is still preserved, or even becomes swollen.
• Effacement of cortical sulci The dilation and tension of the lateral ventricles modifies the
appearance of the midline. As the lateral ventricles are more dilated
The first three are the most useful signs. Although many measurements can than the third ventricle, the latter is pushed downward. The corpus
be derived from these signs (e.g., spleniochiasmal distance, third ventricular
callosum is stretched, thinned, arched upward, more than the columns
splenial distance, mamillocommissural distance [109]), these measurements
of the fornix, so that the distance between them increases (Fig. 8-6A);
are rarely necessary.
the septum pellucidum is stretched and may even be torn (Fig. 8-6B).
The part of the fornix that forms the hippocampal commissure (the
psalterium), normally transverse, becomes verticalized on each side of
patients with cerebral atrophy, and the hippocampus is not displaced the midline (Fig. 8-6D, compare with Fig. 8-1C), which explains why
(96,97) (Fig. 8-7). The rather small size of the temporal lobes and, in the posterior part of the fornix seems to be detached from the under-
particular, the relatively small volume of white matter in the temporal surface of the splenium on the midline sagittal cut (Fig. 8-6A, compare
lobes, is almost certainly the reason for this (98). The presence of tem- with Fig. 8-1A).
poral horn enlargement is not reliable in children with significant tem- In rare cases of chronic severe obstructive hydrocephalus (usually
poral lobe atrophy, such as those with Down syndrome. The Sylvian aqueductal stenosis) the ventricular anatomy may be compounded by
fissures should always be studied to assess the degree of temporal lobe atro- a ventricular diverticulum, inferior expansion of the medial wall of
phy before enlargement of the temporal horns is used to make a diagnosis the ventricular atrium through the choroid fissure behind the thala-
of hydrocephalus. If the Sylvian fissures are enlarged, or other evidence mus, into the supracerebellar and quadrigeminal cisterns (Fig. 8-9)
of temporal lobe atrophy is present, enlarged temporal horns are not a (99–102). The inferomedial atrium is affected because it is the thinnest
reliable sign of hydrocephalus. portion of the atrial wall and has the largest surface of the ventricular

FIG. 8-7. Brain atrophy in a 4-year-old child, with head circumference below the 25th percentile. A. The third and lateral ventricles
are wide with rounding of the lateral angles. However the temporal horns, although large, are small compared to the bodies of the
lateral ventricles; they retain their normal shape and the hippocampi are not displaced medially. The pericerebral spaces are patent,
notably the sylvian fissures. B. The corpus callosum is thin and the fornix appears lowered. However, the anterior third ventricular
recesses are not dilated, the lamina terminalis and the tuber cinereum are both concave. The pineal recess is prominent, but this may
be a normal variant. The suprasellar cisterns are prominent.

Barkovich_Chap08.indd 819 5/6/2011 10:26:06 PM


820 Pediatric Neuroimaging

FIG. 8-8. Various methods in the radiographic diagnosis of hydrocepha-


lus. A. The ventricular index is the ratio of the ventricular diameter at the
level of the frontal horns to the diameter of the brain measured at the same
level. This is not a very sensitive or specific measurement in the detection of
hydrocephalus because the ventricular index is enlarged in cerebral atrophy
as well as in hydrocephalus. B. Enlargement of the temporal horns commen-
surately with the bodies of the lateral ventricles is probably the most sensi-
tive and reliable sign in the differentiation of hydrocephalus from atrophy.
There is significantly less dilatation of the temporal horns than the bodies of
the lateral ventricles in cerebral atrophy. C. The ventricular angle measures
the divergence of the frontal horn. In theory, the angle made by the anterior
or superior margins of the frontal horn at the level of the foramina of Monro
is diminished when concentric enlargement of the frontal horns occurs.
Compare the illustration of hydrocephalus (top) with that of atrophy (bot-
tom). The ventricular index in both instances is 39%; however, the ventricu-
lar angle is markedly reduced in hydrocephalus. D. The frontal horn radius
measures the widest diameter of the frontal horns taken at a 90° angle to‘ the
long axis of the frontal horn. The usefulness of this measurement is dem-
onstrated by the markedly increased frontal horn radius in the patient with
hydrocephalus (top) as opposed to the patient with atrophy (bottom). Over-
all, no one measurement is completely accurate in the diagnosis of hydro-
cephalus; the size of the temporal horns, ventricular angle, the frontal horn
radius, and the size of the ventricles as compared to the cortical sulci should
all be assessed. (This figure courtesy Dr. E, Ralph Heinz, Durham, NC.)

FIG. 8-9. Ventricular diverticulum. Axial FLAIR (A),


coronal T2 (B), and sagittal T2 (C) images show expan-
sion of the left lateral ventricle with the posteromedial
aspect of it evaginating through the choroidal fissure
into the ambient cistern behind the third ventricle. This
diverticulum (d) compresses the adjacent structures
such as the tectal plate and the top of the vermis. On
sagittal cuts it may be mistaken for an arachnoid cyst,
but the continuity with the ventricle is well shown on
axial and coronal planes.

Barkovich_Chap08.indd 820 5/6/2011 10:26:07 PM


Chapter 8 • Hydrocephalus 821

system and, therefore, has the highest wall tension. If not treated, these The disproportionate enlargement of the recesses of the third ven-
atrial diverticula can compress the mesencephalic tectum with the risk tricle probably results from the relatively small resistance to expansion
of significant neurological complications; they can be mistaken for provided by the thin hypothalamus and the cisterns that surround the
arachnoid cysts in the region of the quadrigeminal cistern (Fig. 8-9C). walls of the recesses. In contrast, the thalami, which form the walls of
Coronal images are very helpful in the evaluation of these patients, the body of the third ventricle, provide a great deal more resistance to
demonstrating the continuity of the trigone of the lateral ventricle with expansion. It is important to note that even with a significant ventricu-
the diverticulum (Fig. 8-9B). lar enlargement, the lateral walls of the third ventricle (i.e., the thalami)
The third ventricle is normally slit-like, mildly wider anteriorly remain parallel to each other (Fig. 8-6E); a rounded, circular third ven-
than posteriorly. It is enlarged both in atrophy and hydrocephalus, but tricular lumen indicates the presence of a cyst, usually a suprasellar cyst
in hydrocephalus, it commonly develops rounded recesses: anteriorly with intraventricular expansion (Fig. 8-11). The anterior (chiasmal and
the chiasmal (supraoptic) and infundibular recesses and posteriorly the infundibular) recesses seem to enlarge earlier and more severely than
suprapineal recess. This feature is best appreciated on sagittal images the posterior (suprapineal) recess (105). The pulsating dilated anterior
(Figs. 8-6A, B and 8-10A), in which the anterior wall of the third ven- third ventricle may be so huge that it erodes the dorsum sellae (Figs.
tricle, normally concave anteriorly, becomes straightened, and the floor 8-6A and 8-10A). On axial images, the dilated anterior recesses of the
of the third ventricle, usually concave downward, becomes straight- third ventricle are best detected by noting that the third ventricle is
ened or convex downward. The enlargement and inferior displacement larger at the level of the optic chiasm (Fig. 8-10B) than at the level of
of these recesses can compress the infundibulum and diminish flow the middle of the ventricle. When hydrocephalus is severe and chronic,
within the hypothalamic-pituitary portal venous system, resulting in the floor of the third ventricle may rupture spontaneously, creating an
hypothalamic-pituitary dysfunction (103). When the aqueduct or the internal drainage pathway that allows CSF to escape from the ventricu-
lumen or the outlets of the fourth ventricle are occluded, the tuber lar system into the subarachnoid space (106).
cinereum is characteristically hugely dilated and bulges into the inter- The cerebral aqueduct (of Sylvius) is a narrow craniocaudal curved
peduncular cistern (Figs. 8-6A, B and 8-10A) sometimes wrapping the channel that connects the third and fourth ventricles. When narrowed
head of the basilar artery. or occluded, obstructive hydrocephalus develops (aqueductal stenosis)
The suprapineal recess of the third ventricle is another common (Figs. 8-6 and 8-10). Occlusion may be intrinsic (ependymal, usually
site of ventricular herniation. The dilated recess expands into the pos- postinflammatory) (Fig. 8-12), or extrinsic; the latter may be caused
terior incisural space, displacing the pineal gland inferiorly and, occa- by a tegmental or a tectal mass lesion (Figs. 8-6 and 8-10), or by an
sionally, elevating the vein of Galen. When large, diverticula sometimes extrinsic compression of the midbrain. It has been suggested that, in
extend inferiorly to compress the quadrigeminal plate, with conse- some instances at least, aqueductal stenosis may result from compres-
quent shortening of the tectum in the rostral-caudal direction (104) sion by expanded temporal lobes (Fig. 8-13); in this situation, the
(Fig. 8-10A). The short, thick tectum should not be mistaken for a neo- hydrocephalus may be primarily communicating in nature with aque-
plasm; isointensity with normal brain tissue on T2/FLAIR images and ductal narrowing being a result, rather than a cause, of hydrocephalus
the absence of contrast enhancement exclude a tumor. The suprapineal (104,107,108). In contradistinction, the aqueduct typically is dilated
recess may also enlarge further posteriorly and compress the tectum when the fourth ventricle is obstructed, either due to the force exerted
from the posterior direction, resulting in thinning of the tectum and by the CSF pressure or the mechanical effect of a mass expanding the
narrowing of the aqueduct (104). ventricular lumen (Fig. 8-14).

FIG. 8-10. Aqueductal stenosis caused by a tectal mass. A. Sagittal T2-weighted image shows severe dilation of the third ventricular
recesses, especially the suprapineal recess (S). Dilation of this recess is not consistently seen (see Fig. 8-6A and B); it may depend
on a preexisting prominence of the recess (see Fig. 8-7B), or on the duration or early occurrence of hydrocephalus. B. The anterior
(hypothalamic) portion of the third ventricle presents a more rounded appearance that the more posterior interthalamic portion
(see Fig. 8-6E).

Barkovich_Chap08.indd 821 5/6/2011 10:26:08 PM


822 Pediatric Neuroimaging

FIG. 8-11. Rounded appearance of the third ventricle; suprasellar cyst. A. Axial T2-weighted image shows the whole third ven-
tricle, including its interthalamic portion appearing rounded. B. Sagittal FIESTA image shows a huge suprasellar cyst displacing the
ventricular floor upward.

Due to the pulse amplitude of the CSF through the normal when the aqueduct is patent or large, because the amplitude of the CSF
aqueductal strictures, a striking flow void is normally observed there pulsations is increased (Figs. 8-2A and 8-16 to 8-18).
on the sagittal midline T2 image, extending from the posterior third The fourth ventricle in hydrocephalus ranges from normal to
ventricle to the superior fourth ventricle (76) (Fig. 8-1B). This flow markedly dilated. It is normal in obstructive hydrocephalus when the
void disappears if the aqueduct is occluded (Figs. 8-6A and 8-12), but obstruction is located above it (Figs. 8-6, 8-10, 8-12, 8-13, and 8-15). It
its presence does not exclude a stenosis; flow velocity is increased, and is often normal in communicating hydrocephalus, as well (Fig. 8-18),
therefore signal intensity is decreased, as the aqueductal diameter is so it must be remembered that triventricular hydrocephalic dilation
diminished (Fig. 8-15). Flow is also increased in cases of decreased does not necessary mean aqueductal stenosis! In most cases of commu-
meningeal compliance (e.g., in communicating hydrocephalus), even nicating hydrocephalus, however, the fourth ventricle is mildly dilated

FIG. 8-12. Intrinsic aqueductal stenosis. A. Sagittal T2-weighted image in an infant with previous perinatal hemorrhage shows
narrowing of the middle portion of the aqueduct (white arrow). Aqueductal stenosis likely develops from ependymal inflammation
in the aqueduct. Note adherent recesses in the anterior third ventricle. Normal tectal plate. B. Sagittal FIESTA image in congenital
hydrocephalus shows obstruction of the aqueduct by a transverse occluding membrane or web (white arrow) in its caudal portion;
note widening of the rostral segment and normal tectal plate.

Barkovich_Chap08.indd 822 5/6/2011 10:26:09 PM


Chapter 8 • Hydrocephalus 823

FIG. 8-13. Extrinsic aqueductal stenosis. A. Sagittal T2-weighted image in a patient with congenital hydrocephalus. Note huge dila-
tion of the lateral ventricles and to a lesser degree of the third ventricle. Small posterior fossa. The tectal plate appears compressed by
the expanded lateral ventricle. B. Axial T2. The bilateral ventricular distension results in bilateral compression of the midbrain and
deformity of the tectal plate, likely causing the aqueductal occlusion.

(Figs. 8-2, 8-16, and 8-17). Elevated CSF pressure tends to globally Section “Specific Categories of Hydrocephalus,” dealing with specific
decrease the volume of the brain, so that all ventricles and cisterns causes of hydrocephalus. Univentricular hydrocephalus occurs when
are large (Fig. 8-19A); anatomy normalizes after shunt placement the obstruction sits at one foramen of Monro; the most common cause
(Fig. 8-19B). When both the fourth ventricle and the cisterna magna is SEGA in tuberous sclerosis (see Chapter 6). The affected lateral
are involved, a posterior rotation of the vermis may occur, which is not ventricle is dilated and rounded, and the septum pellucidum bulges
a malformation but just hydrocephalus. Finally, the fourth ventricle toward the normal side. Biventricular hydrocephalus is caused by
may be hugely dilated when both in-flow and out-flow are occluded, the obstruction of both foramina of Monro; common causes include
especially if the lateral ventricles are decompressed (isolated fourth bilateral SEGA, colloid cyst of the third ventricular tela choroidea,
ventricle, discussed in more detail in section Specific categories of suprasellar cyst, or tumor. Triventricular hydrocephalus results from
hydrocephalus of this chapter). when the occlusion is in the distal third ventricle, cerebral aqueduct,
Appearances of the ventricular system usually define the type of or upper fourth ventricle; common causes are inflammatory aqueduc-
an obstructive hydrocephalus. These will be discussed in more detail in tal stenosis and midbrain/pineal, posterior third ventricular tumors.
Quadriventricular hydrocephalus is seen when the four ventricles are
dilated, either because a mass occupies the lower fourth ventricle or
because an obstructive process (typically infection) compromises the
fourth ventricular outlets. A variant of this (“pentaventricular hydro-
cephalus”) is seen when the outlets of the cisterna magna are occluded
(e.g., fibrous arachnoiditis of the posterior fossa cisterns) resulting in
a dilated cisterna magna (the “fifth ventricle”) together with the four
ventricles (Fig. 8-20)

The Cisterns
In hydrocephalus the cisterns may be effaced or dilated, depending on
the specific anatomic/hydrodynamic conditions in each patient. When
the obstruction is intraventricular the cisterns surrounding the por-
tion of the brain that is hydrocephalic tend to be effaced. In case of
aqueductal occlusion, the supratentorial cisterns are effaced, including
the suprasellar and ambient cisterns, the interhemispheric cistern, and
the subarachnoid space over the convexity (Figs. 8-6 and 8-10). In case
of fourth ventricular occlusion, the posterior fossa cisterns (cisterna
magna, cisterns around the brainstem and midbrain) are effaced, as
well (Fig. 8-20).
When the occlusion is cisternal, the cisterns located between the
FIG. 8-14. Aqueductal dilation. Large tumor (anaplastic ependymoma) occlusion and the ventricular outlets are dilated, the cisterns located
expanding the fourth ventricle and occluding its outlets results in hydro- between the occlusion and the main absorption sites are effaced. How-
cephalus with a dilated aqueduct. ever cisternal obstructions often result from diffuse arachnoid fibrosis

Barkovich_Chap08.indd 823 5/6/2011 10:26:10 PM


824 Pediatric Neuroimaging

FIG. 8-15. Incomplete aqueductal stenosis. Midline sagittal T1 (A) and T2 (B) images show aqueductal stenosis; the T2-weighted
image demonstrates a striking signal void due to rapid flow. This doesn’t mean that the aqueduct is normal, but that the stricture
results in an increased CSF flow velocity, compatible with obstructive hydrocephalus.

with multiple occlusive arachnoid adhesions; the cisterns appear enlargement of subarachnoid spaces (also called external hydrocepha-
multiloculated and sometimes frankly multicystic. lus and extraventricular obstructive hydrocephalus [EVOH], see sec-
In case of chronic communicating hydrocephalus, all ventricles tion “Benign Enlargement of Subarachnoid Spaces in Infants” of this
and all cisterns tend to be dilated other than the subarachnoid spaces chapter).
over the cerebral convexities, which are effaced as a result of the expan- It is generally assumed that increased pressure within the ventric-
sion of the lateral ventricles. Note that the sylvian fissures may be quite ular system causes compression of the brain tissue against the inner
dilated in this situation. table of the skull and consequent diminution of sulcal size. In pediatric
Finally, there are instances of hydrocephalus in which the patients, however, this conclusion may be misleading because both atro-
arachnoid spaces over the convexity are dilated; benign infantile phy and hydrocephalus can enlarge both the ventricles and sulci (110).

FIG. 8-16. Communicating hydrocephalus with accentuated signal void in widely patent aqueduct. A. Midline sagittal FIESTA
image shows a large aqueduct, mildly dilated fourth ventricle and, likely, arachnoid loculations in the suprasellar cistern (note the
mass effect upon the tuber cinereum and the pituitary stalk). B. Midline sagittal T2-weighted image shows prominent signal void
through the aqueduct, explained by increased pulsatility of the CSF through the aqueduct; restricted cisternal CSF circulation has
resulted in reduced compliance.

Barkovich_Chap08.indd 824 5/6/2011 10:26:11 PM


Chapter 8 • Hydrocephalus 825

FIG. 8-17. Communicating hydrocephalus. Sagittal T2-weighted image


shows hugely prominent flow void through the aqueduct and the mildly
enlarged fourth ventricle, down to the cistern magna. According to the
Greitz model, this results from a decreased compliance of the theca.

Further complicating the radiologic assessment is the fact that the size
of the ventricles and subarachnoid spaces is quite variable over the first
2 years of life (111). In such a case, the final indicator of hydrocepha-
lus is enlargement of the ventricular system to a degree that is dispro-
portionate to the enlargement of the cortical sulci (98). The failure of
measurements such as the bicaudate index (112,113) and the Evans
ratio (114,115) to accurately differentiate hydrocephalus from atrophy
reflects this difficulty (116). Knowledge of the head size of the infant is
essential; a large or a too-rapidly enlarging head suggests hydrocepha-
lus whereas a small or diminishing head circumference is more com-
patible with atrophy.

FIG. 8-19. Cerebral deformities caused by hydrocephalus. Infant with


macrocephaly. A. At age of 2 months, midline sagittal T1 shows huge dila-
tion of the posterior fossa cisterns, particularly dorsal to the vermis; a
diagnosis of cystic malformation of the posterior fossa was discussed, but
finally a ventriculoperitoneal shunt was inserted in the lateral ventricle.
B. At age of 8 months, midline sagittal T1 shows normal brain anatomy.
Artifact is from the shunt valve.

Acute Parenchymal Changes


Periventricular Interstitial Edema
Acute hydrocephalus is characterized by a periventricular band of low
density on CT, of low T1 and high T2/FLAIR signal on MR, which
reflects periventricular interstitial edema. Rather than an outflow of
fluid from the ventricle to the parenchyma (which in some way would
relieve the ventricular pressure) (47,48), this accumulation of fluid in
the periventricular white matter is felt to represent a failure of normal
parenchymal drainage, with consequent accumulation of fluid within
FIG. 8-18. Communicating hydrocephalus, Midline sagittal T2 image the interstitial spaces of the cerebrum (50). Current thought suggests
shows communicating hydrocephalus with normal fourth ventricle. Note that free exchange of water between the ventricular cavities and the
prominent signal void in aqueduct and fourth ventricle. extracellular spaces of the periventricular white matter is a normal

Barkovich_Chap08.indd 825 5/6/2011 10:26:12 PM


826 Pediatric Neuroimaging

On CT, interstitial edema appears as hypodensity in the periven-


tricular region; the ventricular margins may appear indistinct (Fig.
8-21A and B). On MR, the increase in water appears as a rim of T1
hypointensity and T2/FLAIR hyperintensity surrounding the lateral
ventricles (Fig. 8-21C and D). This rim may be difficult to appreciate
on heavily T2-weighted sequences, where the high signal intensity is
indistinguishable from the ventricular CSF; a proton density or FLAIR
image is much more sensitive. Diffusion-weighted images will reveal
increased water motion in the affected areas. Periventricular interstitial
edema in neonates and young infants may be masked by the normal
high water content of the immature brain.

Structural Parenchymal Changes


With the expansion of the ventricular surface, ependymal cells flat-
ten to maintain the lining but the stretching possibilities are limited
and the ependyma rapidly becomes disrupted, especially over the white
matter and the septum pellucidum (50). Choroid plexuses degener-
ate and become sclerotic, possibly with decreased secretory activity
(50). Multiple experimental and animal studies have demonstrated
that CBF is compromised in hydrocephalus, and may improve after
FIG. 8-20. Quadriventricular hydrocephalus associated with dilated shunt placement (50,117,118); an evaluation of this phenomenon is
cistern magna, due to occluded cistern magna outlets. possible using MR technology (118). Impaired cerebral energy metab-
olism in experimental hydrocephalus has been demonstrated by 31P
MRS and 1H MRS (119), but the same group could not show similar
process. Increased intraventricular pressure prevents the normal flow changes with 1H MRS in human hydrocephalus (120). Finally, some
of fluid toward the ventricles, and by compressing the subependymal white matter tracts are disrupted in acute hydrocephalus, as the ven-
veins, prevents its absorption by the deep medullary venous channels. tricular enlargement stretches the axons of the corpus callosum, the
Periventricular interstitial edema, therefore, is not seen in hyperacute septum pellucidum (which contains the limbic fibers that connect the
hydrocephalus (water cannot accumulate in a short period of time) septal cortex to the cingulate cortex), the fornices, the hippocampal
nor in chronic hydrocephalus (the ventricular pressure is not high commissure, the occipitofrontal, inferior longitudinal and corticospi-
enough), but only in acute/subacute obstructive hydrocephalus such nal tracts, and the optic radiations. Histologically this stretching has
as from an enlarging midline tumor. Periventricular interstitial edema, been shown to be associated with axonal degeneration (50). MRI with
therefore, is a complication of hydrocephalus, not a compensatory pro- Diffusion Tensor Imaging (DTI) DTI has demonstrated microstruc-
cess; it increases the volume of interstitial water, thus increasing the tural changes reflecting compression of the tracts in the white matter
mass effect upon, and causing damage to, the cerebral parenchyma lateral to the ventricles (increased FA, increased longitudinal diffusiv-
(50,51). ity but unchanged ADC) but not in the corpus callosum (decreased

FIG. 8-21. Progressive/acute hydrocephalus due to posterior third ventricular tumor: effects on the parenchyma. A and
B. Axial and parasagittal CT. Markedly enlarged lateral ventricles with low attenuation of the periventricular white matter,
reflecting periventricular interstitial edema that results from the compression of the subependymal veins and consequent
lack of absorption of the interstitial fluid from the deep white matter. This parenchymal edema develops in addition to the
hydrocephalus.

Barkovich_Chap08.indd 826 5/6/2011 10:26:13 PM


Chapter 8 • Hydrocephalus 827

FIG. 8-21. (Continued) C and D. Same case, MRI with axial FLAIR (C) and parasagittal T2 (D) images. The MR changes are similar to the CT changes,
but are more easily seen.

FA with increased ADC) (121). The changes reversed after treatment myelination of glial cells) may persist (50). Demyelination and axonal
(121). degeneration have a similar MR appearance, a ribbon of bright T2/FLAIR
signal in the paraventricular white matter, sometimes accompanied by
Herniation loss of brain volume. Axonal degeneration however does not necessarily
Increased supratentorial pressure with ventricular dilation may lead to imply neuronal loss (122) and damaged axons may regenerate, resulting
downward herniation of the mesial temporal structures along the free in return to a nearly normal appearance of the brain. This may be rather
edge of the tentorium toward the posterior fossa. In the process, the mid- spectacular in infants whose posterior cerebral mantle was compressed
brain and the aqueduct are compressed, and the vessels in the adjacent to a few millimeters thick before shunting. Subependymal fibrosis and
cisterns are stretched and compressed with resultant ischemia/infarction. sclerosis is usually found at autopsy (51); it may appear in young infants
If increased pressure and hydrocephalus occurs in the posterior fossa, as a hypointense ventricular wall interposed between the bright CSF and
those structures may herniate both upward through the tentorial incisura bright periventricular white matter on T2-weighted images.
and downward through the foramen magnum, with similar effects on the A rare late complication of chronic hydrocephalus is the local rup-
midbrain and medulla; in addition, occlusion of the foramen magnum ture of the ventricular wall and the development of a CSF-filled cleft
results in a rapid, tamponade-like increase of intracranial pressure. that dissociates the hemispheric white matter (“ventricular disrup-
tion”). Similar changes have been reported in human fetal hydrocepha-
Circulatory Arrest lus (pseudo-schizencephalies) (123).
Because neither CSF nor brain parenchyma is compressible, the main
complication of a rapid increase of intraventricular/intracranial pres- SPECIFIC CATEGORIES
sure is vascular compression: the subependymal veins are compressed
within the ventricles, the cortical veins are compressed against the
OF HYDROCEPHALUS
cranium, and the engorged intracerebral capillaries are compressed in Hydrocephalus can be divided into major categories (Table 8-3). The
between. The resulting increased capillary pressure causes brain edema first is overproduction of CSF. Essentially all patients in this category
and consequent herniations; herniated tissue fills the tentorial incisura have choroid plexus papillomas, a rare tumor. A very small number
and/or the foramen magnum, increasing the pressure even more and may have diffuse villous hyperplasia of the choroid plexus, mentioned
leading to intracranial circulatory arrest as the perfusion pressure can- earlier in the chapter (43–45). The other major category is obstruc-
not overcome the intracranial pressure. This situation may occur early, tion to normal CSF flow and absorption. This second category, termed
in hyperacute hydrocephalus, or as a later, terminal complication of obstructive hydrocephalus, is generally divided into intraventricular
progressive hydrocephalus. In hyperacute hydrocephalus, the ventricles obstructive hydrocephalus caused by intraventricular obstruction of
remain small but rounded. In both situations, the pericerebral spaces CSF flow, and EVOH, in which there is cisternal obstruction to CSF
are effaced, the parenchyma becomes edematous with loss of gray- flow or diminished absorption of CSF. In addition to these categories,
white matter contrast and, if administered, intravenous contrast pools the introduction of the Greitz model of CSF hydrodynamics based on
in the surface vessels but is not able to penetrate the parenchyma. the arterial pulsatile pressure wave leads to the concept of purely com-
municating hydrocephalus, which assumes no physical obstacle to the
Chronic Parenchymal Changes flow of CSF but an inability of the thecal sac to appropriately buffer the
Although many of the acute changes noted in progressive hydrocephalus systolic pressure waves.
(interstitial edema, demyelination, vascular compromise, axonal loss) are In this section, various causes of hydrocephalus are presented. For
reversible if shunting is performed in a timely fashion, some (ependy- each type of hydrocephalus the most common causes, the mechanisms
mal damage, gliosis, and abnormal myelination, including aberrant by which they cause hydrocephalus, the specifics of the radiologic

Barkovich_Chap08.indd 827 5/6/2011 10:26:14 PM


828 Pediatric Neuroimaging

been hypothesized that, by pulsating into the ventricular lumen, the


TABLE 8-3 Categories of Hydrocephalus papilloma increases the amplitude of the CSF pressure, thus creating
communicating hydrocephalus (27). This theory could apply to hydro-
cephalus associated with choroid plexus hyperplasia as well (Fig. 8-23).
A. Hydrocephalus secondary to overproduction of CSF
The imaging appearance of choroid plexus papillomas on CT and
Choroid plexus papillomas MR was discussed and illustrated in Chapter 7. Briefly, the tumors are
Diffuse villous hyperplasia of the choroid plexus frond-like, enhancing masses that are most commonly located in the
trigones of the lateral ventricles, with the bodies and temporal horns of
B. Hydrocephalus secondary to disturbance of CSF flow or absorption the lateral ventricles, the third ventricle, and the fourth ventricle being
Intraventricular obstructive hydrocephalus the next most common sites. In contrast to adults, in whom the fourth
EVOH ventricle is the most common site for these tumors, fourth ventricu-
lar choroid plexus papillomas are very rare in the pediatric age group.
C. Hydrocephalus secondary to a loss of thecal compliance Resection of the tumor cures the hydrocephalus in most patients.
“Normal-pressure” chronic communicating hydrocephalus
Hydrocephalus Secondary to Intraventricular
Obstruction of CSF Flow (Noncommunicating
Hydrocephalus)
appearance of each cause, and the appearance of the associated
hydrocephalus will be discussed. Tumors and Cysts
Hydrocephalus may result from obstruction of any portion of the ven-
Hydrocephalus Resulting From Excessive tricular system from the lateral ventricles to the fourth ventricular out-
flow foramina of Luschka and Magendie. The most common locations
Formation of CSF (Choroid Plexus Papillomas)
for obstruction are the locations where the CSF pathway is narrowest:
Choroid plexus papillomas (see Chapter 7) (Fig. 8-22) are large aggre- the foramina of Monro, the posterior third ventricle, the aqueduct of
gations of choroidal fronds that are microscopically similar to normal Sylvius, the fourth ventricle, and the fourth ventricular outflow foram-
choroid plexus; these neoplasms can produce great quantities of CSF ina. Tumors are the most common cause of such obstructions in the
(124,125). Choroid plexus papillomas account for 2% to 4% of child- pediatric age group.
hood intracranial tumors. They usually present during infancy with Tumors and arachnoid cysts can grow into and obstruct the
signs of increased intracranial pressure; occasionally, however, they are foramina of Monro from a number of locations. Masses originating
found incidentally at postmortem examination. In the past, the pres- in the lateral ventricles, such as choroid plexus tumors (Figs. 8-22 and
ence of hydrocephalus was thought to be related to the mass effect 8-23), ependymomas, astrocytomas, or meningiomas, typically cause
of the tumor, the presence of proteinaceous CSF, or intraventricular obstruction at the foramina of Monro, whereas masses originating in
hemorrhage with subsequent obstruction of CSF flow (obstructive the third ventricle, such as astrocytomas, choroid plexus papillomas,
hydrocephalus). More recent preoperative and postoperative studies craniopharyngiomas, or pineal region tumors, can grow anteriorly
have supported earlier suggestions that, at least in some cases, overse- or superiorly to obstruct the foramina of Monro or posteriorly and
cretion of CSF by the papilloma produces hydrocephalus (124,125) by inferiorly to obstruct the cerebral aqueduct. Suprasellar tumors and
overcoming the capacity of the system to absorb the fluid. It has also arachnoid cysts may occasionally grow upward to the foramina of

FIG. 8-22. Hydrocephalus and choroid plexus papilloma. Axial (A) and coronal (B) postcontrast T1 images show a dense, avidly
enhancing mass in the left atrium. It does not obstruct the ventricle. The bilateral, symmetrical hydrocephalus is tentatively explained
by an overproduction of CSF, by the high density of protein usually found in the CSF, and by the increased pulsatility of the tumoral
choroid plexus.

Barkovich_Chap08.indd 828 5/6/2011 10:26:14 PM


Chapter 8 • Hydrocephalus 829

FIG. 8-23. Hydrocephalus and choroid plexus hyper-


plasia. Sagittal T2 (A), axial FLAIR (B) and contrast-
enhanced coronal T1 (C) images show massive sym-
metrical enlargement of the ventricles, as well as of the
posterior fossa, suprasellar and anterior interhemispheric
cisterns; choroid plexuses are bilaterally hyperplastic. The
mechanism of the hydrocephalus is assumed to be an
overproduction of CSF.

Monro, pushing the floor of the third ventricle superiorly (Fig. 8-24). benign aqueductal stenosis from stenosis secondary to a tumor in chil-
In children with tuberous sclerosis, giant cell tumors, which originate dren with aqueductal narrowing.
adjacent to the foramina of Monro, often grow medially and obstruct Children with tumors of the fourth ventricle and cerebellum fre-
the foramina (see Chapters 6 and 7). quently have hydrocephalus by the time of presentation. The most
Obstruction of the Sylvian aqueduct is a common cause of hydro- common posterior fossa neoplasms in the pediatric age group are
cephalus. The most common cause of obstruction at this level in chil- medulloblastomas, followed by cerebellar astrocytomas and ependymo-
dren is a tumor of the pineal region (see Chapter 7). Germ cell tumors mas (see Chapter 7). Imaging studies show the tumor in the fourth
(germinoma, endodermal sinus tumor, embryonal cell carcinoma, ventricle, obstructing CSF flow directly (Figs. 8-14 and 8-25), or in
choriocarcinoma, teratoma), tumors of pineal origin (pineoblastoma, the cerebellum compressing the aqueduct and/or ventricle. In contrast
pineocytoma in young adults mostly but also in teenagers), astrocy- to cerebellar or ventricular tumors, hydrocephalus is uncommon in
tomas (from the quadrigeminal plate, thalamus, or the tegmentum pontine neoplasms.
of the midbrain), meningiomas (from the tentorium), supravermian Rarely, tumors of the spine and spinal cord (see Chapter 10) can cause
arachnoid cysts, or varices of the vein of Galen may all cause hydro- hydrocephalus. It is important to think about spinal tumors in all cases
cephalus from compression of the aqueduct (in vein of Galen arterio- of unexplained new onset hydrocephalus in children and to image their
venous malformations, a component of increased venous pressure also spines accordingly. The cause of the hydrocephalus in these cases is not
contributes to the hydrocephalus). CT identifies most of these lesions; obvious, but a number of hypotheses have been proposed. Among these
however, astrocytomas of the posterior third ventricle or arising in the are (1) increased viscosity of CSF secondary to elevated CSF protein,
quadrigeminal plate are often subtle and difficult to identify on CT. (2) obliteration of cisterna magna due to rostral extension of tumor,
Both are easier to identify by MR because MR has inherently better (3) restriction of the thecal space and poor buffering of the systolic
contrast resolution and the ability to image in the sagittal plane. Pos- pressure wave, and (4) blockage of spinal subarachnoid pathways of
terior third ventricular masses are seen as discrete masses within the CSF resorption. All of these mechanisms may have a role in some
ventricle, whereas tumors of the quadrigeminal plate are seen on MR as cases, although the fact that Chiari I malformations (see Chapter 5)
bulbous tectal masses with prolonged T2 relaxation and poorly defined are inconstantly associated with hydrocephalus makes mechanism (2)
margins (Fig. 8-6A, B, and E). Thus, MR is essential to differentiate rather unlikely. Because almost all reported cases have very high CSF

Barkovich_Chap08.indd 829 5/6/2011 10:26:15 PM


830 Pediatric Neuroimaging

FIG. 8-24. Third ventricular cyst. A. Midline sagittal T1 image shows triventricular hydrocephalus suggesting aque-
ductal stenosis, although the aqueduct looks patent. B. Midline sagittal FIESTA shows a thin cyst wall (arrowheads),
indicating that a large CSF-filled cyst occupies most of the ventricle, sparing the anterior and posterior portions only. It
blocks CSF flow through the rostral end of the aqueduct and the foramina of Monro.

protein content, it is likely that dissemination of tumor through CSF siblings of 1% to 4.5% (131). The normal mean cross sectional area of
probably plays a role in the development of hydrocephalus in these the aqueduct at birth is .5 mm2 with a range of 0.2 to 1.8 mm2 (132). In
patients (126). aqueductal stenosis, the aqueduct is focally reduced in size; narrowing
generally occurs either at the level of the superior colliculi or at the inter-
Aqueductal Stenosis collicular sulcus (132). In many instances, aqueductal stenosis is accom-
After the initial closure of the neural tube, its lumen has a relatively panied by branching of the aqueduct into dorsal and ventral channels;
uniform dimension throughout the neural axis. As the brain and spinal the dorsal channel is often divided into a group of several ductules. This
cord mature, the lumen of the neural tube expands in some areas, such condition has been termed aqueductal forking (133). Forking of the
as the cerebral ventricles, but not, or less, in others, such as the spinal aqueduct is often accompanied by fusion of the quadrigeminal bodies,
canal and Sylvian aqueduct. The lumen of the aqueduct decreases in fusion of the third nerve nuclei, and molding or beaking of the tectum.
relative size beginning in the second month of fetal life, continuing until In some patients, the shape of the molded tectum is congruent with the
birth (127). This relative narrowing appears to be caused by growth shape of the medial aspect of the adjacent temporal lobes, which are
pressures upon the aqueduct from adjacent mesencephalic structures. markedly expanded by hydrocephalus. This congruency has motivated
Aqueductal stenosis can be developmental or acquired (128–130) some authors to postulate that aqueductal stenosis may be a secondary
and is present in approximately 20% of patients with hydrocephalus. Its phenomenon in some patients, resulting from communicating hydro-
incidence ranges from 0.5 to 1 per 1000 births with a recurrence rate in cephalus and secondary compression of the quadrigeminal plate by the
dilated cerebral hemispheres (107,108,134) (Fig. 8-13).
The onset of symptoms is usually insidious; it may occur at any
time from birth to adulthood. As in all types of hydrocephalus, the
symptoms depend upon the cause of the hydrocephalus and the age of
the patient at the time of onset.
The CT appearance of benign aqueductal stenosis is dilatation of
the lateral and third ventricles with a normal sized fourth ventricle.
This appearance may be misleading, however, since the fourth ventricle
is normal in a significant percentage of patients with communicating
hydrocephalus. Moreover, as stated earlier, tectal tumors that are large
enough to obstruct the aqueduct can be missed on routine CT scans.
In all patients with suspected aqueductal stenosis the posterior third
ventricle should be carefully scrutinized for the presence of a mass; any
asymmetry of the posterior aspect of the third ventricle is indication for
an MR study to rule out the presence of a mass in the mesencephalon or
posterior thalamus. The MR findings in aqueductal stenosis are more
specific but are nonetheless quite variable. Patients with severe hydro-
cephalus generally have a stenosis in the proximal aqueduct, either at
the level of the superior colliculi or at the entrance to the aqueduct
FIG. 8-25. Fourth ventricular tumor. Midline sagittal postcontrast immediately inferior to the posterior commissure (104). In patients
T1 image shows triventricular hydrocephalus due to the large fourth ven- with mild hydrocephalus, the level of obstruction is more often in the
tricular medulloblastoma. distal portion of the aqueduct. When the distal aqueduct is stenotic,

Barkovich_Chap08.indd 830 5/6/2011 10:26:15 PM


Chapter 8 • Hydrocephalus 831

the dilated proximal aqueduct tends to displace the quadrigeminal patients have Hirschsprung disease, as well, while others may not have
plate posteriorly. MR can nearly always differentiate benign aqueductal hydrocephalus (146). Because the malformation is so complex, and
stenosis from neoplastic aqueductal stenosis. Tectal and tegmental because hydrocephalus seems to be only one small part of the disorder,
gliomas that compress and obstruct the aqueduct appear as bulbous Fransen et al. (147) have suggested that the disorder be renamed the
masses that have high signal intensity on T2/FLAIR images (Fig. 8-6E). CRASH syndrome (corpus callosum hypoplasia, retardation, adducted
These tumors uncommonly enhance (135). thumbs, spastic paraparesis, and hydrocephalus) and others have sug-
A special case of distal aqueductal stenosis is the aqueductal web. gested that L1CAM should be investigated in any X-linked disorder
An aqueductal web is a thin membrane of brain tissue situated in the associated with corpus callosum anomalies (146). As with many genetic
distal aqueduct, restricting the flow of CSF into the fourth ventricle. It disorders, the site of mutation within the L1 protein correlates with the
has been suggested that the membrane may be the result of a small glial severity of the disease (148,149).
occlusion of the caudal aqueduct that becomes an attenuated sheet Pathologic studies show that, in addition to hydrocephalus, affected
of tissue secondary to prolonged pressure from and dilatation of the patients have malformations of cortical development (see Chap-
canal above it (136). The imaging appearance is characteristic, consist- ter 5) with poor differentiation and maturation of cortical neurons
ing of a thin membrane of tissue separating a dilated aqueduct from being identified on histologic examination of the brain (143,144). Of
a normal-sized fourth ventricle (Fig. 8-12B). The importance of rec- interest, some authors have suggested that the aqueduct is narrowed by
ognizing aqueductal webs is that a third ventriculostomy (see section compression and is not primarily stenotic (150,151). Other pathologic
on “ Imaging of treated Hydrocephalus and Resulting Complications”) features include absence or diminution of the size of the corticospinal
is likely to result in complete resolution of the hydrocephalus. There tracts, fusion of the thalami, fusion of the colliculi, and absence of the
also exists a possibility of perforating the membrane via fiberoptic septum pellucidum. The corpus callosum is typically small and may be
ventriculoscopy (137–139), another means of obviating the need of an absent (46,152) (see Chapter 5).
indwelling shunt. Indeed, in some centers endoscopic aqueductoplasty Very few reports of MR studies in X-linked hydrocephalus have
is now being performed for aqueductal stenosis of many causes with been published. The only series of patients with this disorder indicates
promising results (139). that affected patients have enlargement of the massa intermedia (thal-
amic fusion), abnormal flattening of the mesencephalic tectum, a small
X-linked Hydrocephalus (HSAS, CRASH Syndrome) brainstem, and diffuse hypoplasia of cerebral white matter. Interest-
X-linked hydrocephalus, also known as Bickers-Adams syndrome, ingly, according to this report, the Sylvian aqueduct is usually patent
X-linked aqueductal stenosis and hereditary stenosis of the aqueduct (153). Others have reported similar findings of large lateral ventricles,
of Sylvius (HSAS), is a rare hereditary disorder with variable symptoms hypoplastic cerebral white matter, thin cerebral cortex with abnormal
that include mental retardation, hydrocephalus secondary to aqueduc- sulcation, fused thalami, and hypoplasia/hypogenesis/agenesis of the
tal stenosis, spasticity of the lower extremities, and clasped, adducted corpus callosum. The authors have seen very few cases of X-linked
thumbs (140). The disorder is caused by mutation of the L1CAM gene hydrocephalus and, in those cases, the findings were similar (Figs. 8-26
that has been localized to Xq28 (141,142). Identification of this disor- and 8-27).
der is important, not only because of the potential for future siblings
being similarly affected, but also because affected patients have poor Aqueductal Gliosis
neurological outcome despite early shunting (143,144). This complex Aqueductal gliosis is a postinflammatory process that is usually second-
malformation is allelic with (has the same chromosomal location as) ary to a perinatal infection or hemorrhage (128,133,154). It is becoming
the so-called MASA syndrome, which consists of mental retardation, more prevalent as newborns with bacterial meningitis or intracra-
aphasia, shuffling gait, and adducted thumbs (145). Some affected nial hemorrhage survive at increasing rates. As in benign aqueductal

FIG. 8-26. CRASH syndrome, formerly known as X-linked hydrocephalus. Fetal T2-weighted images, sagittal (A) and
coronal (B), show severe ventricular dilatation, thin cerebral mantle, absent or extremely hypoplastic corpus callosum,
compressed aqueduct, and small posterior fossa. The syndrome is related to a developmental failure of the axons to
fasciculate; as a consequence, the white matter is extremely hypoplastic. Hydrocephalus still may develop due to com-
pression of the midbrain.

Barkovich_Chap08.indd 831 5/6/2011 10:26:16 PM


832 Pediatric Neuroimaging

FIG. 8-27. CRASH syndrome, formerly known as X-linked hydrocephalus, in an older child. A. Midline sagittal T1. Small, dysmor-
phic brainstem, with a narrow aqueduct. The lateral ventricles are too large, and the corpus callosum (arrows) is thin and incom-
pletely formed. B. Coronal T1. The thalami (white arrowheads) are small and incompletely separated. The volume of cerebral white
matter is markedly diminished and the cortex has an abnormal gyral pattern with too many sulci that are too shallow.

stenosis, the onset of symptoms, which are those of hydrocephalus, recognize that the fourth ventricle is normally a thin vertical slit in the
is insidious. The ependymal lining of the aqueduct is destroyed and Chiari II malformation (see Figs. 5-162 and 5–163). The presence of a
marked fibrillary gliosis of adjacent tissue is evident at autopsy. normal-appearing or enlarged fourth ventricle in these patients sug-
On imaging studies, differentiation of aqueductal stenosis from gests a shunt malfunction or an isolated fourth ventricle that needs
aqueductal gliosis is not possible. The appearance of the two entities is CSF diversion (see Fig. 5-165).
identical on ultrasound, MR, and CT. Hydrocephalus is present in 70% to 80% of patients with the
Dandy-Walker malformation (see Chapter 5, Section “Overview of
Congenital Anomalies Midbrain and Hindbrain Development”) (157,158). It is also common
Among congenital malformations of the brain, two anomalies are, by in children with cobblestone cortex disorders (Chapter 5, Section Mal-
far, the most common causes of hydrocephalus. The more common formations of cortical development) and rhombencephalosynapsis
of these, the Chiari II malformation, has been extensively discussed (Chapter 5, Section “Classification of Midbrain-Hindbrain Malforma-
in Chapter 5 (Section “Chiari II Malformations”) and will therefore tions”).
be covered only briefly in this section. The Chiari II malformation
accounts for approximately 40% of all hydrocephalus in children (71). The Isolated (Trapped) Fourth Ventricle
Although there is still some dispute as to the cause of hydrocephalus When both the aqueduct of Sylvius and the fourth ventricular out-
in these patients, the most generally accepted theory is that of Russell flow foramina are occluded, the fourth ventricle becomes isolated from
(133). Russell demonstrated that adequate flow of CSF exists between the remaining ventricular system and from the CSF circulation of the
the ventricles and the spinal canal in patients with Chiari II malfor- subarachnoid space. Continued CSF production by the choroid plexus
mations, but that the connection between the lumbar subarachnoid of the fourth ventricle leads to progressive cystic dilatation of the ven-
space and the subarachnoid space over the cerebral convexities is poor. tricle; the dilated ventricle can then act as an expanding mass in the
The implication is that disturbance in CSF flow is most likely the result posterior fossa. The trapped fourth ventricle appears after shunting of
of the abnormal location of the exit foramina of the fourth ventricle the lateral ventricles and seems to result from mechanical or inflamma-
below the foramen magnum, within the cervical spinal canal. Since the tory changes that obstruct the aqueduct and the fourth ventricular out-
spinal canal absorbs much less CSF, hydrocephalus develops. Recent let foramina (159). Thus, CSF from the fourth ventricle cannot drain
experience showing improvement in both posterior fossa herniation proximally through the shunt catheter or distally through normal CSF
and in the degree of fetal ventriculomegaly after fetal myelomeningo- pathways; the ventricle becomes “isolated.”
cele repair does not contradict this theory (155,156). The clinical presentation depends upon the baseline neurological
It is an important clinical concept that patients with the myelom- status of the patient. In those patients who have moderate to severe
eningocele/Chiari II malformation who develop occipital headaches preexisting neurological deficits, the isolated fourth ventricle accen-
at night probably have a shunt malfunction. Fatal shunt malfunctions tuates preexisting deficits without causing prominent posterior fossa
may result in brainstem compression and consequent apneic spells. signs or symptoms. Patients with normal or near normal baseline neu-
Symptoms of brainstem compression in patients with the Chiari II rological examinations tend to present with signs and symptoms of a
malformation are often interpreted as resulting from compression of posterior fossa mass. A history of a recent ataxia, diplopia, or increas-
the medulla by the neural arches of C-1 and C-2 or by the foramen ing drowsiness is common (160). Occasionally, patients are mildly
magnum. These patients may be subjected to unnecessary decompres- symptomatic or asymptomatic and the isolated fourth ventricle can be
sions of the foramen magnum and C-1 and C-2 instead of adjustment an incidental finding (159). In these asymptomatic patients, a steady
or replacement of the ventriculo-peritoneal shunt. It is important to state is presumably reached between production and absorption of the

Barkovich_Chap08.indd 832 5/6/2011 10:26:17 PM


Chapter 8 • Hydrocephalus 833

FIG. 8-28. Isolated (trapped) fourth ventricle resulting from previous septic meningitis. A. Midline sagittal T2. Huge,
elongated fourth ventricle (V) that herniates toward the supratentorial space; it seems to communicate with the caudal
aqueduct, but not with the cistern magna. Note the multiple loculations in the third ventricle. B. Axial FLAIR. The dila-
tion of the fourth ventricle (V) extends into both lateral recesses (white arrows) presumably because of the obstruction
of both foramina of Luschka.

CSF within the fourth ventricle, resulting in the absence of signs of hemorrhage, bacterial infections, or granulomatous meningitis. Cer-
increased intracranial pressure. Close follow-up however is justified as tain features seen on imaging studies can help to differentiate the dif-
complete lack of evolutivity is uncertain and acute decompensation ferent causes of the hydrocephalus. For example, hemorrhage often
may occur. leaves a transient hemosiderin staining of the parenchyma, ependyma,
On imaging studies, the isolated fourth ventricle appears as a or pia-arachnoid (superficial siderosis). The hemosiderin will appear
large rounded or pear-shaped midline cystic structure (Figs. 8-28) in black on T2-weighted spin-echo images (Fig. 8-30B–D) (164,165) and
the posterior fossa (159–161). In some cases the lateral recesses may especially so on T2*-weighted gradient-echo or susceptibility-weighted
be ballooned (Fig. 8-28B) (162). The lateral and third ventricles are images (166,167). Although hemosiderin will remain at the site of
small or only moderately enlarged if the ventriculo-peritoneal shunt hemorrhage for months or years after parenchymal hemorrhages, it
is functioning. The aqueduct may be completely occluded or the distal is absorbed much more quickly from the ependyma and arachnoid.
aqueduct may be dilated (Fig. 8-28A). Treatment is the installation of Both infectious and carcinomatous meningitis may result in meningeal
a fourth ventricle to peritoneal shunt; endoscopic therapy is currently enhancement (Figs. 8-31 and 8-32) after administration of intravenous
experimental (138), as is stenting of the fourth ventricle to allow com- contrast (168–170). Differentiation of subarachnoid tumor from infec-
munication with the subarachnoid space (163). After shunting, there is tion, however, is not possible at this time by radiologic methods.
usually rapid reversal of posterior fossa signs, if they had been present or
progressive. In children with preexisting neurological deficits, a return Hemorrhage
to baseline is generally observed. Occasionally, further treatment, such Intraventricular hemorrhage is common in neonates, particularly
as posterior fossa decompression, may be necessary (159,160). in babies with a gestational age of 32 weeks or less (see discussion in
Chapter 4), with hydrocephalus being a frequent complication. Acute
Hydrocephalus Secondary to posthemorrhagic hydrocephalus is most commonly the result of red
blood cells obstructing the ventricular system or the arachnoid villi;
Extraventricular Obstruction of CSF this acute hydrocephalus has no prognostic implications. Scarring and
EVOH represents approximately 30% of all childhood hydrocephalus fibrosis begin to appear in the subarachnoid spaces approximately 10
(131). After leaving the foramina of the fourth ventricle, CSF normally days after the hemorrhagic event (171). The fibrosis tends to be most
enters the cisterna magna and basilar cisterns and then progresses into prominent in the region of the cisterna magna. This adhesive arach-
the cerebral and cerebellar subarachnoid spaces. Normal CSF drainage noiditis is the cause of the subacute hydrocephalus that occurs after
is jeopardized if the normal CSF pathways are obstructed by thickened germinal matrix hemorrhage in premature infants (Fig. 8-30). The pres-
arachnoid or meninges (Fig. 8-29). (The Greitz model would hypothesize ence of subacute hydrocephalus imparts a poor functional prognosis to
that the obstruction of the cisterns compromises the dampening of the the infant. However, it is important not to mistake ex vacuo ventricular
systolic pressure wave, and that the full force of the wave is then exerted enlargement secondary to periventricular white matter injury for ventric-
against the parenchyma (25,55); see the theory section earlier in this ular enlargement secondary to impaired CSF circulation. Therefore, it is
chapter.) CSF drainage is also impaired if the dural venous sinuses are important to look for evidence of periventricular white matter injury
obstructed or if pressure in the sinuses becomes elevated. Unless venous in all such cases. Irregularity of the ventricular margins, cavitation in
thrombosis is present, it is usually not possible to determine the level of the periventricular white matter and T1 and T2 shortening within the
the extraventricular obstruction to CSF flow from imaging studies. periventricular white matter (see Chapter 4) all suggest that brain injury
Impaired flow of CSF through the subarachnoid spaces (or reduced has occurred. Of course, as with all potential cases of pediatric hydro-
compliance [55]) may result from recent or remote intracranial cephalus, correlation with head circumference measurements is vital.

Barkovich_Chap08.indd 833 5/6/2011 10:26:17 PM


834 Pediatric Neuroimaging

FIG. 8-29. Extraventricular obstruction of the CSF pathways.


A. Midline sagittal T2. The severe hydrocephalus was initially
explained by the large suprasellar cyst that was felt to obstruct
the foramina of Monro and the third ventricle; however, the
prominent flow void in the aqueduct suggests that the obstruc-
tion must be located somewhere else. The patient was treated by
endoscopic fenestration of the third ventricle and cyst. B. Mid-
line sagittal FIESTA image after surgery confirms the diagnosis
of suprasellar cyst and the patency of the foramina of Monro,
third ventricle and aqueduct, but shows that the suprasellar
cistern remains obstructed and the hydrocephalus persists.
C. Midline sagittal T2 image, acquired in the same study as image
B shows an extremely prominent aqueductal-fourth ventricle-
foramen of Magendie signal void, suggesting hyperdynamic flow
due to reduced compliance. This observation strongly suggests
that the hydrocephalus is a result of restricted cisternal CSF flow
due to the suprasellar location of the cyst. See text for further
explanation.

Subarachnoid hemorrhage in full term infants is most often caused granulomatous and fungal meningitis. Hydrocephalus appears to be
by trauma (see Chapter 4). The causes of acute and chronic hydro- related to the duration and severity of the meningeal infections; in gen-
cephalus in these infants are similar to the causes of hydrocephalus eral, the longer the delay in treatment, the worse the prognosis for the
in premature infants with intraventricular hemorrhage; however, the patient. As a general rule, bacterial meningitis tends to produce cerebral
prognostic value of the hydrocephalus is not as great in term infants as cortical arachnoiditis, whereas granulomatous or parasitic meningiti-
in premature infants. des produce cisternal obstruction (Fig. 8-31) (46). Viral meningiti-
des may result in obstruction at either point, albeit rarely. Ventricular
Meningitis dilatation, which presumably results from transiently diminished CSF
Inflammation of the leptomeninges may lead to obstruction the CSF absorption, is the most common finding on ultrasound, CT scans, or
pathways (see Chapter 11, Section “Meningitis”). In the acute phase, unenhanced MR scans in patients with bacterial meningitis. Contrast-
hydrocephalus is caused by blockage of the CSF flow pathways by enhanced MR scans often show localized or diffuse meningeal enhance-
purulent fluid. Another component of acute hydrocephalus may be ment in bacterial and fungal meningitides (see Fig. 8-31 and examples
inflammation of the arachnoid granulations (46,172). In the chronic in Chapter 11) (169). Arachnoidal scarring from meningitis may also
phase, organization of exudate and blood results in fibrosis of the suba- result in arachnoid loculations, which may be indistinguishable from
rachnoid spaces with subsequent obstruction to normal CSF flow and arachnoid cysts.
resultant hydrocephalus (46,172).
Clinically significant hydrocephalus is less common in bacterial CSF Seeding of Tumor
meningitis than in granulomatous and fungal meningitides. Indeed, A number of neoplastic processes can involve the subarachnoid space
hydrocephalus is almost always present at the time of presentation in diffusely (see Chapter 7). The most common of these in children are

Barkovich_Chap08.indd 834 5/6/2011 10:26:18 PM


Chapter 8 • Hydrocephalus 835

FIG. 8-30. Post hemorrhagic hydrocephalus in a former premature infant. A. Midline sagittal FIESTA image shows severe hydro-
cephalus, dehiscent septum pellucidum, and an obstructed aqueduct. Low signal intensity around the brainstem and inferior vermis
suggests hemosiderin from intraventricular hemorrhage. B–D. Axial T2*-weighted images show hypointensity (black arrows) from
magnetic susceptibility effects of blood on the surface of the brainstem and cerebellum (B), choroid plexuses and walls of the lateral
ventricles (C and D).

medulloblastoma, germinoma, leukemia, and lymphoma; it can also be Venous Hypertension


seen in a variant of pilocytic astrocytoma, the pilomyxoid astrocytoma Obstruction of the cerebral veins and sinuses is thought to be a cause of
(173). This is in contrast to adults where adenocarcinoma is the most communicating hydrocephalus in infants (176–178). It appears that the
common tumor to show diffuse meningeal involvement (174). increased intracranial venous pressure may produce either hydroceph-
Older patients with CSF seeding of tumor (also called carcinoma- alus or pseudotumor cerebri, depending upon the patient’s age (176).
tous meningitis) typically present with headache, stiff neck, and cranial Hydrocephalus is more likely if the patient is less than 18th months
nerve palsies. Hydrocephalus occurs only later in the course of the dis- of age, whereas pseudotumor cerebri is noted if the patient is more
ease. As with infectious meningitis, ventricular enlargement is the most than 3 years of age. This difference is thought to result from an expan-
common manifestation of this process on ultrasound, CT, and unen- sile calvarium and softer, less myelinated parenchyma in the infant;
hanced MR scans. Occasionally, diffuse meningeal enhancement may both of these factors allow greater ventricular dilatation under high
be seen on contrast-enhanced CT scans. Significant enhancement of pressure (179–182). Once the cerebrum is myelinated and the sutures
the subarachnoid spaces and cranial nerves (particularly cranial nerves are fused or the calvarium is otherwise prevented from expanding,
5 and 8) is typically seen on contrast-enhanced MR scans (Fig. 8-32) intracranial hypertension occurs without ventricular enlargement and
(168,170,175). results in pseudotumor cerebri (176,179). The hydrodynamic theory

Barkovich_Chap08.indd 835 5/6/2011 10:26:19 PM


836 Pediatric Neuroimaging

FIG. 8-31. Hydrocephalus associated with tuberculous menin-


gitis. Midline sagittal (A), coronal (B), axial (C) T1 images after
contrast administration. The thickened, inflamed leptomeninges
show diffuse enhancement over the surface of the brainstem,
hypothalamus, cerebellum, and cranial nerves, associated with
significant triventricular dilation.

of Greitz would suggest that, in infant with an elastic skull, CSF would the arterial systolic inflow not being properly buffered because of the
accumulate immediately above the obstruction, in the pericerebral increased pressure in the dural sinuses and, thus, the compliance of the
subarachnoid spaces, much more than in the ventricles (25). system is decreased (25).) The resultant increased intracranial pressure
Ventricular enlargement results from skull base abnormalities results in expansion of the calvarium and ventricles in infants until the
in a number of syndromes. These include achondroplasia (183,184) pressure normalizes (186,187) or the hydrocephalus becomes compen-
(Fig. 8-33), craniofacial syndromes that result in multisutural cranio- sated (192). Typical imaging signs of hydrocephalus are not reliable in
synostosis (Apert syndrome, Carpenter syndrome, Pfeiffer syndrome, these patients. For example, temporal horn enlargement results from
Crouzon syndrome, and others described in Chapter 5, Section “Cran- expansion of the middle cranial fossa and frontal horn enlargement may
iosynostosis”), and the Marshall-Smith syndrome (a syndrome of accel- be the result of the enlarged anterior fontanelle, not necessarily from
erated osseous maturation and CNS malformations) (178,185–187). It increased intraventricular pressure. Also, because of the sutural synos-
has been postulated that the ventricular enlargement in all of these syn- tosis, the bodies and trigones of the lateral ventricles may not enlarge
dromes results from diminished venous outflow through the small jugu- until a cranial expansion has been performed (193). The indication
lar foramina that, in turn, result from hypoplasia of the skull base (188) for shunting in these patients seems to be based primarily on the pres-
(Fig. 8-33D). These theories are supported by experimental evidence in ence of severe ventricular dilation or evidence of persistent intracranial
the case of achondroplasia (189,190) and Crouzon syndrome (191). The hypertension such as papilledema (193). A recent study suggests that the
mechanism of this ventricular enlargement is believed to be an increase intracranial pressure normalizes in patients with craniosynostosis syn-
in superior sagittal sinus pressures, leading to a decreased pressure gra- dromes at the age of approximately 6 years as a result of the enlargement
dient across the arachnoid villi, from the subarachnoid spaces to the of stylomastoid emissary veins (transcalvarial veins that allow venous
superior sagittal sinus. (The hydrodynamic theory would suggest that drainage into the stylomastoid venous plexus) (194).

Barkovich_Chap08.indd 836 5/6/2011 10:26:20 PM


Chapter 8 • Hydrocephalus 837

FIG. 8-32. Hydrocephalus associated with leptomeningeal tumoral dissemination (high risk medulloblastoma). Postcontrast
T1-weighted axial (A) and coronal (B) images show a thick enhancing layer of leptomeningeal carcinomatosis (carcinomatous men-
ingitis), mainly overlying the cerebellar folia.

Progressive head enlargement in infants can also result from of choice to evaluate the venous sinuses. However, MR venography can-
thrombosis of the dural venous sinuses, presumably via the same mecha- not determine pressure differential across foci of narrowing in the dural
nism. MR venography can be helpful to detect obstruction of the dural venous sinuses. In difficult cases, it may be necessary to do a catheter
venous sinuses in children with increased CSF pressures with no other venogram and measure pressures in the sinuses.
obvious cause. As two-dimensional (2D) time-of-flight MR venogra-
phy may show apparent narrowing of the distal transverse and sigmoid Normal Pressure Hydrocephalus
sinuses that are caused by complex flow (sometimes by the presence of Normal pressure hydrocephalus (NPH) is a form of hydrocephalus in
an arachnoid granulation or flow from the vein of Labbé) in those areas, which a pressure gradient exists between ventricle and brain paren-
time-resolved contrast-enhanced MR venography (see Chapter 1, Section chyma, despite the fact that CSF pressure is within the normal range
“Magnetic Resonance Angiography and CT Angiography”) is the study at the time of lumbar puncture. Intermittently high pressure probably

FIG. 8-33. Hydrocephalus from venous hypertension in a 15 month old infant with achondroplasia. A. Midline sagittal T1-weighted
image shows the frontal bossing characteristic of the disease. Supratentorial ventricles are large, the posterior fossa is small, the
cranio-vertebral junction and cervical spinal canal are narrow. B. Coronal T2 image shows supratentorial ventricular dilation with
typical rounding of the temporal horns compressing the hippocampi, and a prominent suprapineal recess (black arrow).

Barkovich_Chap08.indd 837 5/6/2011 10:26:21 PM


838 Pediatric Neuroimaging

FIG. 8-33. (Continued) C. Axial FLAIR image shows diffuse ventricular dilation with prominent subarachnoid spaces over the
frontal lobes and anterior interhemispheric fissure (associated subrachnoid enlargement). D. MR venogram shows both sigmoid
sinuses to be interrupted in the jugular foramina. Prominent collateral venous flow can be seen through emissary veins and the
venous plexuses around the foramen magnum (white arrows).

results in a compromise of normal compensatory pressure mechanisms therefore, the venous backpressure that keeps open the venules and
(195,196); this is called a loss of compliance in the hydrodynamic the- capillaries is lost, compromising the optimal perfusion and exchange
ory (25). The fluctuating high pressures may combine with impaired conditions that prevailed between the vessels and the brain (25). This
regional CBF to cause ventricular enlargement and parenchymal model has been confirmed by several reports in which the registration
destruction (131). The most common cause of NPH is communicating of the velocity curve at the aqueduct using cine phase-contrast imaging
hydrocephalus with unidentified arachnoidal damage interfering with in cohorts of patients demonstrated a significant increase of the CSF
normal CSF dynamics, or loss of elasticity of the dural sac. Primary stroke amplitude as compared with normal controls (59–62). On con-
events leading to NPH include neonatal intraventricular hemorrhage, ventional diagnostic this is expressed by a significantly increased signal
spontaneous subarachnoid hemorrhage, intracranial trauma, infec- void at the aqueduct on sagittal T2 FSE/TSE imaging.
tions, and surgery (197). Among the pediatric population, it is most
commonly seen in those who have underlying neurological pathology
such as complications of meningitis or germinal matrix/intraventricu- BENIGN ENLARGEMENT OF THE
lar hemorrhage (198). NPH, therefore, is not merely a disease of adults SUBARACHNOID SPACES IN INFANTS
but occurs in children, as well.
The ultrasound, CT, and MR appearance of NPH in children is Definition
indistinguishable from that of other forms of communicating hydro- A pattern of enlarged CSF spaces with normal to slightly increased
cephalus. However, measurements of CSF flow with flow-sensitive ventricular size in infants (Fig. 8-34) is fairly common. The affected
MRI methods have demonstrated that the CSF stroke volume (vol- children are usually neurologically normal infants without evidence
ume of CSF displaced with each cardiac systole) is reduced by 50% at of prior brain injury (199). This appearance rarely causes clinical
the craniovertebral junction and the venous stroke volume is reduced concern and, indeed, is rarely noticed unless the infant has macro-
by 33% in the dural sinuses in communicating hydrocephalus. At the cephaly or rapid head growth; under these circumstances, the large
same time, intracranial pressure monitoring reports a 6-fold increase CSF spaces often raise concerns for hydrocephalus (200–203). This
of the CSF pulse pressure (25). This association of decreased stroke phenomenon of large CSF spaces in developmentally and neurologi-
volume and increased pressure wave reflects the loss of compliance. cally normal children with macrocephaly has been variously called
It means that the arterial expansion resulting from cardiac systole is benign extra-axial collections of infancy, benign idiopathic external
decreased and that the arterial pulse wave, instead of being transmit- hydrocephalus, EVOH, benign subdural effusions of infancy, benign
ted to the CSF, is transmitted directly to the vascular lumen. Thus, the macrocephaly of infancy, and benign enlargement of the subarachnoid
intravascular pressure is increased in capillaries and veins, preventing spaces (200,202,203). The term benign enlargement of the subarach-
the appropriate absorption of CSF by the parenchyma. Because the noid spaces probably is most appropriate and can be applied to neuro-
pulse wave is transmitted into the vessels rather than via the pericere- logically normal infants with large CSF spaces whether macrocephaly
bral CSF, the brain expands, compressing the cerebral ventricles. The is present or not.
ventricular compression, in turn, causes increased amplitude of the
pressure wave within the ventricles and, especially, in the aqueduct.
Clinical Manifestations
The increased pressure in both the vessels and the ventricles results
in a slowly progressive loss of brain tissue. In addition the compres- In the absence of other anomalies, children with benign enlargement
sion of the draining veins by the pericerebral CSF pulse pressure is lost; of the subarachnoid spaces develop normally (203). If macrocephaly is

Barkovich_Chap08.indd 838 5/6/2011 10:26:22 PM


Chapter 8 • Hydrocephalus 839

FIG. 8-34. Benign enlargement of the subarachnoid spaces.


This 1-year-old infant was referred for macrocephaly, with head
circumference above the 95th percentile. A–C. Axial, coronal and
sagittal T2 images show significant widening of the subarachnoid
spaces over the anterior convexity. With proper windowing, the
bridging veins are clearly seen coursing from the brain surface to
the inner layer of the dura. The lateral ventricles are only mildly
dilated, and the temporal horns are normal. D. Midline sagittal
T2-weighted image shows diffusely prominent CSF spaces. A
faint flow void can be seen through the aqueduct. E. Midline
sagittal FIESTA image shows normal midline anatomy other than
the prominence of the CSF spaces.

Barkovich_Chap08.indd 839 5/6/2011 10:26:22 PM


840 Pediatric Neuroimaging

present, the head circumference tends to be in the high normal range table with veins medial to it. In patients with enlarged subarachnoid
at birth and increases rapidly during the first few months of life; it spaces, the veins course directly into superior sagittal sinus (210,211).
is generally well above the 95th percentile at the time of presenta- Occasionally, patients with clinical courses and CT scans strongly
tion (usually between the ages of 2 and 7 months). It is important to suggestive of benign enlargement of the subarachnoid spaces have had
recognize that this rapid head growth does not necessarily represent bloody subdural collections, superimposed upon large subarachnoid
hydrocephalus or the presence of subdural hematomas, particularly in spaces, detected on MR scans or ultrasound (208,210) (Fig. 8-35).
the absence of developmental delay or signs or symptoms of increased Wilms et al. have reported that infants with bloody subdural collec-
intracranial pressure. In fact, when followed without intervention, tions have a higher incidence of difficult births or postnatal traumatic
the head growth curve of affected infants tends to stabilize along a incidents and are more likely to have symptoms of increased intracra-
curve parallel to the 95th percentile by the age of 18 months. Both nial pressure, such as vomiting and somnolence (210). However, many
the size of the subarachnoid spaces and the head circumference typi- reports describe infants with mildly hemorrhagic subdural collec-
cally return to normal after the second birthday (202). Cisternogra- tions superimposed upon large subarachnoid spaces in which exten-
phy has demonstrated slow flow over the cerebral convexities in these sive investigations have revealed no evidence of significant trauma
patients (201); the enlarged subarachnoid spaces, therefore, are pro- (212–214). It is postulated that the large CSF spaces in these patients
posed to result from delayed maturation of the arachnoid villi or other may make these infants more susceptible to subdural hematoma forma-
absorption sites such as the nasal mucosal-lymphatic pathway under- tion secondary to rather mild trauma, similar to the condition encoun-
lying the cribriform plate (34,40). The fact that this same pattern of tered in patients with middle cranial fossa arachnoid cysts (215,216).
enlarged subarachnoid spaces may be found also in association with All babies with hemorrhagic subdural collections should, nonetheless,
high venous pressure lends credence to this tentative explanation. In be carefully screened by child protective services, as most will be found
most cases, however, benign enlargement of the subarachnoid spaces to have suffered some sort of trauma (217).
is idiopathic; it is sometimes familial (204). Histologic examination in
affected patients has shown thickening of the arachnoid, which may
be a normal developmental phenomenon or perhaps a reactive change
IMAGING OF TREATED HYDROCEPHALUS
that helps to increase resorption of CSF (205). AND RESULTING COMPLICATIONS
As discussed in Sections Classification of hydrocephalus and Clinical
Imaging aspects of hydrocephalus, severe long-lasting hydrocephalus will cause
damage to the brain. In dogs, the brain is not permanently damaged
The mere presence of enlarged subarachnoid spaces is not sufficient until the cerebral mantle is compressed to a thickness of less than 4
to make this diagnosis. Enlarged subarachnoid spaces can be caused mm (218). Persistent compression of the brain results in gliosis, cor-
by a number of factors, including ACTH and corticosteroid therapy tical distortion, damage to neurons, tearing of axons in the internal
(206,207), dehydration, malnutrition (208), total parenteral nutrition, capsule, and myelin destruction (46,218,219). Therefore, treatment of
and cancer chemotherapy, in addition to the mechanisms discussed uncompensated hydrocephalus is mandatory.
previously in this chapter. The presence of these factors should be ruled In hyperacute hydrocephalus, ventricular tapping with external
out before the diagnosis of benign enlargement of the subarachnoid ventricular drainage relieves the ventricular tamponade and prevents
spaces (or atrophy, or hydrocephalus) is considered. the circulatory arrest caused by the compression of the vascular bed
Typical imaging findings in patients with benign enlargement of (see section Clinical aspects of hydrocephalus). Follow-up imaging,
the subarachnoid spaces include slightly enlarged lateral and third
ventricles, and subarachnoid space that is wider than normal in
frontal region, anterior interhemispheric fissure, and Sylvian fissures
(Fig. 8-34) (199). The chiasmatic cistern is also typically enlarged
(Fig. 8-34D and E). The degree of dilation of the lateral ventricles is
roughly proportional to the width of the frontal subarachnoid space.
Imaging studies may not be helpful in the distinction of benign enlarge-
ment of the subarachnoid spaces from hydrocephalus or, for that mat-
ter, from atrophy. As discussed earlier, scrutiny of the anterior and
posterior recesses of the third ventricle and the temporal horns (see
figures in the early sections of this chapter) may be helpful in the dif-
ferentiation from hydrocephalus. Symmetry of the extraparenchymal
fluid is an important finding, especially on CT; asymmetry is unusual
in benign macrocephaly and may indicate the presence of traumatic
subdural collections (209). Both MR and ultrasound can differentiate
subdural collections from enlarged subarachnoid spaces by visualiza-
tion of cortical veins (Fig. 8-34A–C). On MR, examining the first echo
of a T2-weighted sequence or a FLAIR image is optimal. In benign
enlargement of the subarachnoid spaces, veins are identified as curvi-
linear structures that are adjacent to the inner table of the calvarium;
with subdural collections, the veins are displaced from the inner table.
Also most subdural collections have high protein content and have FIG. 8-35. Benign enlargement of the subarachnoid spaces with small
higher signal intensity than CSF on T2-weighted or FLAIR images. On subdural hematoma. Axial CT of a 1-year-old infant with head circumfer-
cranial ultrasound, differentiation is made by angling the transducer ence above the 95th percentile shows prominent subarachnoid spaces over
to follow the veins into the superior sagittal sinus. In infants with sub- the frontal convexity. The right frontal subdural collection of intermediate
dural collections, the echogenic pia-arachnoid is displaced from inner attenuation (black arrows) is highly suggestive for a subdural hematoma.

Barkovich_Chap08.indd 840 5/6/2011 10:26:23 PM


Chapter 8 • Hydrocephalus 841

typically restricted to the acute period, assesses the ventricular size, the choroid plexuses is the procedure of choice, provided the cisterns are
morphological features associated with high intraventricular pressure, patent (223,224).
and the related condition of the parenchyma. Shunt systems are composed of a ventriculostomy tube, a reservoir
In less acute situations, hydrocephalus may be treated by endo- (optional), a valve, and a peritoneal tube. The ventriculostomy tube is
scopic third ventriculostomy or by CSF diversion in the form of a ven- inserted either into the frontal or occipital horn of the lateral ventricle
triculo-peritoneal or a ventriculo-atrial shunt. Third ventriculostomy through a small hole in the calvarium. This is attached to the reser-
is a popular treatment because it eliminates the necessity of having an voir, which lies subcutaneously over the calvarial defect. The valve is
indwelling shunt, which can become obstructed and can be the cause set to a certain pressure that must be attained before drainage occurs;
of hemorrhage, infections, foreign body reactions, slit ventricle syn- the presence of the shunt system restores the compliance of the system
drome, and other complications listed in this section. In third ventricu- and allows both dampening of the systolic pressure wave and drain-
lostomies, a fiberoptic endoscope is used to create a hole in the floor of age of the accumulated fluid. Some valves are pressure-adjusted with
the third ventricle, immediately anterior to the mamillary bodies. This an external magnetic device; although these magnetic devices are not a
hole allows the intraventricular CSF access into the interpeduncular contraindication to MRI, it is almost always necessary to reset the valve
and suprasellar cisterns, from where it can flow laterally and upward after the study. The peritoneal tube is run subcutaneously from the valve
over the cerebral convexities to sites of CSF resorption. Thus, the into the peritoneal cavity. The radiologist is usually asked to assess the
obstructions are bypassed. Third ventriculostomies appear to be most function of the shunt by determining the position of the tip of the ven-
useful in obstructive hydrocephalus; as discussed earlier, obstructive triculostomy tube and the reduction in ventricular size resulting from
hydrocephalus in children is most often the result of obstruction in the its placement. In general, the tip of the ventriculostomy tube should lie
region of the Sylvian aqueduct, fourth ventricular outflow foramina, within the lateral ventricle in the region of the foramen of Monro. The
cisterna magna, or basal cisterns. Third ventriculostomy may also be tube is hypointense on T1- and T2-weighted and FLAIR MR images
used in cases of communicating hydrocephalus, as by-passing the cau- (Fig. 8-36A–C) and has high attenuation on CT (Fig. 8-36D). Some
dal ventricular channels (aqueduct, fourth ventricle) might increase diminution in ventricular size should be apparent soon after the place-
the compliance (220). However, third ventriculostomy is not effec- ment of a shunt; if the ventricular size is unchanged after 2 to 3 days, the
tive when loculations are present in the subarachnoid spaces, such as patency of the shunt system should be questioned. It has been shown
the prepontine, interpeduncular or suprasellar cisterns. Studies have that if the cerebral mantle measures 2 cm or more in thickness after
shown that third ventriculostomies result in successful resolution of shunting, patients usually attain average intellectual development (46).
hydrocephalus in 80% of children so treated (221). If the third ven- Surgical cannulation of the aqueduct was proposed in the 1970s
triculostomy does not result in adequate resolution of hydrocephalus, to 1980s for treating aqueductal stenosis (128,225) and reintroduced
a ventricular shunt is placed. Advantages of endoscopic third ventricu- in the past decade using fiberoptic ventriculoscopy, with or without
lostomy include a more physiologic CSF circulation, lower incidence of stenting (139,226). It is uncommonly used and the reader is unlikely to
infections, and better response of infection to antibiotic therapy. encounter such cases.
Third ventriculostomies are less useful in young infants, in whom a
higher incidence of parenchymal injury and vascular injury is reported; Radiologic Assessment of Third
in addition, the communication through the floor of the ventricle is
Ventriculostomies
more likely to spontaneously close (222). In countries where appro-
priate follow-up of patients is difficult for economic or geographic Although sonography, CT, or MR can be used to assess shunted hydro-
reasons, third ventriculostomy associated with cauterization of the cephalus, MR is the study of choice for the assessment of patients

FIG. 8-36. Appearance of a ventriculoperitoneal shunt (white arrows) on MR (A and C) and CT (D). On MR the shunt appears
as a hypointense linear structure extending from the ventricle to the extracranial soft tissues. It appears as a high attenuation linear
structure on CT.

Barkovich_Chap08.indd 841 5/6/2011 10:26:24 PM


842 Pediatric Neuroimaging

FIG. 8-36. (Continued)

treated with third ventriculostomy, as only by MR can one assess the physician is most comfortable. In addition, detailed morphological
surgically induced defect in the floor of the third ventricle and the CSF analysis can be provided by using CISS/FIESTA submillimeter imag-
pulsing through it. ing. This sequence is not sensitive to flow but demonstrates the ventric-
In children who have undergone successful third ventriculostomies, ulostomy well and can identify occlusive septations in the surrounding
the reduction in ventricular size is usually slower and less dramatic cisterns better that flow-sensitive sequences.
than in those who have been treated using ventricular shunts (227).
The ventricular size after third ventriculostomy usually decreases over
Shunt Malfunctions
several months, instead of several days as in patients with ventricu-
lar shunt catheters (228). On follow-up, the ventricles remain larger Shunt malfunctions are manifested clinically by symptoms of increased
than they would usually be after shunt placement. This difference in intracranial pressure, persistent bulging of the anterior fontanel, exces-
magnitude of ventricular size reduction is likely related to the fact that sive rate of head growth in the infant, or seizures (131,232). On imag-
indwelling ventriculostomy catheters dampen the pulsations of CSF ing studies, shunt malfunction is usually manifested by increasing
emanating from the choroid plexus or possibly to a reduced absorp- ventricular size (Fig. 8-39A and B). In infants, this is usually assessed
tion capacity in the affected children (229). by sonography. In older children, CT is the standard study, but devel-
Imaging assessment of the third ventriculostomy is aimed at opment of fast imaging (half-Fourier single-shot RARE sequences and
demonstrating flow passing through the hole in the floor of the third PROPELLER sequences) now allow rapid assessment of ventricular size
ventricle. This can be best accomplished noninvasively by the use of by MR without sedation and without ionizing radiation (233–236). If
MR. Lev et al. used phase-contrast MR flow studies to determine ratios time is available on the MR scanner, serious consideration should be
of CSF flow velocities in patients with third ventriculostomies (230). given to using these techniques. Shunt failure does not always result
They found that the ratio of velocity of CSF in the prepontine space in ventricular enlargement, however. Some patients have considerable
to that in the anterior cervical spinal subarachnoid space was signifi- scarring in and around the ventricular walls, causing decreased com-
cantly higher in patients with patent third ventriculostomies than in pliance of the brain and no enlargement or minimal enlargement of
normal patients. Those patients with velocity ratios lower than normal the ventricular system in the presence of shunt malfunction (see sub-
needed revisions (230). Fischbein et al. found that sagittal 3 to 4 mm sequent section on the “Slit Ventricle Syndrome” [237,238]). The most
fast spin-echo sequences show hypointensity in the region of the ven- common cause of malfunction is occlusion of the ventricular catheter
triculostomy secondary to rapid pulsatile flow. In their study, the fast by choroid plexus or glial tissue that grows into the lumen of the cath-
spin-echo studies were as sensitive as cine phase-contrast studies (227) eter. This diagnosis can only be inferred from imaging studies, which
(Fig. 8-37). Kim et al. used a combination of T2-weighted images and will show enlarging lateral ventricles in spite of a well-placed ventricu-
phase-contrast images (228). Hoffman et al. used a steady-state free lostomy tube and intact connections of the shunt system.
precession sequence and consistently found loss of the steady state (and Disconnection of the shunt components can occur at any point
consequent loss of signal) in the inferior third ventricle and suprasellar within the system; it most commonly occurs where the various compo-
cistern in patients with patent third ventriculostomies but no loss of nents are joined. Although the site of shunt malfunction (ventricular
the steady state (persistent high signal intensity in the inferior third portion versus peritoneal portion) can often be determined clinically,
ventricle) in patients with occluded third ventriculostomies (231) (Fig. plain film studies of shunt components (Fig. 8-39C), positive contrast
8-38). As all of these techniques rely on detecting flow in the inferior plain film radiography, or radionuclide studies are sometimes neces-
third ventricle and suprasellar/interpeduncular/prepontine cisterns, it sary (239–243). If radionuclide studies are performed, both dynamic
is likely that all of them work. Therefore, choice of the precise tech- and static studies should be obtained (239). The “scout” film from a
nique to be used should depend upon which techniques are available on CT scan will sometimes show a disconnection at the valve that is not
the MR scanner being used and with which technique(s) the imaging appreciated on the axial images; therefore, the scout film should always

Barkovich_Chap08.indd 842 5/6/2011 10:26:24 PM


Chapter 8 • Hydrocephalus 843

FIG. 8-37. Endoscopic third ventriculocisternostomy (ETV) efficacy assessment. A. Midsagittal FIESTA image shows a wide defect
(black arrow) in the anterior part of the floor of the third ventricle. B and C. Cine phase-contrast imaging demonstrates good CSF
flow (hypointensity in [B], hyperintensity in [C]) across the defect in the floor of the ventricle. D. Sagittal T2-weighted image dem-
onstrates a prominent signal void (black arrow) at the ventriculostomy site. FSE T2 is felt to be similarly diagnostic as cine phase-
contrast imaging for evaluating the patency of an ETV.

be analyzed. In addition to showing the location of obstruction or dis- carefully, flow rates within the shunt can be calculated (246). Parameters
connection, shunt function studies are of value when symptoms of are set to detect flow with velocity greater than a certain minimum. If
shunt malfunction are vague and clinical testing of shunt function is flow is seen within the shunt tubing, therefore, one may confidently
equivocal. Note that the reservoir is not always radio-opaque and, there- conclude that a rate of flow above that minimum is present within the
fore, may appear as a discontinuity of the shunt system on plain radio- shunt system. However, CSF production and, therefore, CSF flow, vary
graphs. It is essential to communicate with the treating neurosurgeon from minute to minute (247); therefore, a negative study (a study show-
or perform a positive contrast or radionuclide study in order to dif- ing no flow) is indeterminate. CSF may be flowing through the shunt
ferentiate a disconnection from the presence of a radiolucent shunt at a velocity below the minimum detectable rate (245). In addition,
component. the amount of CSF flowing through the shunt may vary with position
Using special techniques described in Chapter 1, CSF flow can be (e.g., upright versus supine). As a result of these considerations, a lack
analyzed by MR techniques (81,82). When used properly, the CSF flow of detection of CSF flow on an MR study is not necessarily diagnostic
techniques can be used to analyze flow in ventricular catheters and of shunt failure. Therefore, if no flow is detected, a more conventional
in connecting tubing (244,245); when done properly and calibrated radionuclide or positive contrast study becomes necessary.

Barkovich_Chap08.indd 843 5/6/2011 10:26:25 PM


844 Pediatric Neuroimaging

FIG. 8-38. Failed endoscopic third ventriculocisternostomy.


A. Coronal T2 image shows persistent evidence of hydrocepha-
lus, with large ventricular bodies and temporal horns. B. Sagittal
FIESTA image shows wide opening in the third ventricular floor
(arrows). C. Sagittal T2-weighted image shows no signal void
through the opening.

Shunt Infections loculations are within the brain parenchyma, not in the ventricles; they
result from ischemic or toxic parenchymal damage that causes cavita-
The risk of infection resulting from ventricular shunting has decreased tion of the white matter (see section on “Complications of meningitis”
steadily over the years and is now generally between 1% and 5% in Chapter 11). Patients with multiple loculations are more difficult to
(131,248,249); the rate is higher in infants (249,250). Most shunt infec- treat because each loculation must be drained separately. The use of the
tions develop shortly after insertion; however, as many as 10% develop fiberoptic ventriculoscopy to fenestrate the loculations has aided the
months to years later, usually as a result of systemic infection, peri- treatment of these cases (252). In order to guide the ventriculoscopy, it is
tonitis, trauma, or surgery (252,2002). The major clinical manifesta- essential to image the affected patients in 3 orthogonal planes; ideally a
tion of shunt infection is fever, which is usually intermittent and low 3D image is acquired and multiple contiguous images can be viewed
grade. Anemia, dehydration, hepatosplenomegaly, and stiff neck can in three orthogonal planes at a workstation. Therefore, sonography or
also be presenting signs. Shunt infection may also be manifested by MR is the study of choice; intraoperative sonography or neuronaviga-
swelling and redness over part or all of the shunt tract or by such gen- tion using MR may be a useful adjunct in identifying and localizing
eralized symptoms as peritonitis. The only aspect of shunt infection each loculation within the ventricular system.
that is visualized on imaging studies is that of ventriculitis, which is
seen on contrast-enhanced imaging studies as enlarged lateral ventri-
Subdural Hematoma Formation
cles with an irregular, enhancing ventricular wall. Occasionally, after
severe ventriculitis, multiple fluid loculations develop and distort the Subdural, clinically significant collections are uncommon. They develop
brain, resulting in a confusing image; fortunately, these appear to be in the weeks or months following the treatment of the hydrocephalus.
increasingly uncommon. In the vast majority of these cases, the fluid It is hard to tell if they develop directly as subdural blood collections,

Barkovich_Chap08.indd 844 5/6/2011 10:26:26 PM


Chapter 8 • Hydrocephalus 845

FIG. 8-39. Ventriculoperitoneal shunt disconnection. (Early


infantile hydrocephalus.) A. Axial CT from a routine follow-up
study shows small ventricles, with the interhemispheric fissure
clearly patent. B. Follow-up axial CT due to complaints of new
headaches. Significant bilateral ventricular enlargement with
effacement of the interhemispheric CSF spaces. C. Plain film
evaluation of the shunt. Disconnection of the tube (white arrow)
in the right lateral the neck.

or primarily as simple hygromas which may themselves become however, surgical decompression of post-shunt subdural hemato-
hemorrhagic (Fig. 8-40). It is difficult to understand why the ventricu- mas is very uncommon, as the ventricular decompression results in a
lar collapse would result in a late subdural collection. The pericerebral considerable buffer to increases in intracranial pressure. Because it is
spaces typically become mildly dilated, and one would expect efface- very sensitive to the presence of very small fluid collections, MR will
ment of (noncommunicating) ventricles without brain restoration to be often show small subdural hematomas in children who have recently
accompanied by a simple expansion of the subarachnoid spaces. A pos- been shunted. These small hematomas are almost never of clinical sig-
sibility is that the arachnoid septations have not enough time to adapt nificance. Large subdural hemorrhages which can result in increased
and therefore are pulling the inner dural cellular layers inward, inducing intracranial pressure, are usually seen in children over 3 years of age
the inner dural vessels to bleed. In the case of a hygroma, interstitial after drainage of markedly enlarged ventricles. The use of high-pres-
fluid would accumulate instead of blood; then, like during the evolution sure valves in the shunting of these patients has reduced the incidence
of posttraumatic hygromas (253), neoangiogenesis could take place in of significant subdural hematomas. Hygromas or hemorrhagic hygro-
the inner membrane of the collection and secondarily bleed (Fig. 8-40). mas may develop secondarily within weeks and are usually not con-
Whatever their mechanism, these collections tend to attenuate and cerning, as they evolve and disappear over months (Fig. 8-40).
characteristically disappear spontaneously. Rarely, they may become Using contrast-enhanced MR or CT, chronic subdural hematomas
chronic; they have been suspected of being the origin of the occasionally can be distinguished from postshunt meningeal fibrosis (meningeal
observed fibrous meningeal thickening, but the pathophysiology could callus [254]), in which a thick collagenous scar develops in the sub-
also be that of the chronic intracranial hypotension. dural space, presumably as a reaction to chronic subdural hematoma
When present, the appearance of the subdural collections is similar (254–256) (Fig. 8-42). Whereas the liquid portions of chronic sub-
to that of traumatic subdural hematomas (see Chapter 4) (Fig. 8-41); dural hematomas will not enhance after administration of contrast,

Barkovich_Chap08.indd 845 5/6/2011 10:26:27 PM


846 Pediatric Neuroimaging

FIG. 8-40. Development and evolution of subdural collections


after ventriculoperitoneal shunt placement in an eight month old
infant with Chiari I related hydrocephalus. A. T2-weighted axial
MR 1 month after shunt placement shows persistent ventricular
enlargement with a subdural hygroma over the right convexity.
B. Axial CT 2 months after shunt placement shows that the size
of the ventricles has clearly decreased. Bilateral subdural collec-
tions of intermediate attenuation, higher on the right than on the
left. C. Three months post surgery, the CT shows that the ven-
tricles have enlarged, but both collections are much smaller. Their
attenuation remains increased. D. Four months post surgery, the
CT shows the collection on the left to have completely resorbed.
The collection on the right now has high attenuation. Ventricles
are unchanged. E. Nine months post surgery, the collections have
completely resolved. Ventricles are slightly larger, commensurate
with the amount of subdural fluid resorbed.

Barkovich_Chap08.indd 846 5/6/2011 10:26:28 PM


Chapter 8 • Hydrocephalus 847

FIG. 8-41. Subdural hematomas after shunt placement. A. Axial CT shows bilateral large subdural collections of intermediate
attenuation associated with ventricular compression. B. Axial MR FLAIR in a different patient shows bilateral moderate-sized col-
lections of different signal intensity, associated with ventricular compression.

meningeal fibrosis enhances dramatically, presumably as a result of having the “slit ventricle syndrome” (238,257–261). Some debate exists
vascular granulation tissue intermixed with the collagen bundles in the as to what constellation of clinical and/or pathological findings define
subdural space (256). this syndrome (238,262). It has become clear that there are multiple
different entities lumped together under this name; therefore, affected
Slit Ventricle Syndrome patients may require several different treatments (263).
Di Rocco has divided slit ventricle syndrome into three groups:
Definition (1) patients with inability of the ventricles to expand due to stiff-
Shunted hydrocephalic patients may become symptomatic from shunt ness at the subependymal level (normal intracranial pressure),
failure without evidence of ventricular enlargement on ultrasound, CT, (2) patients with chronic low pressure from overdrainage or siphoning
or MR. Patients who exhibit this phenomenon have been labeled as of CSF through a shunt catheter or chronic CSF leak (low intracranial

FIG. 8-42. Post shunt meningeal fibrosis and calvarial thickening. Constellation of findings in chronic intracranial hypotension.
Child with congenital hydrocephalus. A. At 5 years of age, axial CT shows prominent ventricles, but the pericerebral spaces in the
sulci and interhemispheric fissure are visible. B. At age 7 years, contrast-enhanced CT showed much smaller ventricles. The skull is
markedly thicker than at 5 years. There is a thick layer of intermediate density (white arrows) between the subarachnoid space and the
skull. It could be mistaken for a fluid collection but its irregular, enhancing inner margin suggests meningeal fibrosis.

Barkovich_Chap08.indd 847 5/6/2011 10:26:29 PM


848 Pediatric Neuroimaging

FIG. 8-42. (Continued) C. At the age of 16, axial FLAIR shows marked bone thickening especially the frontal bone. The ventricles
are small and surrounded by a thin band of white signal, presumably periventricular gliosis/fibrosis. A thick layer of tissue (white
arrows) is interposed between the brain and the calvarium and between the occipital lobes, most likely dural fibrosis. D–F. T1 post
contrast axial (D and E) and sagittal (F) images from the same study show that the thick fibrotic calvarial, tentorial, and falcine dura
enhances avidly. The dural venous sinuses are prominent. The cerebellar tonsils are low and compressed. The pituitary gland is
prominent.

pressure), and (3) patients with craniocerebral disproportion second- that can cause headaches without detectable ventricular enlargement.
ary to early suture fusion (high intracranial pressure) (264). Rekate Other than Rekate’s category no. 3, which may show calvarial thicken-
(257) has a different classification of the various disorders composing ing, imaging cannot differentiate these categories. Bruce and Weprin
the slit ventricle syndrome. He divides them into (1) extreme low pres- suggest that patients with symptoms of shunt malfunction in the
sure headaches, probably from siphoning of CSF from the brain by absence of ventricular enlargement require an emergency shunt func-
the shunt; (2) intermittent obstruction of the proximal shunt catheter; tion study, followed by shunt revision with revision of the ventricular
(3) normal volume hydrocephalus (in which the buffering capacity catheter and valve (261).
of the CSF is diminished, see following paragraph); (4) intracranial
hypertension associated with working shunts (probably linked to Pathophysiology
venous hypertension); and (5) headaches in shunted children unre- The syndrome most commonly referred to by the name “slit ventricle
lated to intracranial pressure or shunt function. Thus, shunted patients syndrome” is the result of a combination of decreased intracranial
may experience a number of disorders, with very different treatments, volume; resultant diminished CSF buffering capacity (diminished

Barkovich_Chap08.indd 848 5/6/2011 10:26:30 PM


Chapter 8 • Hydrocephalus 849

ability to compensate for the normal variations in intracranial pres- Diagnosis


sure); diminished brain compliance; and, possibly, intermittent or Affected patients typically present with recurrent, transient symptoms
partial shunt obstruction (237,238). Intracranial volume decreases of shunt failure. Imaging studies, however, will show small ventricu-
because shunting causes an immediate reduction in ventricular, and lar size (Fig. 8-43). Shunt function studies show the shunt system to
hence brain, size. Thus, the brain expansion that maintains the patency be patent, although flow may be reduced. Treatment is shunt revi-
of the cranial sutures is temporarily reversed and the sutures close. By sion or replacement with a valve having higher opening pressure (Fig.
the time the brain grows enough to once again fill the calvarium, the 8-43D). If this fails, bilateral temporal craniotomies are performed
sutures have fused and the calvarium cannot enlarge as rapidly as the in order to expand the volume of intracranial CSF. Theoretically, the
brain. When the overlying calvarium is too small, the CSF spaces are expanded intracranial space is better able to compensate for the tran-
not large enough to provide adequate buffering of normal variations sient changes in intracranial pressure that accompany the variations
in intracranial pressure. Thus, unless the shunt is functioning perfectly, in CSF production.
patients have recurrent episodes of increased intracranial pressure. One finding on imaging studies that may help in the determination
However, because of the small size of the CSF spaces and the compres- of which patients have diminished buffering capacity is that of progres-
sion of the brain, the lateral ventricles do not enlarge significantly dur- sive calvarial thickening. Such thickening may or may not be associ-
ing these episodes of increased pressure. ated with premature synostosis of the cranial sutures; either way, the

FIG. 8-43. Slit ventricles. A. Initial CT scan demonstrating significant hydrocephalus. B. First post surgery CT shows that the
ventricles are still large, with rounded temporal horns but markedly improved. C. Two years later the child complains of headaches.
A new CT demonstrates almost completely effaced ventricles and pericerebral spaces (sulci and cisterns). D. After shunt revision,
headaches have resolved. The CT shows normal-looking ventricles.

Barkovich_Chap08.indd 849 5/6/2011 10:26:31 PM


850 Pediatric Neuroimaging

result is a diminished reservoir of CSF to buffer the normal variations


in intracranial pressure (238).
It is less important for the radiologist to know all the possible
causes of small ventricles in a symptomatic patient than to realize that
the presence of very small ventricles after placement of a ventriculo-
peritoneal shunt is not diagnostic of, or even suggestive of, the “slit
ventricle syndrome.” This syndrome is a clinical, not a morphologic syn-
drome. Small ventricles are not, in the majority of cases, a problem;
they are the desired result of many surgeons after treatment of hydro-
cephalus and, even when extremely small, their only implication in the
vast majority of patients is that the shunt is functioning.

Imaging of Intracranial Hypotension


Beside the early development of subdural collections and the isolated
delayed “slit ventricle”, too much depletion of the ventricles may lead to
the progressive development of the imaging constellation characteris-
tic of intracranial hypotension, findings identical to those observed in
idiopathic cases, iatrogenic cases or cases related to pathologies such as
Marfan syndrome. Intracranial hypotension is important to recognize
as its clinical symptoms are similar to those of intracranial hyperten-
FIG. 8-44. Iatrogenic Chiari I secondary to placement of lumboperito-
sion, while the therapeutic goals are diametrically opposed. The main neal shunt. Midline sagittal T2-weighted image shows elongated cerebellar
symptom is a postural headache exacerbated by standing and relieved tonsils herniating through the foramen magnum.
by reclining. Other nonspecific neurological manifestations may be
observed, including cranial neuropathies and, in severe cases, coma.
When the condition is chronic, head MRI is diagnostic, showing a Other Complications
characteristic constellation of nonspecific abnormalities (265) (Fig.
8-42). The most typical findings are diffuse pachymeningeal (dural) Other complications resulting from ventriculo-peritoneal shunting
enhancement and thickening (PME/PMT) (100%) (Fig. 8-42B, C, E, are much less common. Complications within the abdomen typically
and F), and the venous distension sign (VDS) (100%) which reflects result from the shunt becoming too short while the child grows, and its
venous engorgement resulting from the low intracranial pressure (265) peritoneal end remaining in the same area instead of being displaced
(Fig. 8-42D and F). PMT may be seen without contrast administra- by the bowel displacements; they include ascites, pseudocyst formation,
tion on FLAIR images. VDS may be recognized on T1 imaging without perforation of a viscus or of the abdominal wall, intestinal obstruction
contrast as well, from observing the convexity of the sinus walls (265); (241,269), and catheter degradation (270). Very rarely, patients may
it is the first anomaly to resolve after normalization of the intracra- develop a granulomatous reaction adjacent to the shunt tube either
nial pressure (265). Other features may include subdural hygromas within or near the ventricle (271). These granulomatous lesions appear
or hematomas, effacement of the cisterns, and downward brain her-
niation (which can mimic the Chiari 1 malformation, Fig. 8-42F). An
increased height of the pituitary gland with convex upper border is
commonly observed as well (265) (Fig. 8-42F).
Another presentation of chronic intracranial overdrainage is that of
the complicated acquired Chiari I malformation (266). As mentioned
before, CSF diversion produces a major reduction of intracranial capac-
ity due to calvarial thickening, premature closure of the sutures (pos-
sible induced), and expansion of the paranasal sinuses and mastoid air
cells. The craniocerebral disproportion may be further complicated by
the development of intracranial hypertension with tonsillar herniation
that can only be relieved by a decompressive supratentorial craniotomy
(266). Imaging in this situation shows a small posterior fossa with ton-
sillar herniation through the foramen magnum, small ventricles, mark-
edly reduced pericerebral CSF spaces and prominent thickening of the
calvarium (266). As in all instances of intracranial hypotension, there
may be a pachymeningeal thickening, avidly enhancing after contrast
administration, and prominent widening of the dural venous sinuses.
Clinical features suggest intracranial hypertension with symptoms
pointing to the lower brainstem (266).
This is different from another variety of “acquired Chiari I malfor-
mation” (more accurately, another form of intracranial hypotension)
that is a fairly common complication of lumboperitoneal shunting FIG. 8-45. Interstitial edema and cyst formation secondary to shunt
(267) (Fig. 8-44). It is assumed to result from a CSF pressure differen- malfunction. A. Axial FLAIR image in a shunted 12-year-old hydrocepha-
tial between the cranium and the spine, but this explanation has been lic child shows the normal hypointensity of the ventriculostomy catheter
challenged (268). (arrow) coursing through the cerebrum.

Barkovich_Chap08.indd 850 5/6/2011 10:26:32 PM


Chapter 8 • Hydrocephalus 851

FIG. 8-45. (Continued) B. Two years later, an area of edema surrounds the shunt. The central focus of low signal (arrow) likely
represents a cyst forming around the shunt. The child still is asymptomatic. C. At the age of 17, both the edematous area and the cyst
have increased in size, leading to a shunt revision.

as irregular, contrast-enhancing masses along the course of the shunt 11. Miyan JA, Nabiyouni M, Zendah M. Development of the brain: a vital role
tube. Calcification may be noted within the mass. for cerebrospinal fluid. Can J Physiol Pharmacol 2003;81:317–328.
One final appearance of shunt malfunction should be mentioned. 12. Cutler RW, Page L, Galicich J, Watters GV. Formation and absorption of
Rarely, when the ventricular end of the shunt becomes partially cerebrospinal fluid in man. Brain 1968;91:707–720.
13. McComb JG. Recent research into the nature of cerebrospinal fluid forma-
occluded, CSF may track along this shunt and enter the interstices of
tion and absorption. J Neurosurg 1983;59:369–383.
the centrum semiovale. On these occasions, the CT and MR appear- 14. McComb JG. Cerebrospinal fluid physiology of the developing fetus. Am J
ance is that of edema (low density in the white matter on CT, pro- Neuroradiol 1992;13:595–599.
longed T1 and T2 relaxation time in the white matter on MR) in the 15. Artru AA. Spinal cerebrospinal fluid chemistry and physiology. In: Yaksh
area around the shunt. Eventually, an actual cyst may form in the white TL, ed. Spinal Drug Delivery. Amsterdam: Elsevier, 1999:177–238.
matter surrounding the ventricular catheter (Fig. 8-45). It should be 16. Bulat M, Lupret V, Orehkovic D, Klarica M. Transventricular and transpial
recognized that the presence of edema in these patients is not the result absorption of cerebrospinal fluid into cerebral microvessels. Coll Antropol
of infection or tumor but is a sign of shunt malfunction. The edema 2008;32(Suppl 41):43–50.
and cyst will resolve after shunt revision. 17. Olivero WC, Rekate HL, Chizek JH, et al. Relationship between intracranial
and sagittal sinus pressure in normal and hydrocephalic dogs. Pediatr Neu-
rosci 1988;14:196–201.
References 18. McCormick JM, Yamada K, Rekate HL, Miyake H. Time course of intraven-
tricular pressure change in a canine model of hydorcephalus: its relation-
1. Bartelmez GW, Dekaban AS. The early development of the human brain. ship to sagittal sinus elastance. Pediatr Neurosurg 1992;18:127–133.
Contrib Embryol Carnegie Instn 1962;37:13–32. 19. Worthington, Jr W, Cathcart RS III. Ependymal cilia: distribution and
2. ten Donkelaar HJ, van der Vliet T. Overview of the development of the activity in the adult human brain. Science 1963;139:221–222.
human brain and spinal cord. In: ten Donkelaar HJ, Lammens M, Hori S, 20. Cathcart III RS, Worthington, Jr W. Ciliary movement in the rat cerebral
eds. Clinical Neuroembryology. Berlin-Heidelberg: Springer, 2006:1–46. ventricles: clearing action and direction of currents. J Neuropathol Exp
3. Johanson C, Jones H. Promising vistas in hydrocephalus and cerebrospinal Neurol 1964;23:609–618.
fluid research. Trends Neurosci 2001;24:631–632. 21. Welch K, Friedman V. The cerebrospinal fluid valves. Brain 1960;83:
4. Brodbelt A, Stoodley M. CSF pathways: a review. Br J Neurosurg 454–469.
2007;21:510–520. 22. Bosanovic-Sosic R, Mollanji R, Johnston MG. Spinal and cranial contribu-
5. McLone DG. The subarachnoid space: a review. Childs Brain 1980;6: tion to total cerebrospinal fluid transport. Am J Physiol Regulatory Integra-
113–130. tive Comp Physiol 2001;281:R909–R916.
6. Osaka K, Handa R, Matsumoto S, Yasuda M. Development of the cerebro- 23. Edsbagge M, Tisell M, Jacobsson L, Wikkelso C. Spinal CSF absorption in
spinal fluid pathway in the normal and abnormal human embryos. Childs healthy individuals. Am J Physiol Regul Integr Comp Physiol 2004;287:R1450–
Brain 1980;6:26–38. R1455.
7. Brocklenhurst G. The development of the human cerebrospinal fluid path- 24. Zenker W, Bankoul S, Braun J. Morphological indications for considerable
way with particular reference to the roof of the fourth ventricle. J Anat diffuse reabsorption of cerebrospinal fluid in spinal meninges particularly
1969;55:40–48. in the areas of meningeal funnels. An electronmicroscopical study includ-
8. Dooling EC, Chi Je G, Gilles FH. Ependymal changes in the human fetal ing tracing experiments in rats. Anat Embryol (Berl) 1994;189:243–258.
brain. Ann Neurol 1977;1:535. 25. Greitz D. Radiological assessment of hydrocephalus: new theories and
9. Kuban KCK, Gilles FH. Human telencephalic angiogenesis. Ann Neurol implication for therapy. Neurosurg Rev 2004;27:145–165.
1985;17:539–548. 26. Greitz D, Hannerz J. A proposed model of cerebrospinal fluid circula-
10. Norman MG, O’Kusky JR. The growth and development of microvasculature tion: observations with radionuclide cisternography. Am J Neuroradiol
in the human cerebral cortex. J Neuropathol Exp Neurol 1986;45:222–232. 1996;17:431–438.

Barkovich_Chap08.indd 851 5/6/2011 10:26:32 PM


852 Pediatric Neuroimaging

27. Di Rocco C, Pettorossi VE, Caldarelli M, Mancinelli R, Velardi F. 51. Del Bigio MR, Wilson MJ, Enno T. Chronic hydrocephalus in rats and
Communicating hydrocephalus induced by mechanically increased ampli- humans: white matter loss and behavior changes. Ann Neurol 2003;53:
tude of the intraventricular cerebrospinal fluid pressure: experimental 337–346.
studies. Exp Neurol 1978;59:40–52. 52. Shirane R, Sato S, Sato K, et al. Cerebral blood flow and oxygen metabolism
28. Greitz D, Greitz T, Hindmarsh T. A new view on the CSF circulation with in infants with hydrocephalus. Childs Nerv Syst 1992;8:118–123.
the potential for pharmacological treatment of childhood hydrocephalus. 53. Hill A, Volpe JJ. Decrease in pulsatile flow in the anterior cerebral arteries in
Acta Pediatr 1997;1997:125–132. infantile hydrocephalus. Pediatrics 1982;69:4–10.
29. James AE, Strecker EP, Sperber E, et al. An alternative pathway of cere- 54. Bannister CM, Chapman SA. Ventricular ependyma of normal and hydro-
brospinal fluid absorption in communicating hydrocephalus. Radiology cephalic subjects: a scanning electron microscopic study. Dev Med Child
1974;111:143–146. Neurol 1980;22:725–731.
30. Greitz D. Thesis: Cerebrospinal fluid circulation and associated intracranial 55. Greitz D, Greitz T. The pathogenesis and hemodynamics of hydrocephalus.
dynamics. A radiologic investigation using MR imaging and radionuclide cis- Int J Neuroradiol 1997;3:367–375.
ternography. [Ph. D.], Karolinska Institute, 1993. 56. Greitz D, Franck A, Nordell B. On the pulsatile nature of intracranial and
31. Greitz D, Wirestam R, Franck A, Nordell B, Thomsen C, Ståhlberg F. spinal CSF-circulation demonstrated by MR imaging. Acta Radiologica
Pulsatile brain movement and associated hydrodynamics studied by mag- 1993;34:1–8.
netic resonance imaging. The Monroe-Kellie doctrine revisited. Neuroradi- 57. Du Boulay GH. Pulsatile movements in the CSF pathways. Br J Radiol
ology 1992;34:370–380. 1966;39:255–262.
32. Johnston M, Zakharov A, Papaiconomou C, Salmasi G, Armstrong D. Evi- 58. Huang T-Y, Chung H-W, Chen M-Y, et al. Supratentorial cerebrospinal fluid
dence of connections between cerebrospinal fluid and nasal lymphatic production rate in healthy adults: quantification with two-dimensional
vessels in humans, non-human primates and other mammalian species. cine phase-contrast MR imaging with high temporal and spatial resolu-
Cerebrospinal Fluid Res 2004;1:2. tion. Radiology 2004;233:603–608.
33. Papaiconomou C, Zakharov A, Azizi N, Djenic J, Johnston M. Reassessment 59. de Marco G, Idy-Peretti I, Didon-Poncelet A, Baledent O, Onen F, Feugeas
of the pathways responsible for cerebrospinal fluid absorption in the neo- MC. Intracranial fluid dynamics in normal and hydrocephalic states: sys-
nate. Childs Nerv Syst 2004;20:29–36. tems analysis with phase-contrast magnetic resonance imaging. J Comput
34. Silver I, Li B, Szalai J, Johnston M. Relationship between intracranial pres- Assist Tomogr 2004;28:247–254.
sure and cervical lymphatic pressure and flow rates in sheep. Am J Physiol 60. Luetmer PH, Huston J, Friedman J, et al. Measurement of cerebrospinal
1999;277:R1712–R1717. fluid flow at the cerebral aqueduct by use of phase-contrast magnetic reso-
35. Bradbury MWB, Cole DF. The role of the lymphatic system in drainage of nance imaging: technique validation and utility in diagnosing idiopathic
cerebrospinal fluid and aqueous humor. J Physiol 1980;299:353–365. normal pressure hydrocephalus. Neurosurgery 2002;50:534–543.
36. McComb JG, Hyman S. Lymphatic drainage of cerebrospinal fluid in the 61. Mase M, Yamada K, Banno T, Miyachi T, Ohara S, Matsumoto T. Quantita-
primate. In: Johansson BB, Owman C, Widner H, eds. Pathophysiology of tive analysis of CSF flow dynamics using MRI in normal pressure hydro-
the Blood-Brain Barrier. Amsterdam: Elsevier, 1990:421–438. cephalus. Acta Neurochir Suppl 1998;71:350–353.
37. McComb JG, Hyman S, Weiss MH. Lymphatic drainage of cerebrospinal 62. Tsunoda A, Mitsuoka H, Bandai H, Endo T, Arai H, Sato K. Intracranial
fluid in the cat. In: Shapiro K, Marmarou A, Portnoy H, eds. Hydrocephalus. cerebrospinal fluid measurement studies in suspected idiopathic normal
New York: Raven, 1984:83–98. pressure hydrocephalus, secondary normal pressure hydrocephalus, and
38. Ludemann W, Berens Von Rautenfeld D, Samii M, Brinker T. Ultrastructure brain atrophy. J Neurol Neurosurg Psychiatry 2002;73:552–555.
of the cerebrospinal fluid outflow along the optic nerve into the lymphatic 63. Mori K. Current concept of hydrocephalus: evolutions of new classifica-
system. Childs Nerv Syst 2004. tions. Childs Nerv Syst 1995;11:523–532.
39. Fox RJ, Walji AH, Mielke B, Petruk KC, Aronyk KE. Anatomic details of 64. Hirsch J-F. Consensus: long term outcome in hydrocephalus. Childs Nerv
intradural channels in the parasagittal dura: a possible pathway for flow of Syst 1994;10:64–69.
cerebrospinal fluid [Report]. Neurosurgery 1996;39:84–90. 65. Levine D, Trop I, Mehta TS, Barnes PD. MR imaging appearance of fetal
40. Johnston M, Armstrong D, Koh L. Possible role of the cavernous sinus veins cerebral ventricular morphology. Radiology 2002;223:652–660.
in cerebrospinal fluid absorption. Cerebrospinal Fluid Res 2007;4:3. 66. Patel MD, Filly RA, Hersh D, Goldstein RD. Isolated mild fetal cere-
41. Zervas NT, Lisczak TM, Mayberg MR, et al. Cerebrospinal fluid may nour- bral ventriculomegaly: clinical course and outcome. Radiology 1994;192:
ish cerebral vessels through pathways in the adventitia that may be analo- 759–764.
gous to systemic vasa vasorum. J Neurosurg 1982;56:475–481. 67. Simon EM, Goldstein RB, Coakley FV, et al. Fast MR imaging of fetal CNS
42. Oi S, Di Rocco C. Proposal of “evolution theory in cerebrospinal fluid anomalies in utero. Am J Neuroradiol 2000;21:1688–1698.
dynamics” and minor pathway hydrocephalus in developing immature 68. Rosseau GL, McCullough DC, Joseph AL. Current prognosis in fetal ven-
brain. Childs Nerv Syst 2006;22:662-669. triculomegaly. J Neurosurg 1992;77:551–555.
43. Davis LE. A physio-pathological study of the choroid plexus with the report 69. Garel C, Luton D, Oury JF, Gressens P. Ventricular dilatations. Childs Nerv
of a case of villous hypertrophy. J Med Res 1924;44:521–534. Syst 2003;19:517–523.
44. Hirano H, Hirahara K, Asakura T, et al. Hydrocephalus due to villous 70. Cavalheiro S, Moron AF, Zymberg ST, SDastoli P. Fetal hydrocephalus –
hypertrophy of the choriod plexus in the lateral ventricles. J Neurosurg prenatal treatment. Childs Nerv Syst 2003;2003:7–8.
1994;80:321–323. 71. Laurence KM. The pathology of hydrocephalus. Ann R Coll Surg Engl
45. Welch K, Strand R, Bresnan M, et al. Congenital hydrocephalus due to 1959;24:388.
villous hypertrophy of the telencephalic choroid plexuses. J Neurosurg 72. Warkany J, Lemire RJ, Cohen MM, Jr. Mental Retardation and Congenital
1983;59:172–175. Malformations of the Central Nervous System. Chicago: Yearbook, 1981.
46. Friede RL. Developmental Neuropathology, 2nd ed. Berlin: Springer-Verlag, 73. Yakovlev PI. Paraplegias of hydrocephalics. Am J Ment Defic 1947;51:
1989. 561–577.
47. Milhorat TH. Classifications of the cerebral edemas with reference 74. Muñoz A, Hinojosa J, Esparza J. Cisternography and ventriculography
to hydrocephalus and pseudotumor cerebri. Childs Nerv Syst 1992;8: gadopentate dimeglumine-enhanced MR imaging in pediatric patients:
301–306. preliminary report. Am J Neuroradiol 2007;28:889–894.
48. Milhorat TH. Hydrocephaly. In: Myrianthopoulos NC, ed. Malformations, 75. Iskandar BJ, Sansone JM, Medow J, Rowley HA. The use of quick-brain
vol 50. New York: Elsevier, 1987:285–300. magnetic resonance imaging in the evaluation of shunt-treated hydroceph-
49. Weller RO, Schulman K. Infantile hydrocephalus, clinical, histological, and alus. J Neurosurg: Pediatrics. 2004;101:147–151.
ultrastructural study of brain damage. J Neurosurg 1972;36:255–260. 76. Bradley WG, Jr, Kortman KE, Burgoyne B, Eng D. Flowing cerebrospi-
50. Del Bigio MR. Neuropathological changes caused by hydrocephalus. Acta nal fluid in normal and hydrocephalic states: appearance on MR images.
Neuropathol 1993;85:573–585. Radiology 1986;159:611–616.

Barkovich_Chap08.indd 852 5/6/2011 10:26:33 PM


Chapter 8 • Hydrocephalus 853

77. Sherman JL, Citrin CM, Gangarosa RE, Bowen BJ. The MR appearance 104. Barkovich AJ, Newton TH. Aqudeuctal stenosis: evidence of a broad
of CSF flow in patients with ventriculomegaly. Am J Neuroradiol 1986;7: spectrum of tectal distortion. Am J Neuroradiol 1989;10:471–475.
1025–1031. 105. El Gammal T, Allen MB, Jr, Brooks BS, Mark EK. MR evaluation of hydro-
78. Sherman JL, Citrin CM. Magnetic resonance of normal CSF flow. Am J cephalus. Am J Neuroradiol 1987;8:591–597.
Neuroradiol 1986;7:3–6. 106. Rovira A, Capellades J, Grivé E, et al. Spontaneouls ventriculostomay:
79. Atlas SW, Mark AS, Fram EK. Aqueductal stenosis: evaluation with gradi- report of three cases revealed by flow-sensitive phase-contrast cine MR
ent echo rapid MR imaging. Radiology 1988;169:449–453. imaging. Am J Neuroradiol 1999;20:1647–1652.
80. Enzmann DR, Pelc NJ. Normal flow patterns of intracranial and spi- 107. Borit A, Sidman RL. New mutant mouse with communicating hydro-
nal cerebrospinal fluid defined with phase-contrast cine MR imaging. cephalus and secondary aqueductal stenosis. Acta Neuropathol 1972;21:
Radiology 1991;178:467–474. 316–331.
81. Levy LM, Di Chiro G. MR phase imaging and cerebrospinal fluid flow in 108. Williams B. Is aqueductal stenosis the result of hydrocephalus? Brain
the head and spine. Neuroradiology 1990;32:399–406. 1973;96:399–412.
82. Nitz WR, Bradley WG, Jr, Watanabe AS, et al. Flow dynamics of cerebro- 109. Segev Y, Metser U, Beni-Adani L, Elran C, Reider-Groswasser I-I, Constan-
spinal fluid: assessment with phase-contrast velocity MR imaging per- tini S. Morphometric study of the midsagittal MR imaging plane in cases
formed with retrospective cardiac gating. Radiology 1992;183:395–405. of hydrocephalus and atrophy and in normal brains. Am J Neuroradiol
83. Taylor GA, Madsen JR. Neonatal hydrocephalus: hemodynamic response 2001;22:1674–1679.
to fontanelle compression–correlation with intracranial pressure and 110. Fitz CR, Harwood-Nash DC. Computed tomography of hydrocephalus.
need for shunt placement. Radiology 1996;201:685–689. Comput Tomogr 1978;2:91–108.
84. Filly RA, Goldstein RB. Fetal ventricular atrium: 4th down and 10 mm to 111. Kleinman PK, Zito JL, Davidson RI, Raptopoulos V. The subarachnoid
go. Radiology 1994;193:315–317. spaces in children: normal variations in size. Radiology 1983;147:455–457.
85. Garel C, Chantrel E, Sebag G, Brisse H, Elmalah M, Hussan M. Le devel- 112. Gomori JM, Steiner I, Melamed E, Cooper G. The assessment of changes
oppment du cerveau foetal: atlas IRM et biometrie. Editions Sanramps in brain volume using combined linear measurements. A CT scan study.
Medical, Montpelier, France, 2000. Neuroradiology 1984;26:21–24.
86. Girard N, Raybaud C, Poncet M. In vivo MR study of brain maturation in 113. Hahn FJY, Rim K. Frontal ventricular dimensions on normal computed
normal fetuses. Am J Neuroradiol 1995;16:407–413. tomography. Am J Roentgenol 1976;126:593–396.
87. Gaglioti P, Danelon D, Bontempo S, Mombro M, Cardaropoli S, Todros T. 114. Haug G. Age and sex dependence of the size of normal ventricles on com-
Fetal cerebral ventriculomegaly: outcome in 176 cases. Ultrasound Obstet puted tomography. Neuroradiology 1977;14:201–204.
Gynecol 2005;25:372–377. 115. Evans WA. An encephalographic ratio for estimating ventricular enlarge-
88. Patel MD, Goldstein RB, Tung S, Filly RA. Fetal cerebral ventricular atrium: ment and cerebral atrophy. Arch Neurol Psych 1942;47:931–937.
difference in size according to sex. Radiology 1995;194:713–715. 116. Gooskens RHJM, Gielen CCAM, Hanlo PW, Faber JAJ, Willemse J. Intrac-
89. Vergani P, Locatelli A, Strobelt N, et al. Clinical outcome of mild fetal ven- ranial spaces in childhood macrocephaly: comparison of length measure-
triculomegaly. Am J Obstet Gynecol 1998;178:218–222. ments and volume calculations. Dev Med Child Neurol 1988;30:509–519.
90. Garel C. MRI of the Fetal Brain. Normal Development and Cerebral Pathol- 117. da Silva MC, Michowicz S, Drake JM, Chumas PD, Tuor UI. Reduced local
ogies. Berlin: Springer, 2004. cerebral blood flow in periventricular white matter in experimental neo-
91. Girard N, Gire C, Sigaudy S, et al. MR imaging of acquired fetal brain natal hydrocephalus-restoration with CSF shunting. J Cereb Blood Flow
disorders. Childs Nerv Syst 2003;19:490–500. Metab 1995;15:1057–1065.
92. Griffiths PD, Reeves RJ, Morris JE, et al. A prospective study of fetuses 118. Leliefeld PH, Gooskens RHJM, Vincken KL, et al. Magnetic resonance
with isolated ventriculomegaly investigated by antenatal sonography and imaging for quantitative flow measurement in infants with hydrocepha-
in utero MR imaging. Am J Neuroradiol 2010;31. lus: a prospective study. J Neurosurg: Pediatrics. 2008;2:163–170.
93. Glenn O. MR imaging of the fetal brain. Pediatr Radiol 2010;40:68–81. 119. Braun KPJ, Dijkhuizen RM, de Graaf RA, et al. Cerebral ischemia and
94. Beeghly M, Ware J, Soul J, et al. Neurodevelopmental outcome of fetuses white matter edema in experimental hydrocephalus: a combined in vivo
referred for ventriculomegaly. Ultrasound Obstet Gynecol 2010;35: MRI and MRS study. Brain Res 1997;757:295–298.
405–416. 120. Braun KPJ, Gooskens RHJM, Vandertop WP, Tulleken CAF, van der Grond
95. Baker LL, Barkovich AJ. The large temporal horn: MR analysis in devel- J. 1-H magnetic resonance spectroscopy in human hydrocephalus. J Magn
opmental brain anomalies versus hydrocephalus. Am J Neuroradiol Reson Imaging 2003;17:291–299.
1992;13:115–122. 121. Assaf Y, Ben-Sira L, Constantini S, Chang LC, Beni-Adani L. Diffusion
96. Heinz ER, Ward A, Drayer BP, DuBois PJ. Distinction between obstructive tensor imaging in hydrocephalus: initial experience. Am J Neuroradiol
and atrophic dilatation of ventricles in children. J Comput Assist Tomogr 2006;27:1717–1724.
1980;4:320–325. 122. Ding Y, McAllister JP, Yao B, Yan N, Canady AI. Neuron tolerance during
97. Sjaastad O, Skalpe IO, Engeset A. The width of the temporal horn and the hydrocephalus. Neuroscience 2001;106:659–667.
differential diagnosis between pressure hydrocephalus and hydrocephalus 123. Humphreys P, Muzumdar DP, Sly LE, Michaud J. Focal cerebral mantle
ex vacuo. Neurology 1969;19:1087–1093. disruption in fetal hydrocephalus. Pediatr Neurol 2007;36:236–243.
98. LeMay M, Hochberg FH. Ventricular differences between hydrostatic 124. Milhorat TH, Hammock MK, Davis DA, Fenstermacher JD. Choroid
hydrocephalus and hydrocephalus ex vacuo by computed tomography. plexus papilloma: I. Proof of cerebrospinal fluid overproduction. Childs
Neuroradiology 1979;17:191–195. Brain 1977;5:273–277.
99. Childe AE, McNaughton FL. Diverticula of the lateral ventricles extending 125. Eisenberg HM, McComb JG, Lorenzo AV. Cerebrospinal fluid overpro-
in the posterior fossa. Arch Neurol Psychiatry 1942;47:768–778. duction and hydrocephalus associated with choroid plexus papilloma.
100. Jabaudon D, Charest D, Porchet F. Pathogenesis and diagnostic pitfalls of J Neurosurg 1974;40:381–385.
ventricular diverticula: case report and review of the literature. Neurosur- 126. Cinalli G, Sainte-Rose C, Lellouch-Tubiana A, Sebag G, Renier D, Pierre-
gery 2003;52:209–212; discussion 212. Kahn A. Hydrocephalus associated with intramedullary low-grade
101. Naidich TP, McLone DG, Hahn YS, Hanaway J. Atrial diverticula in severe glioma. Illustrative cases and review of the literature. J Neurosurg 1995;83:
hydrocephalus. Am J Neuroradiol 1982;3:257–266. 480–485.
102. Osuka S, Takano S, Enomoto T, Ishikawa E, Tsuboi K, Matsumura A. 127. Turkewitsch N. Die Entwicklung des Aquaeductus Cerebri des Menschen.
Endoscopic observation of pathophysiology of ventricular diverticulum. Morphologisches Jahrbuch 1935;76:421–477.
Childs Nerv Syst 2007;23:897–900. 128. Lapras C, Tommasi M, Bret P. Stenosis of the aqueduct of Sylvius. In:
103. Kim CS, Bennett DR, Roberts TS. Primary amenorrhea secondary to Myrianthopoulos NC, ed. Malformations, vol 50. New York: Elsevier,
noncommunicating hydrocephalus. Neurology 1969;19:533–537. 1987:301–322.

Barkovich_Chap08.indd 853 5/6/2011 10:26:33 PM


854 Pediatric Neuroimaging

129. Sarnat HB. Ependymal reactions to injury. A review. J Neuropathol Exp 156. Tulipan N, Sutton LN, Bruner JP, Cohen BM, Johnson M, Adzick NS. The
Neurol 1995;54:1–15. effect of intrauterine myelomeningocele repair on the incidence of shunt-
130. Castro-Gago M, Alonso A, Eiris-Punal J. Autosomal recessive hydrocepha- dependent hydrocephalus. Pediatr Neurosurg 2003;38:27–33.
lus with aqueduuctal stenosis. Childs Nerv Syst 1996;12:188–191. 157. Hirsch JF, Pierre-Kahn A, Renier D, Sainte-Rose C, Hoppe-Hirsch E.
131. Gabriel RS, McComb JG. Malformations of the central nervous system. In: The Dandy-Walker malformation: a review of 40 cases. J Neurosurg
Menkes JH, ed. Textbook of Child Neurology. Philadelphia: Lea and Febiger, 1984;61:515–522.
1985:234–253. 158. Raimondi AJ, Sato K, Shimoji T. The Dandy-Walker Syndrome. Basel: S.
132. Woollam DHM, Millen JW. Anatomical considerations in the pathology Karger, 1984.
of stenosis of the cerebral aqueduct. Brain 1953;76:104–112. 159. Scotti G, Musgrave MA, Fitz CR, Harwood-Nash DC. The isolated fourth
133. Russell DS. Observations on the Pathology of Hydrocephalus. London: His ventricle in children: CT and clinical review of 16 cases. Am J Roentgenol
Majesty’s Stationery Office, 1949. 1980;135:1233–1238.
134. Raimondi AJ, Clark SJ, McLone DG. Pathogenesis of aqueductal occlusion 160. James HE. Spectrum of the syndrome of the isolated fourth ventricle in
in congenital murine hydrocephalus. J Neurosurg 1976;45:66–77. posthemorrhagic hydrocephalus of the premature infant. Pediatr Neuro-
135. Boydston WR, Sanford RA, Mauhlbauer MS, et al. Gliomas of the tectum surg 1990–1991;16:305–308.
and periaqueductal region of the mesencephalon. Pediatr Neurosurg 161. Hall TR, Choi A, Schellinger D, Grant EG. Isolation of the fourth ventricle
1991–92;17:234–238. causing transtentorial herniation: neurosonographic findings in prema-
136. Turnbull I, Drake C. Membranous occlusion of the aqueduct of Sylvius. ture infants. Am J Roentgenol 1992;159:811–815.
J Neurosurg 1966;24:24–33. 162. Owler BK, Jacobson EE, Johnston IH. Lateral recess cysts in two cases of
137. Schroeder HWS, Gaab MR. Endoscopic aqueductoplasty: technique and isolated fourth ventricle. Childs Nerv Syst 2001;17:363–365.
results. Neurosurgery 1999;45:508–518. 163. Hamada H, Hayashi N, Kurimoto M, Endo S. Endoscopic aqueductal
138. Fritsch MJ, Kienke S, Mehdorn HM. Endoscopic aqueductoplasty: stent or stenting via the fourth ventricle under navigating system guidance: tech-
not to stent? Childs Nerv Syst 2003. nical note. Neurosurgery 2005;56:E206.
139. Er&scedil;ahin Y. Endoscopic aqueductoplasty. Childs Nerv Syst 164. Gomori JM, Grossman RI, Goldberg HI, Zimmerman RA, Bilaniuk
2007;23:143–150. LT. Intracranial hematomas: imaging by high field MR. Radiology
140. Bickers DS, Adams RD. Hereditary stenosis of the aqueduct of Sylvius as a 1985;157:87–93.
cause of congenital hydrocephalus. Brain 1949;72:246–262. 165. Gomori JM, Grossman RI, Goldberg HI, et al. High field spin echo MR
141. Willems PJ, Dijkstra I, Van der Auwera BJ, et al. Assignment of X-linked imaging of superficial and subependymal siderosis secondary to neonatal
hydrocephalus to Xq28 by linkage analysis. Genomics 1990;8:367–370. intraventricular hemorrhage. Neuroradiology 1987;29:339–342.
142. Willems PJ, Vits L, Raeymaekers P, et al. Further localization of X-linked 166. Unger EC, Cohen MS, Brown TR. Gradient echo imaging of hemorrhage
hydrocephalus in the chromosomal region Xq28. Am J Hum Genet at 1.5T. Magn Reson Imaging 1989;7:163–167.
1992;51:307–315. 167. Ashwal S, Holshouser BA, Tong K. Use of advanced neuroimaging tech-
143. Holmes LB, Nash A, ZuRhein GM, et al. X-linked aqueductal steno- niques in the evaluation of pediatric traumatic brain injury. Dev Neurosci
sis: clinical and neuropathological findings in two families. Pediatrics 2006;28:309–326.
1973;51:697–704. 168. Rippe DJ, Boyko OB, Friedman HS, et al. Gd-DTPA-enhanced MR imag-
144. Viseskul C, Gilbert EF, Opitz JM. X-linked hydrocephalus. Further obser- ing of leptomeningeal spread of primary intracranial CNS tumors in
vations. Z Kinderheilk 1975;119:111–121. children. Am J Neuroradiol 1990;11:329–332.
145. Fransen E, Schrander-Stumpel C, Vits L, Coucke P, Van Camp G, Willems 169. Mathews VP, Kuharit MA, Edwards MK, D’Amour PG, Azzarelli B,
PJ. X-linked hydrocephalus and MASA syndrome present in one family Dreesen RG. Gadolinium enhanced MR imaging of experimental bacte-
are due to a single missense mutation in exon 28 of the L1CAM gene. Hum rial meningitis: evaluation and comparison with CT. Am J Neuroradiol
Mol Genet 1994;3:2255–2256. 1988;9:1045–1050.
146. Basel-Vanagaite L, Straussberg R, Friez M, et al. Expanding the phenotypic 170. Matthews VP, Broome DR, Smith RR, Bognanno JR, Einhorn LH, Edwards
spectrum of L1CAM-associated disease. Clin Genet 2006;69:414–419. MK. Neuroimaging of disseminated germ cell tumors. Am J Neuroradiol
147. Fransen E, Lemmon V, Van Camp G, Vits L, Coucke P, Willems PJ. CRASH 1990;11:319–324.
syndrome: clinical spectrum of corpus callosum hypoplasia, retardation, 171. Bagley C. Blood in the cerebrospinal fluid. Resultant functional and
adducted thumbs, spastic paraparesis and hydrocephalus due to mutation organic alterations in the central nervous system. A. Experimental data.
in one single gene, L1. Eur J Hum Genet 1995;3:273–284. Arch Surg 1928;17:18–38.
148. Yamasaki M, Thompson P, Lemmon V. CRASH syndrome: mutations in 172. Hughes CP, Gado M. Pathology of hydrocephalus and brain atrophy. In:
L1CAM correlate with severity of the disease. Neuropediatrics 1997;28: Newton TH, Potts DG, eds. Radiology of the Skull and Brain. Vol III. Anat-
175–178. omy and Pathology. St. Louis: C.V. Mosby, 1977:Chapter 97.
149. Weller S, Gartner J. Genetic and clinical spects of X-linked hydrocephalus 173. Linscott LL, Osborn AG, Blaser S, et al. Pilomyxoid astrocytoma: expand-
(L1 disease): Mutations in the L1CAM gene. Hum Mutat 2001;18:1–12. ing the imaging spectrum. Am J Neuroradiol 2008;29:1861–1866.
150. Willems PJ, Brouwer OF, Dijkstra I, Wilmink J. X-linked hydrocephalus. 174. Little JR, Dale AJD, Okazaki H. Meningeal carcinomatosis: clinical mani-
Am J Med Genet 1987;27:921–928. festations. Arch Neurol 1974;30:138–143.
151. Varadi V, Csecsei K, Szeifert GT, Toth Z, Papp Z. Prenatal diagnosis 175. Kochi M, Mihara Y, Takada A, et al. MRI of subarachnoid dissemination of
of X linked hydrocephalus without aqueductal stenosis. J Med Genet medulloblastoma. Neuroradiology 1991;33:264–267.
1987;24:207–209. 176. Rosman NP, Shands KN. Hydrocephalus caused by increased intracranial
152. Wong EV, Kenwrick S, Willems P, Lemmon V. Mutations in the cell adhe- venous pressure: a clinicopathological study. Ann Neurol 1978;3:445–450.
sion molecule L1 cause mental retardation. Trends Neurosci 1995;18: 177. Kalbag RM, Woolf AL. Cerebral Venous Thrombosis. London: Oxford,
168–172. 1967.
153. Yamasaki M, Arita N, Hiraga S, et al. A clinical and neuroradiological 178. Sainte-Rose C, LaCombe J, Peirre-Kahn A, et al. Intracranial venous sinus
study of X-linked hydrocephalus in Japan. J Neurosurg 1995;83:50–55. hypertension: cause or consequence of hydrocephalus in infants? J Neuro-
154. Jellinger G. Anatomopathology of non-tumoral aqueductal stenosis. surg 1984;60:727–736.
J Neurosurg Sci 1986;30:1–16. 179. Epstein F, Hochwald GM, Ransohoff J. Neonatal hydrocephalus treated by
155. Bouchard S, Davey MG, Rintoul NE, Walsh DS, Rorke LB, Adzick NS. compressive head wrapping. Lancet 1973;1:634.
Correction of hindbrain herniation and anatomy of the vermis after 180. Hochwald GM, Epstein F, Malhan C, et al. The role of skull and dural
in utero repair of myelomeningocele in sheep. J Pediatr Surg 2003;38: in experimental feline hydrocephalus. Dev Med Child Neurol 1972;14
451–458. (Suppl 27): 65–69.

Barkovich_Chap08.indd 854 5/6/2011 10:26:33 PM


Chapter 8 • Hydrocephalus 855

181. Shapiro K, Fried A, Takei F, Kohn I. Effect of the skull and dura on neural 208. Barkovich AJ, Edwards MSB. Applications of neuroimaging in
axis pressure-volume relationships and CSF hydrodynamics. J Neurosurg hydrocephalus. Pediatr Neurosurg 1992;18:65–83.
1985;63:76–81. 209. Maytal J, Alvarez LA, Elkin CM, Shinnar S. External hydrocephalus: radio-
182. Shapiro K, Fried A, Maramou A. Biomechanical and hydrodynamic char- logic spectrum and differentiation from cerebral atrophy. Am J Neurora-
acterization of the hydrocephalic infant. J Neurosurg 1985;63:69–75. diol 1987;8:271–278.
183. Hecht JT, Butler IJ. Neurologic morbidity associated with achondoplasia. 210. Wilms G, Vanderschueren G, Demaerel PH, et al. CT and MR in infants
J Child Neurol 1990;5:84–97. with pericerebral collections and macrocephaly: benign enlargement of
184. Kao SCS, Waziri MH, Smith WL, Sato Y, Yuh WTC, Franken EA, Jr. MR the subarachnoid spaces versus subdural collections. Am J Neuroradiol
imaging of the craniovertebral junction, cranium, and brain in children 1993;14:855–860.
with achondoplasia. Am J Roentgenol 1989;153:565–569. 211. Chen C-Y, Chou T-Y, Zimmerman RA, Lee C-C, Chen F-H, Faro SH.
185. Pappas CTE, Rekate HL. Cervicomedullary junction decompression in a Pericerebral fluid collection: differentiation of enlarged subarachnoid
case of Marshall-Smith syndrome. J Neurosurg 1991;75:317–319. spaces from subdural collections with color Doppler US. Radiology
186. Moss ML. The pathogenesis of artificial cranial deformation. Am J Phys 1996;201:389–392.
Anthropol 1958;16:269–286. 212. Mori K, Sakamoto T, Nishimura K, Fujiwara K. Subarachnoid fluid col-
187. Moss ML. Functional anatomy of cranial synostosis. Childs Brain lection in infants complicated by subdural hematoma. Childs Nerv Syst
1975;1:22–33. 1993;9:282–284.
188. Cinalli G, Sainte-Rose C, Kollar EM, et al. Hydrocephalus and cranio- 213. McNeely PD, Atkinson JD, Saigal G, O’Gorman AM, Farmer JP. Subdural
synostosis. J Neurosurg 1998;88:209–214. hematomas in infants with benign enlargement of the subarachnoid
189. Steinbok P, Hall J, Flodmark O. Hydrocephalus in achondroplasia: spaces are not pathognomonic for child abuse. Am J Neuroradiol 2006;27:
the possible role of intracranial venous hypertension. J Neurosurg 1725–1728.
1989;71:42–48. 214. Ravid S, Maytal J. External hydrocephalus: a probable cause for subdural
190. Lundar T, Bakke SJ, Nornes H. Hydrocephalus in an achondroplastic child hematoma in infancy. Pediatr Neurol 2003;28:139–141.
treated by venous decompression at the jugular foramen. Case report. 215. Galassi E, Tognetti F, Gaist G, et al. CT scan and metrizamide cisternog-
J Neurosurg 1990;73:138–140. raphy in arachnoid cysts of the middle cranial fossa: calssification and
191. Martinez-Perez D, Vander Woude DL, Barnes PD, Scott RM, Mulliken pathophysiological aspects. Surg Neurol 1982;17:363–369.
JB. Jugular foraminal stenosis in Crouzon syndrome. Pediatr Neurosurg 216. Naidich TP, McLone DG, Radkowski MA. Intracranial arachnoid cysts.
1996;25:252–255. Pediatr Neurosci 1986;12:112–122.
192. McLone DG, Partington MD. Arrest and compensation of hydrocephalus. 217. Morris MW, Smith S, Cressman J, Ancheta J. Evaluation of infants with
Neurosurg Clin North Am 1993;4:621–624. subdural hematoma who lack external evidence of abuse. Pediatrics
193. Collmann H, Sörensen N, Krauss J. Hydrocephalus in craniosynostosis: a 2000;105:549–553.
review. Childs Nerv Syst 2005;21:125-131. 218. Yamada H, Yokota A, Furuta A, Horie A. Reconstitution of shunted mantle
194. Taylor W, Hayward R, Lasjaunias P, et al. Enigma of raised intracranial in experimental hydrocephalus. J Neurosurg 1992;76:856–862.
pressure in patients with complex craniosynostosis: the role of abnormal 219. Wright LC, McAllister JP, II, Katz SD, et al. Cytological and cytoarchitec-
intracranial venous drainage. J Neurosurg 2001;94:377–385. tural changes in the feline cerebral cortex during experimental infantile
195. Di Rocco C, McLone DG, Shimoji T, Raimondi AJ. Continuous interven- hydrocephalus. Pediatr Neurosurg 1990–1991;16:139–155.
tricular cerebrospinal fluid pressure recording in hydrocephalic children 220. Greitz D. Paradigm shift in hydrocephalus research in legacy to Dandy’s
during wakefulness and sleep. J Neurosurg 1975;42:683–689. pioneering work: rationale for third ventriculostomy in communicating
196. Di Rocco C, Maira G, Rossi G, Vignati A. Cerebrospinal fluid pressure hydrocephalus. Childs Nerv Syst 2007;23:487–489.
studies in normal pressure hydrocephalus and cerebral atrophy. Eur Neurol 221. Teo C, Jones R. Management of hydrocephalus by endoscopic third
1976;14:119–128. ventriculostomy in patients with myelomeningocele. Pediatr Neurosurg
197. Stein BM, Fraser RA, Tenner MS. Normal pressure hydrocephalus: com- 1996;25:57–63.
plication of posterior fossa surgery in children. Pediatrics 1972;49:50–52. 222. Drake JM, Canadian Pediatric Neurosurgery Study Group. Endoscopic
198. Barnett GH, Hahn JF, Palmer J. Normal pressure hydrocephalus in chil- third ventriculostomy in pediatric patients: the Canadian experience.
dren and young adults. Neurosurgery 1987;20:904–907. Neurosurgery 2007;60:881–886.
199. Prassopoulos P, Cavouras D, Golfinopoulos S, Nezi M. The size of the 223. Warf BC, Campbell JW. Combined endoscopic third ventriculostomy
intra-and extraventricular cerebrospinal fluid compartments in children and choroid plexus cauterization as primary treatment of hydrocephalus
with idiopathic benign widening of the frontal subarachnoid space. Neu- for infants with myelomeningocele: long-term results of a prospective
roradiology 1995;37:418–421. intent-to-treat study in 115 East African infants. J Neurosurg Pediatr 2008;2:
200. Barlow CF. CSF dynamics in hydrocephalus—with special attention to 310–316.
external hydrocephalus. Brain Dev 1984;6:119–127. 224. Warf BC, Kulkarni AV. Intraoperative assessment of cerebral aqueduct pat-
201. Briner S, Bodensteiner J. Benign subdural collections of infancy. Pediatrics ency and cisternal scarring: impact on success of endoscopic third ventric-
1980;67:802–804. ulostomy in 403 African children. J Neurosurg Pediatr 2010;5:204–209.
202. Carolan PL, McLaurin RL, Tobin RB, Tobin JA, Egelhoff JC. Benign extra- 225. Crosby RM, Henderson CM, Paul RL. Catheterization of the cerebral aque-
axial collections of infancy. Pediatr Neurosci 1986;12:140–144. duct for obstructive hydrocephalus in infants. J Neurosurg 1973;38:596–
203. Nickel RE, Gallenstein JS. Developmental prognosis for infants with 601.
benign enlargement of the subarachnoid spaces. Dev Med Child Neurol 226. Miki T, Nakajima N, Wada J, Haraoka J. Indications for neuroendoscopic
1987;29:181–186. aqueductoplasty without stenting for obstructive hydrocephalus due to
204. Karantanas AH, Bakratsi E. Benign enlargement of peripheral subarach- aqueductal stenosis. Minim Invasive Neurosurg 2005;48:136–141.
noid spaces in twins. Comput Med Imaging Graph 2002;26:47–48. 227. Fischbein NJ, Ciricillo SF, Barr RM, et al. Endoscopic third ventriculoscis-
205. Kim D, Choi J, Yoon S, Suh J, Kim T. Hypertrophic arachnoid membrane ternostomy: MR assessment of patency with 2D cine phase-contrast versus
in infantile external hydrocephalus: neuroradiologic and histopathologic T2 weighted fast spin echo technique. Pediatr Neurosurg 1998;28:70–78.
correlation. Radiology 1993;189(P):195. 228. Kim S-K, Wang K-C, Cho B-K. Surgical outcome of pediatric hydro-
206. Gordon N. Apparent cerebral atrophy in patients on treatment with cephalus treated by dndoscopi third ventriculostomy: prognostic fac-
steroids. Dev Med Child Neurol 1980;22:502–506. tors and interpretation of postoperative neuroimaging. Childs Nerv Syst
207. Ito M, Takao P, Okumo T, Mikawa H. Sequential CT studies of 24 chil- 2000;16:161–169.
dren with infantile spasms on ACTH therapy. Dev Med Child Neurol 229. St George E, K N, Sgouros S. Changes in ventricular volume in hydro-
1983;25:475–480. cephalic children following successful endoscopic third ventriculostomy.
Childs Nerv Syst 2004.

Barkovich_Chap08.indd 855 5/6/2011 10:26:33 PM


856 Pediatric Neuroimaging

230. Lev S, Bhadelia RA, Estin D, Heilman CB, Wolpert SM. Functional analysis 251. Vinchon M, Lemaitre M, Vallee L, Dhellemmes P. Late shunt infection:
of third ventriculostomy patency with phase contrast MRI velocity mea- incidence, pathogenesis, and therapeutic implications. Neuropediatrics
surements. Neuroradiology 1997;39:175–179. 2002;33:160–173.
231. Hoffmann K, Lehmann T, Baumann C, Felix R. CSF flow imaging in the 252. Lewis AI, Keiper GL, Crone KR. Endoscopic treatment of loculated hydro-
management of third ventriculostomy with a reversed fast imaging with cephalus. J Neurosurg 1995;82:780–785.
steady-state precession sequence. Eur Radiol 2003;13:1432–1437. 253. Lee K, Bae W, Bae H, Yun I. The fate of traumatic subdural hygroma in
232. Faillace WJ, Canady AI. Cerebrospinal fluid shunt malfunction signaled by serial computed tomography scans. J Korean Med Sci 2000;15:560–568.
new or recurrent seizures. Childs Nerv Syst 1990;6:37–40. 254. Falhauer K, Schmitz P. Overdrainage phenomena in shunt treated hydro-
233. Forbes K, Pipe J, Karis J, Farthing V, Heiserman J. Brain imaging in the cephalus. Acta Neurochir (Wien) 1978;45:89–101.
unsedated pediatric patient: comparison of periodically rotated overlap- 255. Emery JL. Intracranial effects of long-standing decompression of the
ping parallel lines with enhanced reconstruction and single-shot fast spin- brain in children with hydrocephalus and meningomyelocele. Dev Med
echo sequences. Am J Neuroradiol 2003;24:794–798. Child Neurol 1965;7:302–309.
234. Patel M, Klufas R, Alberico R, Edelman R. Half-fourier acquisition single- 256. Destian S, Heier LA, Zimmerman RD, et al. Differentiation between men-
shot turbo spin-echo (HASTE) MR: comparison with fast spin-echo MR ingeal fibrosis and chronic subdural hematoma after ventricular shunt-
in disease of the brain. Am J Neuroradiol 1997;18:1635–1640. ing: value of enhanced CT and MR scans. Am J Neuroradiol 1989;10:
235. Pipe J. Motion correction with PROPELLER MRI: applciation to head 1021–1026.
motion and free-breathing cardiac imaging. Magn Reson Med 1999;42: 257. Rekate HL. Classification of slit-ventricle syndrome using intracranial
963–969. pressure monitoring. Pediatr Neurosurg 1993;19:15–20.
236. Sugahara T, Korogi Y, Hirai T, et al. Conmparison of HASTE and segment- 258. Hyde-Rowan MD, Rekate HL, Nulsen FE. Reexpansion of previously
ed-HASTE sequences with a T2-weighted fast spin-echo sequence in the collapsed ventricles: the slit ventricle syndrome. J Neurosurg 1982;56:
screening evaluation of the brain. Am J Roentgenol 1997;169:1401–1410. 536–539.
237. Oi S, Matsumoto S. Infantile hydrocephalus and the slit ventricle syn- 259. Epstein FJ, Fleisher AS, Hochwald GM, Ransohoff J. Subtemporal
drome in early infancy. Child’s Nerv Syst 1987;3:145–150. craniectomy for recurrent shunt obstruction secondary to small ventricles.
238. Epstein F, Lapras C, Wisoff JH. “Slit ventricle syndrome”: Etiology and J Neurosurg 1974;41:29–31.
treatment. Pediatr. Neurosci 1988;14:5–10. 260. Epstein FJ, Hochwald GM, Wald A, Ransohoff J. Avoidance of shunt
239. May CH, Aurisch R, Kornrumpf D, Vogel S. Evaluation of shunt function dependency in hydrocephalus. Dev Med Child Neurol 1975;35:71-77.
in hydrocephalic patients with the radionuclide 99m-Tc-pertechnetate. 261. Bruce DA, Weprin B. The slit ventricle syndrome. Neurosurg Clin N Am
Childs Nerv Syst 1999;15:239–245. 2001;36:709–717.
240. Evans RC, Thomas MD, Williams LA. Shunt blockage in hydrocephalic 262. Serlo W, Heikkinen E, Saukkonen A-L, Wendt L. Classification and man-
children. The use of the valvogram. Clin Radiol 1976;27:489. agement of the slit ventricle syndrome. Childs Nerv Syst 1985;1:194–199.
241. Guertin SR. Cerebrospinal fluid shunts. Evaluation, complications, and 263. Benzel EC, Reeves JD, Kesterson L, Hadden TA. Slit ventricle syndrome
crisis management. Pediatr Clin North Am 1987;34:203–217. in children: clinical presentation and treatment. Acta Neurochir (Wien)
242. Howman-Giles R, McLaughlin A, Johnston I, Whittle I. A radionuclide 1992;117:7–14.
method of evaluating shunt function and CSF circulation in hydrocepha- 264. Di Rocco C. Is the slit ventricle syndrome always a slit ventricle syndrome?
lus. Technical note. J Neurosurg 1984;61:604–605. Childs Nerv Syst 1994;10:49–58.
243. Sweeney LE, Thomas PS. Contrast examination of cerebrospinal fluid 265. Forghani R, Farb RI. Diagnosis and temporal evolution of signs of
malfunction in infancy and childhood. Pediatr Radiol 1987;17:177–183. intracranial hypotension on MRI of the brain. Neuroradiology 2008;50:
244. Martin AJ, Drake JM, Lemaire C, Henkelman RM. Cerebrospinal fluid 1025–1034.
shunts: flow measurements with MR imaging. Radiology 1989;173:243–247. 266. Caldarelli M, Novegno F, Di Rocco C. A late complication of CSF shunt-
245. Castillo M, Hudgins PA, Malko JA, Burrow BK, Hoffman JC, Jr. Flow- ing: acquired Chiari I malformation. Childs Nerv Syst 2009;25:443–452.
sensitive MR imaging of ventriculoperitoneal shunts: in vitro findings, 267. Chumas PD, Armstrong DC, Drake JM, et al. Tonsillar herniation: the rule
clinical applications, and pitfalls. Am J Neuroradiol 1991;12:667–672. rather than the exception after lumboperitoneal shunting in the pediatric
246. Norbash A, Pelc N, Shimakawa A, Enzmann D. Shunt flow measurement population. J Neurosurg 1993;78:568–573.
and evaluation of valve oscillation with a spin-echo phase-contrast MR 268. Di Rocco C, Velardi F. Acquired Chiari I malformation managed by
sequence. Radiology 1994;190:560–564. supratentorial enlargement. Childs Nerv Syst 2003;19:800–807.
247. Minns RA, Brown JK, Engleman HM. CSF production rate: “real time” 269. Davidson RI. Peritoneal bypass in the treatment of hydrocephalus:
estimation. Z Kinderchir 1987;47:36–40. historical review of abdominal complications. J Neurol Neurosurg Psychia-
248. Sgouros S, Malluci C, Walsh AR, Hockley AD. Long term complications of try 1976;39:640–648.
hydrocephalus. Pediatr Neurosurg 1995;23:127–132. 270. Elisevich K, Mattar A, Cheeseman F. Biodegradation of distal shunt cath-
249. Drake JM, Kestle JRW, Tuli S. CSF shunts 50 years on—past, present and eters. Pediatr Neurosurg 1994;21:71–76.
future. Childs Nerv Syst 2000;16:800–804. 271. Citrin CM, Hammock MK. Computed tomographic representation of
250. Casey A, Kimmings E, Kleinlugtebeld A, Taylor W, Harkness W, Hayward granulomatous reaction from intraventricular shunt. Comput Tomogr
R. The long term outlook for hydrocephalus in childhood. Pediatr Neuro- 1979;3:177–180.
surg 1997;27:63–70.

Barkovich_Chap08.indd 856 5/6/2011 10:26:33 PM


CHAPTER

Congenital Anomalies
9 of the Spine
ERIN SIMON SCHWARTZ AND A. JAMES BARKOVICH

Normal and abnormal embryogenesis of the spine: Anterior sacral meningocele


an overview Sacrococcygeal teratoma
Gastrulation and neurulation Anomalies of development of the notochord
Canalization and retrogressive differentiation The split notochord syndrome spectrum (complex spinal
Formation of the vertebral column dysraphisms)
Anomalies of spinal formation: concepts Split cord malformation (diastematomyelia and diplomyelia—
Clinical manifestations of spinal anomalies complex spinal dysraphism)
Terminology Malformations of unknown origin
Counting anomalous vertebrae Segmental spinal dysgenesis (a complex spinal dysraphism)
Imaging techniques Dorsal meningocele (a closed spinal dysraphism with a
Imaging of patients with idiopathic scoliosis subcutaneous mass)
Abnormalities of neurulation (disorders in which the spinal Lateral meningocele
cord does not completely fuse posteriorly) Congenital tumors of the spine
Disorders resulting from nondisjunction Teratoma
Disorders resulting from premature disjunction—spinal lipomas Dermoid and epidermoid tumors
Anomalies of the caudal cell mass Hamartoma
Normal conus medullaris and filum terminale Syringohydromyelia
The terminal ventricle Definition
Anomalies of the filum terminale/tight filum terminale Clinical presentation and course
(a simple spinal dysraphism) Classification and causes
Syndrome of caudal regression (a complex spinal dysraphism) Theories of pathogenesis
Terminal myelocystocele (a closed spinal dysraphism with a Imaging
subcutaneous mass)

streak. At days 15 and 16, cells enter the primitive pit and migrate
NORMAL AND ABNORMAL inward between the ectoderm and endoderm and then course laterally
EMBRYOGENESIS OF THE SPINE: and undergo epithelial-to-mesenchymal transition, to form the inter-
AN OVERVIEW posed mesoderm. Initially, no cells migrate in the midline; later, these
mesodermal cells will join in the midline to form the notochordal pro-
An understanding of the normal embryonic developmental sequence
cess, which will eventually roll into a tube, separate from the endoderm,
of the spine is invaluable to the understanding of spinal anomalies.
and become the notochord. During a process known as intercalation,
Furthermore, combining knowledge of embryology with understand-
the notochordal process fuses with the endoderm, creating a commu-
ing of the anatomic relationships in the mature congenital lesion allows
nication of the central canal of the notochordal process with the yolk
insight into the time and stage of development at which the normal
sac. As the central canal is already in communication with the amniotic
sequence was altered; this insight leads to a better understanding of
cavity through the primitive pit, a transient communication is present
developmental lesions. Normal development of the spine will therefore
all the way from the yolk sac to the amniotic sac; this communica-
be discussed in some detail.
tion is called the primitive neurenteric canal. Once formed, the noto-
chord induces the formation of a plate of ectodermal cells in the dorsal
Gastrulation and Neurulation
midline, beginning immediately cephalad to Hensen node (Fig. 9-1).
On about the 15th day of embryonic life, ectodermal cells proliferate Under the influence of bone morphogenetic proteins (BMPs), most
to form a plate, the primitive streak, along the surface of the embryo. of the ectoderm is prevented from differentiating into neuroectoderm
A rapidly proliferating group of cells forms at one end of the primi- (1). However, suppression of the BMPs by antagonists such as chor-
tive streak; this nodular proliferation, surrounding a small primitive din, noggin, and follistatin, emanating from the primitive node, allows
pit (known as Hensen node), defines the cephalic end of the primitive neuroectoderm to form in the midline (1,2). The lateral edges of this

857

Barkovich_Chap09.indd 857 5/6/2011 10:28:20 PM


858 Pediatric Neuroimaging

FIG. 9-1. Schematic showing the development of the neu-


ral plate. The primitive streak forms along the surface of the
embryo by about 15 days of embryonic life. A small primitive
pit lies at what will become the cephalic end of the primitive
streak; a nodular proliferation of cells surrounding the primi-
tive pit becomes known as Hensen node. On about days 15
and 16, cells enter the primitive pit and migrate cephalad in
the midline to form the notochordal process, which will even-
tually become the notochord. The notochordal process and
notochord induce formation of a plate of ectodermal cells
dorsally in the midline; this is the neural plate.

midline structure, now known as the neural plate, are contiguous with overlying ectoderm separates from the neural tissue and the edges of
the ectoderm from which the plate has differentiated, now known as the ectoderm meet in the midline and fuse, forming a continuous ecto-
superficial or cutaneous ectoderm. dermal covering of the neural structures, with the mesenchymal cells of
After the neural plate is formed, it is shaped into an elongated the neural crest migrating between the cutaneous and neural ectoderm
structure that is broad at the anterior (cranial) end and narrow at the layers (2). Progressive folding and closure of the neural structures and
posterior (caudal) end. The major driving force of the shaping is a separation from ectoderm then proceed both cranially and caudally
mediolateral elongation of a group of cells, along with development of from each point of initial closure, ultimately resulting in complete
polarized cellular protrusions that enable the cells to migrate medially closure (10–13). The initial closure of the neural tube in humans, the
and intercalate with neighboring cells close to the midline (3). This posterior neuropore, is believed to be at the hindbrain-cervical junction,
midline convergence of cells causes an anterior-posterior elongation from which closure extends in both directions. A second site of closure
and narrowing of the neural plate (4,5). This process is strongly related in mice is at the forebrain-midbrain boundary; this site has not been
to the development of cell polarity (4,5). Along with the process of confirmed in humans. The necessity of this site even in mice is ques-
shaping, the neural plate also bends. At approximately 17 days of gesta- tionable, as about 80% of mice that lack the second closure point still
tion, the lateral portions of the neural plate begin to thicken bilaterally, achieve complete cranial closure (14). The “third” site of closure occurs
forming the neural folds; the process of bending elevates these folds at the most rostral extremity of the forebrain, the lamina terminalis
and brings them to the dorsal midline (6). The process involves the for- (2). The exact site of the most caudal end of the neurulation-formed
mation of “hinge points” at two sites: the median hinge point (MHP) neural tube has been debated, but most experts believe that it is at the
in the ventral midline and extending over the rostrocaudal extent of S-2 level (12,15–21).
the neural plate and the paired dorsolateral hinge points (DLHPs),
which form mainly at the levels of the developing brain (2) and lower
Canalization and Retrogressive Differentiation
spine. The formation of these hinge points is controlled by secretion of
the signal transduction protein sonic hedgehog by the notochord (7), A portion of the neural tube develops caudal to the posterior neu-
as well as an inhibitory interaction between BMP2 and noggin, par- ropore. This elongation is not due to primary neurulation but is the
ticularly in the lower spine (8). After formation of the hinge points, the result of another process known as canalization (or secondary neu-
more lateral aspects of the neural plate are elevated around the MHP, rulation). In this process, a caudal cell mass (also called the tail bud),
bringing the DLHPs upward and toward the midline (Fig. 9-2). This composed of undifferentiated, pleuripotential cells (the residua of the
elevation is accomplished by a poorly understood process called apical primitive streak [2,21]), forms in the tail fold as a result of fusion of
constriction, in which columnar cells of the neural tube are converted neural epithelium (at the caudal end of the embryo) with the noto-
into wedge-shaped cells (9). Eventually, the lateral folds contact one chord. Ventral to the caudal cell mass, with the notochord interposed,
another in the dorsal midline and adhere to one another, with their lies the cloaca, which will form the cells of the anorectal and lower
fusion forming the neural tube (neurulation). Neurulation seems to genitourinary tract structures.
begin separately at at least two different levels in humans, when cel- By the time the embryo is 30 days old, multiple microcysts and
lular protrusions (possibly cilia) project medially from the most dorsal clumps of cells begin to appear in the caudal cell mass. These micro-
cells of the neural folds on either side. Cell recognition and adhesion cysts coalesce to form an ependyma-lined tubular structure that unites
occur under the influence of many molecules (Ephrin-A5, EphA7, with the neural tube above (Fig. 9-3). This process is not as organized as
neural cell adhesion molecule, and neural cadherin among them [3]), the process of neurulation; the multiple accessory lumina and ependy-
closing the tube at each point. Immediately following closure, the mal rests in the normal filum terminale and distal conus medullaris

Barkovich_Chap09.indd 858 5/6/2011 10:28:20 PM


Chapter 9 • Congenital Anomalies of the Spine 859

FIG. 9-2. Normal and abnormal neurulation. A–E. Normal neurulation. The neural plate is composed of neural ectoderm, which is continuous with
cutaneous ectoderm on either side. The cells at the junction of the neural ectoderm and cutaneous ectoderm will eventually differentiate into neural crest
cells (A). At approximately 17 days of gestation, the lateral portions of the neural plate begin to thicken, forming the neural folds (B). Contractile filaments
located in the neuroepithelial cells in the neural folds contract, causing the neural folds to bend dorsally along the entire length of the neuroaxis, bringing the
edges of the neural folds toward one another in the midline (C). Neurulation (closure of the neural tube) begins when the neural folds meet in the midline.
At the time of closure, the overlying ectoderm separates from the neural tissue and fuses in the midline, forming a continuous ectodermal covering of the
neural structures. At the same time, the neural crest cells are extruded from the neural tube to form a transient structure immediately dorsal to the tube
(D). Eventually these neural crest cells will migrate to form dorsal root ganglia and multiple other structures (E). F–G. Abnormal neurulation. When there
is premature disjunction of neural ectoderm from cutaneous ectoderm, the surrounding mesenchyme gains access to the inner surface of the neural tube.
When mesenchyme comes in contact with this primitive ependymal lining, it evolves into fat. This is believed to be the process underlying the formation
of spinal lipomas (F). Complete nondisjunction of cutaneous ectoderm from neural ectoderm results in the formation of myelomeningoceles (Fig. 9-4).
Focal nondisjunction results in a persistent epithelium-lined connection between the central nervous system and the skin (G). This persistent connection
has been labeled a dorsal dermal sinus.

of the adult are believed to result from the disorder of this process. cell death (apoptosis) of the portion derived from primary neurula-
Several neuronal markers expressed in vertebrate embryos are thought tion (21) and necrosis of the portion derived from secondary neuru-
to modulate the differentiation of structures derived from the caudal lation (23). This process has been named retrogressive differentiation
cell mass and be involved in the maturation of the caudal spinal cord. (Fig. 9-3). The caudal segment (formed by canalization and retrogres-
These include N-CAM, synaptophysin, 3A10, and NeuN (22). The sive differentiation) eventually becomes the most caudal portion of
final stage in the formation of the distal spinal cord begins at about the conus medullaris, the filum terminale, and a focal dilatation of the
38 days of gestation, at which time the cell mass and central lumen central canal (within the conus medullaris) known as the ventriculus
of the caudal neural tube decrease in size as a result of programmed terminalis (15–20,24).

Barkovich_Chap09.indd 859 5/6/2011 10:28:21 PM


860 Pediatric Neuroimaging

FIG. 9-3. Canalization and retrogressive differentiation. After formation of the neural tube, a caudal cell mass forms in the tail fold as a result of fusion of
neural epithelium at the caudal end of the embryo with the notochord (A). By the age of 30 days, multiple cysts and clumps of cells appear in the caudal cell
mass (B). These cysts coalesce to form a tubular structure that unites with the neural tube above (C). At about 38 days, the cell mass and central lumen of the
caudal neural tube decrease in size through cell necrosis in a process known as retrogressive differentiation. The segment formed by this process eventually
forms the distalmost conus medullaris, the filum terminale, and the terminal ventricle (D).

Formation of the Vertebral Column mentioned intersegmental fissures. The inferior half of one sclerotome
then fuses with the superior half of the subjacent sclerotome across the
Formation of the vertebral column was discussed in Chapter 2 and fissure to form a vertebral body. This process proceeds bilaterally and
illustrated in Figure 2-23. A review of the subject is given here, as well, symmetrically so that the fusion of the sclerotomes on each side forms
for convenience of the reader. Development of the vertebral column half of the vertebral body on that side. As a consequence of this reseg-
can be divided into three periods. The first of these is membranous
mentation, the intersegmental arteries and veins become located in the
development. At about the 25th gestational day, the notochord sepa-
center of the new vertebral bodies.
rates from the primitive gut and neural tube to create two zones, the
During the second (chondrification) stage of vertebral development,
ventral subchordal and dorsal epichordal zones. These zones are filled
chondrification centers appear within sclerotomes at the cervicotho-
with mesenchyme, which migrates to them from its initial position lat-
racic junction region during the sixth embryonic week and then extend
eral to the neural tube.
rostrally and caudally. Six centers will form each vertebral level, with
The mesenchyme lateral to the closing neural tube organizes into
somites, which are separated by small intersegmental fissures. Somite two in the vertebral centrum, two forming the posterior vertebral arches
formation progresses from rostral to caudal (25). Each somite is and spinous process, and two within the transverse processes and cos-
divided into a medial (medial sclerotome) and a lateral (lateral myo- tal arch (27). Notochordal remnants persist between the newly formed
tome) portion. The medial sclerotome contributes to the formation of vertebral bodies and become incorporated into the intervertebral discs
the vertebrae, while the lateral myotome gives rise to the paraspinous as the nuclei pulposi. Portions of the thoracic sclerotomes later migrate
musculature. After the neural tube has closed and becomes separated ventrolaterally to form ribs. The anterior and posterior longitudinal
from the superficial ectoderm, mesenchyme also migrates dorsal to the ligaments form during chondrification, from mesodermal cells.
neural tube to form the precursors of the neural arches (in addition In the final stage of vertebral development (ossification), the chon-
to the meninges and paraspinous muscles). Differentiation of the ven- dral skeleton ossifies to complete the formation of the vertebrae. Ossi-
tral and dorsolateral mesenchyme, which will form the vertebral body, fication starts in three centers, one in the middle of the vertebral body
results from induction by the sonic hedgehog protein, whereas the dif- and one in each vertebral arch. The thoracolumbar junction region is
ferentiation of the dorsal mesenchyme, which will form the posterior the first to ossify, with rostral and caudal ossification to follow. The
elements, results from induction by a protein called BMP4 (26). Sub- formation of the vertebrae at the caudal end of the embryo proceeds
sequently, the sclerotomes separate transversely along the previously by a different, less organized process. A mass of cells composed of

Barkovich_Chap09.indd 860 5/6/2011 10:28:22 PM


Chapter 9 • Congenital Anomalies of the Spine 861

notochord, mesenchyme, and neural tissue merely divides into somites After disjunction, the cutaneous ectoderm fuses in the midline,
to form the sacral and coccygeal levels. Regression results in reduction dorsal to the closed neural tube. At the same time, perineural mes-
and fusion of most of these segments. As in development of the caudal enchyme migrates into the newly created space between the neural
neural tube, this apparent disorganization of the caudal cell mass leads tube and cutaneous ectoderm, surrounds the neural tube, and is
to frequent anomalous development. Caudal regression syndromes, induced to form the meninges, the bony spinal column, and the
lipomas, and teratomas can result (28,29). paraspinous musculature (38). The mesenchyme normally remains
isolated from the newly formed central canal of the spinal cord
Anomalies of Spinal Formation: Concepts because the neural tube closes immediately prior to, or simultane-
ously with, disjunction.
Embryological Concepts and Theories Defective disjunction can explain a number of diverse patho-
Embryological explanations of anomalies of the developing spine are logic lesions. For example, focal unilateral premature disjunction of
continually evolving. As new facts are discovered, old theories some- the neural ectoderm from the cutaneous ectoderm (prior to clo-
times have to be discarded and new theories developed. In contrast sure of the neural tube) allows the perineural mesenchyme to gain
to mathematical theories, embryological theories cannot be proved access to the neural groove and come in contact with the primi-
correct; they can only be verified or disproved. In clinical medicine, a tive ependymal lining of the groove. Mesenchyme that is exposed
theory is useful if it explains observations and helps to organize a clas- to the interior of the neural tube seems to develop into fat, either
sification; the ability to predict future observations is an added bonus. by receiving signals promoting adipocyte (fat cell) production or by
The classification scheme used in this chapter is based primarily upon the blockage of signals that prevent adipocyte production (39,40).
a theory of spinal dysraphism developed by David McLone, MD, PhD Therefore, focal premature disjunction of neural ectoderm from the
and Thomas Naidich, MD when they worked together at the Children’s superficial ectoderm could explain the development of spinal lipo-
Memorial Hospital in Chicago, Illinois; this work was based on original mas and lipomyelomeningoceles (41) (Fig. 9-2F). Spinal lipomas
concepts by Della Rovere (30). The more recent clinical-radiological situated rostral to the filum terminale seem to be formed in this
classification scheme proposed by Tortori-Donati et al. (31) incorpo- manner. Lipomas caudal to the conus medullaris more likely form
rates much of the work of the older classification schemes and adds a as a result of other processes, such as abnormal development of the
clinical component; however, it is not entirely in agreement with the caudal cell mass.
earlier works. This chapter discusses aspects of both classifications, Other anomalies of the spine can be explained by failure of dis-
as they are the most useful theories we have found for classifying and junction. Dermal sinus tracts may be the consequence of focal failure
explaining anomalies of the spine. of disjunction, resulting in a focal ectodermal-neural ectodermal tract
Developmental neurogenetics are largely ignored in the discussion (Fig. 9-2G). A related entity has been recently described, the dermal-
of spine anomalies in this chapter. This is not in any way an attempt sinus-like stalk, in which there is a solid stalk composed of fibrous
to minimize the value of developmental neurogenetics. Indeed, spi- tissue and fat (with or without nervous tissue) rather than the epithe-
nal anomalies result from a multitude of factors and genetic fac- lium-lined tract of the typical dermal sinus (42). The tract should pro-
tors are likely among the most important (interested readers can see hibit mesenchyme from migrating between the neural ectoderm and
[2,3,9,32–37]). However, despite many major advances in genetics over cutaneous ectoderm at that site, forming a focal spina bifida as is com-
the past 10 years, the details of developmental neurogenetics of normal monly seen in children with dermal sinus tracts. Moreover, the adhe-
and abnormal spinal development are still being worked out, and the sion between the ectoderm and neural ectoderm could explain the
inclusion of the bits and pieces that are known would likely prove more correlation of the dermatome level of the cutaneous lesion with the
confusing than enlightening to most practicing clinicians. neuroectodermal level of the central nervous system termination of
the tract. Open spinal dysraphism (OSD) (myelocele and myelomenin-
Neurulation Anomalies gocele) can be explained as a large area of nondisjunction (Figs. 9-2
The process of separation of the neural tube from the cutaneous and 9-4). These concepts are discussed in more detail in later sections
ectoderm during closure of the neural tube is known as disjunction. of this chapter.

FIG. 9-4. Open spinal dysraphism (Myelocele and Myelomeningocele). A. Myelocele. The neural placode is a flat plaque of neural tissue that is exposed to
the air. The dura is deficient posteriorly; the pia and arachnoid line the ventral surface of the placode and dura, forming an arachnoid sac that is continuous
with the subarachnoid space superiorly and inferiorly. Both the dorsal and ventral roots arise from the ventral surface of the placode. B. Myelomeningocele.
This is identical to the myelocele with the exception that there is an expansion of the ventral subarachnoid space, which posteriorly displaces the placode.

Barkovich_Chap09.indd 861 5/6/2011 10:28:23 PM


862 Pediatric Neuroimaging

Association of Spinal Anomalies with Other TERMINOLOGY


Systemic Anomalies
The term spinal dysraphism refers to a heterogeneous group of spinal
Spinal malformations are very commonly associated with anomalies of
anomalies. In spite of their heterogeneity, all lesions within this group
the viscera, as well as those of the spinal column. As discussed previ-
have incomplete midline closure of mesenchymal, osseous, and nervous
ously, the caudal cell mass forms in close anatomic proximity to the
tissue (20).
cloaca, the region of origin of the lower genitourinary tract and ano-
Spina bifida refers to incomplete closure of the bony elements of
rectal structures. As a result of this close embryological relationship,
the spine (lamina and spinous processes) posteriorly (56,57).
patients with anorectal and urogenital anomalies have a high incidence
OSD includes the myelocele, a midline plaque of neural tissue that
of lumbosacral hypogenesis, terminal myelocystoceles, and tethered
lies exposed at and is flush with the skin surface, and the myelomenin-
spinal cords (and vice versa) (43–45). Moreover, the notochord is
gocele, a myelocele that has been elevated above the skin surface by
associated with the induction of normal formation of thoracic and
expansion of the subarachnoid space ventral to the neural plaque.
abdominal viscera, as well as the neural tube. Therefore, patients with
Closed spinal dysraphisms are a group of lesions that develop
spinal anomalies secondary to abnormal formation of the notochord
beneath an intact dermis and epidermis; that is, there is no exposed
will often have anomalies of the upper gastrointestinal tract or the
neural tissue (58). The clinical-radiological classification system of
respiratory tract. Finally, it is important to remember that the devel-
Tortori-Donati, Rossi, and Cama subdivides this group into lesions
opment of the vertebral column is influenced by many of the same
with a subcutaneous mass (usually the result of a subcutaneous lipoma
factors that influence development of the spinal cord. Therefore, any
or a simple meningocele) and those without a subcutaneous mass (31)
condition that results in vertebral anomalies, such as the VACTERL
(Table 9-1). Included in the category of closed spinal dysraphism with
syndrome (46), the Klippel-Feil syndrome (47), and lumbosacral
a subcutaneous mass are lipomas with dorsal defect of the bony spi-
hypogenesis (48,49) should be investigated for associated anomalies
nal canal (lipomyelocele and lipomyelomeningocele), meningocele,
of the spinal cord.
and myelocystocele. Anomalies with closed spinal dysraphism lacking
a subcutaneous mass include the simple dysraphic states (fatty and/or
CLINICAL MANIFESTATIONS OF SPINAL fibrous thickening of the filum terminale, intraspinal lipoma, spina
bifida, and persistent terminal ventricle) and the complex dysraphic
ANOMALIES states (SCM, dorsal dermal sinus, caudal regression syndrome, segmen-
Many patients with spinal anomalies present either with external tal spinal dysgenesis, neurenteric cyst, and dorsal enteric fistula).
manifestations of the anomaly or with the tight filum terminale (teth-
ered cord) syndrome. The external manifestations vary with the type
of anomaly and are described in more detail in the sections on the
specific anomalies themselves. They include cutaneous hemangiomas, TABLE 9-1 Clinical-Radiological
dimples, hairy patches, atypical scoliosis, and large subcutaneous Classification System
lipomas. The syndrome of the tight filum terminale, also known as the
tethered spinal cord syndrome, denotes a complex of neurologic and for Spinal Dysraphism
orthopedic deformities. This clinical syndrome may be associated
with split cord malformation (SCM), with spinal lipoma, or with a Open Spinal Dysraphism
short, thick filum terminale; frequently the conus medullaris is in a (Hemi-)myelomeningocele
low position. Patients may present with new onset of symptoms at (Hemi-)myelocele (also known as myeloschisis)
any age (50); the authors have seen patients in nearly every decade
of life (as late as the eighth decade) become suddenly symptomatic Closed Spinal Dysraphism
from their tethered spinal cords. Patients suffer from difficulty with With a Subcutaneous Mass
locomotion, ranging from muscle stiffness to actual weakness; some Lipoma with dorsal defect (lipomyelomeningocele and
exhibit abnormal lower extremity reflexes. The patients can also lipomyelocele)
exhibit bladder dysfunction (usually manifest as a low-pressure, drib- Myelocystocele (terminal or cervical)
bling urine stream), sensory changes with abnormal somatosensory Meningocele
evoked potentials, orthopedic deformities of the lower extremities Cervical myelomeningocele
(most commonly clubfoot), and back pain (particularly with exer-
tion). Urodynamic studies are typically abnormal. Bowel dysfunc- Without a Subcutaneous Mass
tion is uncommon. Although scoliosis frequently accompanies this Simple Dysraphic States
syndrome, it is rarely the sole complaint (20,51–53). Symptoms are Spina bifida
frequently worse in the mornings and after exercise. An increased Persistence of the terminal ventricle
incidence of tethered spinal cords is seen in lumbosacral hypogenesis, Intraspinal lipoma (intradural or filum terminale)
in the VACTERL syndrome (46,49), and in anorectal malformations Tight filum terminale (fibrous thickening)
(imperforate anus), including low lesions (54). Complex Dysraphic States
Adults with tethered spinal cords may present somewhat differently Dorsal dermal sinus
than affected children. Adults present more often with pain, possibly Caudal regression syndrome
because of accompanying degenerative changes, and less commonly Split notochord syndrome (dorsal enteric fistula and neurenteric cyst)
with incontinence, weakness, or scoliosis (50,53). Indeed, it has been Split cord malformation (diastematomyelia and diplomyelia)
suggested that tethering of the spinal cord accelerates disc degenera- Segmental spinal dysgenesis
tion. Adults who present with symptoms of lumbar disc degeneration
without magnetic resonance imaging (MRI) evidence of disc disease Adapted from Tortori-Donati et al. (31).
should be investigated for a tight filum terminale (55).

Barkovich_Chap09.indd 862 5/6/2011 10:28:24 PM


Chapter 9 • Congenital Anomalies of the Spine 863

Counting Anomalous Vertebrae (92,94). In these patients, MRI should be used to look for neoplasm,
Chiari I malformation, syringohydromyelia, tethered spinal cord or
Identifying the specific level in patients with vertebral anomalies, such other cord anomaly, and intradural or extradural cysts. Any of these
as hemivertebrae and butterfly vertebrae, can be challenging. One way conditions can lead to scoliosis; more importantly, surgical treatment
to approach this is to count all of the vertebrae (normal ones, butter- of the scoliosis before treatment of the underlying disorder may result
fly vertebrae, and hemivertebrae) in the numbering, but to designate in increased neurologic disability.
the hemivertebra as being extra by adding an “h” and the butterfly by
adding a “b.” For example, if C-6 is a hemivertebra, it is referred to as
C-6h. A butterfly T-8 would be T-8b. A report of a patient with these ABNORMALITIES OF NEURULATION
two segmentation anomalies would state that the spine is not a C-7/T- (DISORDERS IN WHICH THE SPINAL
12/L-5/S-5 (i.e., normal) spine, but a C-8(1h)/T-12(1b)/L-5/S-5 spine
with the hemivertebra at C-6 and the butterfly vertebra at T-8. This
CORD DOES NOT COMPLETELY FUSE
system can be useful for transmitting the necessary clinical data to the POSTERIORLY)
referring physician. Accurate determination of vertebrae can only be
Disorders Resulting from Nondisjunction
performed by labeling from the craniocervical junction and proceed-
ing inferiorly. Open Spinal Dysraphism—Myelocele and
Myelomeningocele
Myeloceles and myelomeningoceles most likely result from a localized
IMAGING TECHNIQUES lack of closure of the neural tube, resulting from a lack of expression of
The spine can be imaged using a variety of techniques. Sonography, specific receptors on the surface of the neuroectodermal cells (34,95).
x-ray computed tomography (CT), and MRI can all give exquisite Most authors use the term “myelomeningocele” to refer to any OSD; we
images of the diverse anatomic anomalies that characterize these enti- use the term OSD to encompass the myelocele and myelomeningocele.
ties (40,47,48,58–89). Therefore, an understanding of the characteristic All other anomalies are considered closed spinal dysraphisms.
anatomic features that distinguish these various entities may be more Open neural tube defects cause neurological disabilities, including
important than the imaging modality for the diagnosis and presurgical paraplegia, hydrocephalus, incontinence, sexual dysfunction, skeletal
planning of these entities. deformities, and, often, cognitive impairment (96). Experimental evi-
An important concept to remember, however, is that a single dence suggests that the neurological deficits of patients with OSD are
patient may have multiple spinal anomalies. For example, a patient not entirely caused by the open neural tube defect, but also by chronic
with a lipomyelomeningocele may have, in addition, SCM, a tight filum mechanical injury and amniotic fluid-induced chemical trauma that
terminale, or a dorsal dermal sinus. Therefore, it is important to image progressively damages the exposed fetal neural tissue during gestation
the entire spine if a cutaneous anomaly or vertebral anomaly suggests (97,98). Early in utero repair of OSD in fetal sheep has been shown
the presence of a malformation. The ability to noninvasively perform to interrupt the spinal cord injury and improve spinal cord function
high-resolution imaging of the entire spine in the sagittal and coronal at the time of birth (97). Several institutions have performed in utero
planes in a very few sequences without ionizing radiation is an advan- OSD repairs in human fetuses; reports suggest postoperative improve-
tage of MR that other modalities cannot match. For example, one study ment of the associated Chiari II malformation, ventriculomegaly,
comparing MR and ultrasound in the infant spine found that, while a lower extremity function, and brainstem function (99–102) in fetuses
normal ultrasound was sufficient to rule out most spinal pathology, treated before 25 gestational weeks. Thus, prenatal diagnosis of OSD
additional pathology is picked up by MR in 20% of cases (90). Detec- by ultrasound or MRI (Fig. 9-5) has made postnatal diagnosis much
tion of a thick or fatty filum terminale in a patient whose conus medul- less common, in regions where these modalities are widely available.
laris terminates at a normal level may be difficult with sonography but The ongoing prospective multi-institutional study will determine the
is easy on axial MR images. Finally, sonography becomes progressively benefits of prenatal repair of this spinal anomaly on neurological and
less useful as ossification of the posterior elements proceeds during the functional outcome.
first year of life while MR remains superb. Typical MR sequences for The exact cause of the impaired closure of the neural tube in
spine imaging are discussed in Chapter 1. It should be added that spiral patients with OSD has not been discovered. Environmental factors
CT, with the rapid scanning time and easy curvilinear and planar ref- implicated in increasing the risk include socioeconomic class, mater-
ormations (in any plane) allow excellent evaluation of osseous spinal nal age, maternal diet, maternal diabetes and obesity, and exposure
anomalies and, if intrathecal contrast is administered, good evalua- to antiepileptic drugs. Genetic components include association with
tion of intraspinal lesions. In our practices, MRI is currently the initial trisomy 13, 18, and 21; association with genetic syndromes (Meckel
imaging modality of choice because it is noninvasive and does not use syndrome and anal stenosis); racial differences in incidence rates;
ionizing radiation but, for some cases, CT myelography gives impor- increased risk for a second affected child (3–5×) for couples who have
tant supplementary information. one affected infant; and a tenfold increase in risk for siblings of affected
children compared to the general population (103,104). Strong evi-
dence has accumulated for a protective role of folate in this disorder.
Imaging of Patients with Idiopathic Scoliosis
Animal work suggests that the folate deficiency impairs biosynthesis
While the necessity of preoperative imaging with MRI was previously of pyrimidine, the presence of which may help to compensate for an
controversial, there is now general agreement that patients with scolio- underlying genetic predisposition (105). Localized differences in folate
sis should have their spinal columns imaged by MR before undergoing metabolism exist early during neural tube formation, and it is believed
distraction and instrumentation of their spines (91). This is particu- that folate may modulate proliferation in the midline via effects on
larly true in any patient presenting with clinical deterioration, atypi- Aldh1l1+ cells (106). Offspring of women whose diet is supplemented
cal features of scoliosis (rapid progression, abnormal location of the with folate have a significantly reduced incidence of neural tube defects
curve, or a left thoracic curve), neurological signs or symptoms (92), (10,107–109). This has led to investigations of genes involved in folate
age less than 11 years (93), or severe scoliosis (Cobb angle > 45°–50°) metabolism as being potentially related to development of OSD.

Barkovich_Chap09.indd 863 5/6/2011 10:28:24 PM


864 Pediatric Neuroimaging

FIG. 9-5. Prenatal imaging of an OSD (myelomeningocele). A. Sagittal sonogram using 4-MHz transducer shows a focal loss of the normal echogenicity
of the posterior elements of the vertebrae, indicating a bony spina bifida. The meningocele is seen as a hypoechogenic region (arrows) extending dorsally
at the level of the spina bifida. B. Higher resolution image using 8-MHz transducer shows curvilinear echoes (arrows) representing neural tissue running
through the meningocele. C. Sagittal image through the spine shows the lumbosacral spina bifida with the dorsal myelomeningocele (small white arrows).
The Chiari II malformation (larger white arrows) is seen at the craniocervical junction. Note the massive hydrocephalus. D. Axial image shows the dorsal
bony spina bifida. At this level, the placode (black arrows) is still within the spinal canal.

Two identified polymorphisms (C677T and possibly A1298C) in placode. The posterior (dorsal) surface of the placode is made up of the
the homocysteine remethylation gene MTHFR are associated with a tissue that would normally form the internal, ependymal lining of the
1.8-fold increase in the risk of an open neural tube defect (14). Interest- neural tube. The anterior (ventral) surface of the placode corresponds
ingly, although offspring of obese women (defined as weighing more to what would normally be the external surface of the spinal cord
than 70 kg) have an increased incidence of neural tube defects, no risk (pia mater) (Fig. 9-4).
reduction has been found among these women after administration of The lack of separation of the placode from the cutaneous ectoderm
folate (110,111), possibly because women with high body mass indi- prevents the mesenchyme from migrating into the area posterior to the
ces have low folate levels compared with nonobese women, even when neural ectoderm; it is forced to remain anterolateral to the nervous tis-
controlling for folate intake (112). sue. Thus, the pedicles and lamina (which are formed from this mesen-
In patients with OSD, the neural folds do not fuse in the midline chyme) are everted, facing posterolaterally instead of posteromedially
to form a neural tube in the affected zone; instead, the neural tube (Fig. 9-4). As a result of the external rotation of the lamina and pedi-
remains open and the neural folds remain in continuity with cutane- cles, the spinal canal undergoes a fusiform enlargement throughout
ous ectoderm at the skin surface (nondisjunction). The region of open the extent of the posterior osseous defect. The maximum enlargement
spinal cord, located at the skin surface and open to the air in the pos- of the canal occurs when the laminae are in the sagittal plane; further
terior midline, is a region of reddish tissue referred to as the neural rotation of the lamina diminishes the size of the canal (20).

Barkovich_Chap09.indd 864 5/6/2011 10:28:24 PM


Chapter 9 • Congenital Anomalies of the Spine 865

The vertebral bodies can be nearly normal or can have anomalies Imaging studies are rarely required in the newborn with an OSD
of segmentation ranging from a single hemivertebra to a jumbled (Fig. 9-6). The exposed placode is obvious upon visual examination
mass of malsegmented vertebral components. Segmentation anoma- and is usually repaired within 48 hours. After repair, the infants typi-
lies result in a short radius kyphoscoliosis in approximately one-third cally have a neurological deficit that is stable. They should not further
of patients with myelomeningoceles (20). Another 65% of affected deteriorate if the accompanying hydrocephalus, which is almost always
patients develop a (less severe) kyphoscoliosis as a result of a neuro- present unless the patient has had a prenatal repair, is controlled. Neu-
muscular imbalance (113,114). roradiologic examination of the spine is indicated if patients deterio-
In children with lumbar or lumbosacral OSD, the spinal cord is rate neurologically in spite of adequate treatment of hydrocephalus or
always tethered. The nerve roots in affected patients radiate in a spoke- if they have an unusual neurological examination, such as an asymmet-
wheel pattern as they leave the placode and travel rostrally, laterally, ric lower extremity neurological deficit, which should raise suspicion
and caudally to their respective neural foramina. that the patient has a SCM and hemiOSD.

FIG. 9-6. Myelomeningocele in a neonate. A and B. Sagittal T1-weighted (A) and T2-weighted images (B) show the spinal cord coursing caudally within
the spinal canal to the sacral level. The cord is expanded by syringomyelia (s), with some air (small white arrows) producing some susceptibility artifact
within the syrinx. The neural placode (p) extends dorsally through the bony spina bifida. The asterisks mark a wet cloth covering the placode. C. Axial
T1-weighted image shows the spinal cord (arrows) traversing the subarachnoid space from the spinal canal to the skin surface where it will form a placode.
D. Axial T1-weighted image at a lower level shows the placode (p) at its dorsal extent, covered by the wet cloth (asterisk).

Barkovich_Chap09.indd 865 5/6/2011 10:28:26 PM


866 Pediatric Neuroimaging

FIG. 9-7. Hemimyelocele. A. Sagittal T1-weighted image shows a repaired myelomeningocele (curved arrows) at the thoracolumbar level and focal syrin-
gohydromyelia (straight arrows) in the cervicothoracic spinal cord. B. Axial T1-weighted image reveals a SCM in the thoracic region. The right hemicord
(arrow) is dilated by hydromyelia. C. At the level of the myelomeningocele repair, the right hemicord is a flattened placode (small arrows), which is tethered
to the dura and epidural fat. The smaller left ventral hemicord (large arrow) was not appreciated at the time of repair.

HemiOSD of the two hemicords exhibits a small OSD that tends to lie on one
Cameron (115), Emery and Lendon (116), and Pang (117) have shown side of the midline, whereas the other hemicord is either normal, teth-
that between 31% and 46% of patients with OSD have associated SCM. ered by a thickened filum terminale (Fig. 9-7), or has a smaller OSD
The spinal cord may be split above (31%), below (25%), or at the same at a much lower level. The two hemicords usually lie in separate dural
level (22%) as the OSD (116). In addition to the patients with frank tubes that are separated by a fibrous or bony spur. Occasionally, the two
SCM, 5% of patients with OSD have a duplication of the central canals hemicords lie within a single dural tube, which becomes deficient at the
of the spinal cord cephalic to and at the level of the placode, indicating level of the hemiOSD. In general, affected patients have impaired neu-
a mild form of splitting that is insufficient to affect the gross contour rological function on the side of the hemiOSD but normal or nearly
of the cord (116). normal function on the normal side (118). Imaging studies (Fig. 9-7)
The hemiOSD, a special form of OSD with SCM, is observed in will show the splitting of the spinal cord, the extent and symmetry (or
less than 10% of patients with OSD (116,118). In the hemiOSD, one lack thereof) of the OSD, the presence or absence of the bony spur,

Barkovich_Chap09.indd 866 5/6/2011 10:28:27 PM


Chapter 9 • Congenital Anomalies of the Spine 867

and any other anomalies that may alter the surgical approach. SCM is 1 cm/s during CSF diastole (at which time, CSF is moving rostrally
discussed more fully later in this chapter. at a velocity of up to 2 cm/s) and moves rostrally during CSF systole
(122). In patients with spinal cord tethering, this motion is damped
Postoperative Complications and the cord velocity is reduced (121,122). However, it is not trivial
After repair of an OSD, patients usually have a stable neurological defi- to calibrate your MR scanner in order to make these measurements
cit. Deterioration in neurologic function, therefore, suggests the pres- accurately. In addition, the presence of scoliosis may reduce cord
ence of a complication. There are five major causes of postoperative motion in the absence of tethering. Moreover, the sensitivity and
neurological deterioration in this group of patients: (i) the spinal cord specificity of the phase contrast examination in making this diagno-
and placode may be tethered by scar or by a second, previously unrec- sis has still not been established in large scale trials. Therefore, this
ognized malformation; (ii) the dura may be pulled too tightly when technique is not widely used at the time that this edition is being
approximated over the placode, forming a constricting dural ring; (iii) written.
the spinal cord may be compressed by a dermoid or epidermoid tumor Associated malformations are common. SCM (Figs. 9-7 and 9-8)
or an arachnoid cyst; (iv) ischemia may occur because of vascular com- or dermal sinuses are seen in up to 46% of patients with OSD. Hydro-
promise; (v) there may be syringohydromyelia (119,120). cephalus and the Chiari II hindbrain malformation frequently develop
Retethering of the cord by scar is a diagnosis that essentially can in the absence of prenatal repair. Therefore, patients with neurological
only be made by exclusion of other causes. The purpose of imaging deterioration after OSD repair must have their entire craniospinal axis
patients after the repair of OSD is to identify one of these causes. evaluated. We typically perform sagittal and axial T1- and T2-weighted
If no other cause for neurological deterioration is identified, the study of the brain, followed by a coronal study of the entire spine (to
clinical diagnosis of retethering is made. It is possible that the diag- assess for vertebral anomalies and a split spinal cord malformation).
nosis of retethering may be suggested or verified by demonstrating Finally, sagittal T1- and T2-weighted images are obtained of the entire
diminished motion of the cervical spinal cord by phase contrast MRI spine, with sagittal FLAIR and axial T1- and T2-weighted imaging of
(121,122). The normal spinal cord moves caudally at a velocity of the lumbar spine.

FIG. 9-8. Patient with multiple causes of


neurologic deterioration after myelom-
eningocele repair at birth. A and B. Sagittal
T1-weighted images show multiloculated
hydromyelia (arrows) in the lower cervical
and upper thoracic spinal cord. C and D.
Sagittal T1-weighted image in the lower
thoracic region shows thinning of the cord
(small arrows), another possible cause of
neurologic deterioration, and a large
bony spur (large arrow). E and F. Axial
T1-weighted images show SCM, with two
hemicords (black arrowheads) and a bony
spur (white arrow). The right hemicord is
dilated by hydromyelia.

Barkovich_Chap09.indd 867 5/6/2011 10:28:28 PM


868 Pediatric Neuroimaging

Dermoids and epidermoids can appear as developmental tumors or insult. Sonography, CT myelography, or MRI can be used to locate this
as tumors that result from lumbar puncture or OSD repair (119,123– abrupt narrowing. A narrowing of the dural sac, suggesting that the
126). They may be more common following in utero OSD repair (101); meninges have been approximated too tightly at the time of the closure
however, this cannot be confirmed until the results of the multicenter of the lesion, may be difficult to demonstrate by MRI and is more easily
prospective trial are available. On imaging studies, dermoids and epi- seen by myelography and sonography.
dermoids appear as intraspinal masses that may be intramedullary or Hydromyelia develops in between 29% and 77% of patients with
adherent to the spinal cord, the roots of the cauda equina, or the wall of OSD (Figs. 9-7 and 9-8) (115,116,129). The exact frequency depends
the thecal sac (64,66,127,128). It is not possible to distinguish between upon the efficacy of treatment of the patient’s hydrocephalus (129–
dermoids and epidermoids by CT or myelography; on MRI, some 132). The hydromyelia in repaired OSD is likely a result of the pas-
dermoids will show a markedly shortened T1 relaxation time. More sage of CSF through the obex from the fourth ventricle into the central
commonly, both epidermoids and dermoids are difficult to identify by canal of the spinal cord: hydromyelia is not usually seen caudal to
MRI because they can be isointense with cerebrospinal fluid (Fig. 9-9); the placode. Hydromyelia is most easily diagnosed by MRI, where it
however, compression of the adjacent cord or displacement of nerve appears as a dilated (CSF-intensity) central canal, causing a fusiform
roots by the tumor will indicate presence of the mass (64). The mass enlargement of the cord, most frequently in the lower cervical or upper
can be confirmed by imaging using a FLAIR sequence, on which CSF thoracic region. The hydromyelia can also involve a short segment,
is hypointense and the dermoid/epidermoid is hyperintense, or by dif- typically beginning a small distance above the site of the placode, or
fusion-weighted imaging, on which the dermoid/epidermoid will have may involve the entire central canal from the cervicomedullary junc-
high signal intensity in contrast to the hypointense CSF. tion down to the placode.
Arachnoid cysts can be congenital (true cysts) or acquired (arach- Untreated hydromyelia can cause the rapid development of sco-
noid loculations), as described in Chapters 5 and 10. They can occur liosis (83,129–133). In patients with OSD and hydrocephalus, rapidly
at any level and may be dorsal or ventral to the cord. Because they are progressive scoliosis may be the sign of a nonfunctioning shunt or an
filled with CSF, they are isointense to the subarachnoid space and must encysted fourth ventricle. Because scoliosis in these patients may result
be identified on MRI by their mass effect upon the adjacent spinal cord from shunt malfunction, encystment of the fourth ventricle, or hydro-
(Fig. 9-10). Although cysts too small to exhibit mass effect will not be myelia unrelated to the above, all patients with scoliosis that is rapidly
detected, myelography is rarely necessary, as cysts that do not have progressive, has an atypical curve, or is associated with neurological
mass effect will not produce symptoms. deficits should have imaging studies of the entire craniospinal axis to
Segmental ischemic injury of the spinal cord is identified by an eliminate these treatable causes of scoliosis. MRI is the imaging study
abrupt diminution in caliber of the cord at the level of the ischemic of choice in these circumstances (83,133,134).

FIG. 9-9. Epidermoid tumor in a patient who under-


went fetal myelomeningocele repair. A–D. The epi-
dermoid (arrows) can be difficult to detect by routine
sagittal and axial T1 and T2 MR sequences. E. Sagit-
tal FLAIR with fat suppression shows the epidermoid
clearly (arrow). Diffusion imaging can also be valuable
(see Chapter 7).

Barkovich_Chap09.indd 868 5/6/2011 10:28:29 PM


Chapter 9 • Congenital Anomalies of the Spine 869

FIG. 9-10. Patient who had myelomeningocele repair at birth, now with worsening neurologic exam secondary to arachnoid scarring/loculations.
A. Sagittal T2-weighted image shows the conus medullaris (white arrow) extending caudally to the L-5 level. Based on this image, it cannot be determined
whether the neurologic symptoms are the result of retethering or another process. This patient emphasizes the need to image the entire spinal cord. The
levels of spina bifida (white arrowheads) are those without spinous processes. B. Sagittal T2-weighted image of the cervical and upper thoracic spine shows
a thin spinal cord (black arrows) that is dorsally displaced due to scarring and arachnoid loculations. C. Axial T1-weighted image at the midthoracic level
shows the spinal cord (white arrows) to have a crescentic shape, being displaced by the scarring and loculations.

Barkovich_Chap09.indd 869 5/6/2011 10:28:30 PM


870 Pediatric Neuroimaging

The Chiari II Malformation ascent through the sinus tract (144–146); meningitis is a risk factor for
The Chiari II malformation is an anomaly of the cervical spinal cord, poor outcome (147). Occasionally, the meningitis is chemical in nature,
brainstem, and hindbrain that is observed in varying degrees in all resulting from the release of cholesterol crystals or other contents of
patients with myelomeningoceles. Because of the close association of dermoid or epidermoid cysts into the cerebrospinal fluid (147,148).
Chiari II malformations with OSD, a brief discussion is appropriate A related entity, the spinal dermal-sinus-like stalk, was recently
in this section, in spite of the fact that this malformation is discussed reported as a discrete entity in a small cohort of patients (42). These
extensively in Chapter 5. patients did not have a tract with an epithelium-lined lumen commu-
The Chiari II malformation can be best considered as an entity in nicating with a cutaneous orifice, but rather a solid stalk of connective
which the posterior fossa is too small (135) due to collapse of the ven- tissue, fat, and occasionally neural tissue that ran from a skin dimple
tricular system from leakage of CSF through the open neural tube. As without an ostium to the intradural space or spinal cord. The dural
a result of the small bony posterior fossa and the reduced pressure of sleeve associated with the stalk was directed towards the skin surface,
the spinal subarachnoid space from the CSF leakage, normal contents unlike the dural sleeve of the dermal sinus, which is directed towards the
of the posterior fossa are distorted as they are squeezed out through an spinal cord; this observation suggests an etiology distinct from that of
enlarged foramen magnum. The brainstem is stretched inferiorly and the dermal sinus. The authors postulate that disjunction between cuta-
narrowed in the anteroposterior diameter, often lying at the level of the neous and neural ectoderm occurred, but that intervening mesodermal
foramen magnum or in the cervical spinal canal. The cervical spinal cord cells may have formed a tight, persistent connection between the two.
is displaced inferiorly and the upper cervical nerve roots have to ascend
Pathology
toward their respective neural foramina. The medulla is also displaced
inferiorly. In 70% of patients, it folds caudally at the cervicomedullary Pathologically, dermal sinuses are thin, epithelium-lined channels
junction, dorsal to the cervical spinal cord (which is tethered by the den- that course inward from the skin surface through the subcutaneous
tate ligaments and therefore limited in its vertical descent), forming a tissues, extending into the spinal canal in 50% to 70% of cases. The
characteristic cervicomedullary kink (Figs. 5-162 and 5–163). The cer- sinus may reach the dura without passing through it; in such cases,
ebellar vermis often herniates inferiorly, forming a tongue of tissue pos- the dura and arachnoid are tented dorsally at the attachment of the
terior to the medulla that usually extends down to the C-2 or C-4 level. sinus tract to the dura. This tenting may be the only manifestation
Rarely, it extends down into the upper thoracic segments of the canal. of the sinus tract on myelography. If they pass through the dura, the
The cerebellum wraps around the brainstem (Fig. 5-163). The fourth sinuses may empty into the subarachnoid space or may traverse the
ventricle is vertical in orientation (Figs. 5-162 and 5–163), extending subarachnoid space to terminate within the conus medullaris, filum
inferiorly between the medulla and cerebellar vermis; occasionally, it terminale, a nerve root, a fibrous nodule on the dorsal aspect of the
extends down below the medulla, posterior to the cervical spinal cord, cord, or a dermoid or epidermoid tumor (Fig. 9-11) (149–151). About
in a cyst-like fashion. The quadrigeminal plate is stretched posteriorly half of dorsal dermal sinuses end in dermoid or epidermoid tumors.
and inferiorly (Figs. 5-161 and 5–162). The small posterior fossa and the
downward herniation of its contents result in a low-lying, abnormally
vertical tentorium cerebelli (136–141). Many of these changes resolve
after fetal repair of the OSD, as discussed in Chapter 5.
Dorsal Dermal Sinuses (also considered as a complex
spinal dysraphism)
Clinical Features
Dorsal dermal sinuses are epithelium-lined tubes that extend inward from
the skin surface for varying distances and frequently connect the body
surface with the central nervous system or its coverings. This anomaly
seems to result from a focal area of incomplete disjunction of cutaneous
ectoderm from neural ectoderm during the process of neurulation (Fig.
9-2). When the spinal cord later becomes surrounded by mesenchyme and
undergoes its relative ascent with respect to the spinal column, this adher-
ence remains and forms a long, epithelium-lined tract. The incidence of
dermal sinuses is low in the cervical region where the neural folds first
fuse into the primitive neural tube, and relatively great in the lumbosacral
and occipital regions, which are the last portions of the neural tube to
close. In a series of 120 cases of dorsal dermal sinuses, 1 was sacrococ-
cygeal, 72 were lumbosacral, 12 thoracic, 2 cervical, and 30 occipital; the
remaining 10 were mostly ventral to the skull and spinal column (142).
Dermal sinuses in the frontonasal region are discussed in Chapter 5.
Dorsal dermal sinuses occur equally frequently in males and females
and are usually detected any time from early childhood through the
third decade of life. Physical examination reveals a midline (rarely para-
FIG. 9-11. Schematic of dorsal dermal sinus. A tuft of hair, nevus, or heman-
median) dimple or pinpoint ostium that is frequently associated with
gioma frequently marks the ostium of the sinus. The dura is often tented when
a hyperpigmented patch, hairy nevus, or capillary angioma (143). The the sinus penetrates the dura. The sinus may terminate in a CNS structure, the
patients become symptomatic either by infection or because of com- dura, or external to the dura. Dermoid or epidermoid tumors develop along
pression of neural structures by an associated dermoid or epidermoid the course of the sinus in 50% of affected patients. (Reprinted from Barkovich
tumor. Meningitis or abscesses in the subcutaneous, epidural, subdural, AJ, Edwards MSB, Cogen PH. MR evaluation of spinal dermal sinus tracts in
subarachnoid, and/or subpial spaces may develop as a result of bacterial children. Am J Neuroradiol 1991;12:123–129, with permission.)

Barkovich_Chap09.indd 870 5/6/2011 10:28:31 PM


Chapter 9 • Congenital Anomalies of the Spine 871

Conversely, 20% to 30% of dermoids and epidermoids are associated two spinous processes. In other cases, the sinus may be associated with
with dermal sinus tracts (150–154). Dermal sinuses can be midline or a groove in the upper surface of the spinous process and lamina of the
paramedian in location. Those with midline orifices are usually associ- vertebra, a hypoplastic spinous process, a single bifid spinous process,
ated with midline dermoid tumors. Paramedian orifices are more com- focal multilevel spina bifida, or a laminar defect (145,150,152).
monly associated with epidermoids, which can be located laterally in When a dermoid or epidermoid tumor is present, adjacent nerve
the epidural, subdural, or subarachnoid spaces. In some patients, the roots are frequently bound down to the capsule of the cyst. The
dermal sinus courses horizontally beneath the skin to the dura mater. cord may be displaced and compressed by extramedullary dermoids
In others, it courses subcutaneously for some distance before reaching or epidermoids, or expanded by intramedullary lesions (20,64).
the dura and then ascends within the spinal canal to the level of the Nerve roots may be clumped because of adhesive arachnoiditis from
conus medullaris. The exact course of the sinus seems to vary from previous infections or rupture of a dermoid. If abscesses form, they
patient to patient. Therefore, the complete course of each dermal sinus may remain confined near the tract and entry site of the sinus or may
must be determined by narrow, contiguous sections above and below extend cephalad and caudad for considerable distances (64,146,155).
the level of the external orifice.
The bony abnormalities associated with dermal sinuses are vari- Imaging
able. Bone abnormalities may be absent if the dermal sinus extends Sonography, CT, and MRI can all demonstrate the subcutaneous and
intraspinally through a ligamentous defect at the interspace between extracanalicular extent of the dermal sinus tract (Figs. 9-12 to 9-14).

FIG. 9-12. Dorsal dermal sinus with dermoid


adjacent to conus medullaris. A. Sagittal
T1-weighted image shows the dermal sinus
(black arrowheads) coursing through subcu-
taneous fat and into the subarachnoid space.
Part of the subarachnoid portion of the tract is
bright (white arrowheads) because it contains
fat. The hyperintense circle (white arrow) at the
skin surface is a vitamin E capsule marking the
opening of the sinus. B. Sagittal T2-weighted
image shows the sinus tract (large black arrow-
heads) more clearly as it courses through the
subarachnoid space. Incidentally noted is a
thickened filum terminale (small black arrows),
likely related to the low level (bottom of L-3, one
full vertebral body level too low) of the conus
medullaris. C and D. Axial T1-weighted images
show smudgy soft tissue intensity (white arrows)
dorsolateral to the conus medullaris. At surgery,
this was found to represent dermoid tumor.

Barkovich_Chap09.indd 871 5/6/2011 10:28:32 PM


872 Pediatric Neuroimaging

FIG. 9-13. Lumbar dorsal dermal sinus with intramedullary


dermoid. A and B. Sagittal T1-weighted and T2-weighted
images show an intramedullary mass (asterisk) that has sim-
ilar signal intensity to CSF. Note that the sinus tract is not
within the portion of the spine visualized on these images.
C. Sagittal T2-weighted image of the lumbar spine shows a
hypointense curvilinear structure (black arrowheads) cours-
ing down the dorsal aspect of the subarachnoid space, then
terminating (black arrow) at the mid-L-5 level. As seen in (D),
this is the level where the tract exits through the dermal sinus.
D. Sagittal T1-weighted image shows the dermal sinus cours-
ing (white arrowheads) below the L-5 spinous process and
(black arrowheads) through the subcutaneous fat. The ostium
is marked by the small white arrow.

Barkovich_Chap09.indd 872 5/6/2011 10:28:33 PM


Chapter 9 • Congenital Anomalies of the Spine 873

FIG. 9-14. Dorsal dermal sinus with infected dermoid and epidural abscess.
A and B. Sagittal T1-weighted images show the importance of proper windowing.
In (A), filmed with wide windows, the subcutaneous portion of the sinus (black
arrows) is easily seen. In (B), windowed to see the intraspinal portions of the sinus
and dermoid, the subcutaneous portion of the tract cannot be identified. In (B),
the lower aspect of the thecal sac (arrows) is very heterogeneous. However, it can-
not be determined from this study whether the heterogeneous signal is the result
of clumping of nerve roots from infection or from (epi)dermoid tumor. At surgery,
this patient was found to have a large, infected epidermoid at the L-5 to S-1 levels
and an epidural abscess at the L-3 to L-4 levels. C and D. Sagittal T2-weighted (C)
and fat-suppressed postcontrast T1-weighted (D) images allow better distinction
between the epidural (e) and intradural (i) components of the infection. E. Axial
T1-weighted image at the L-5 level. The contents of the thecal sac (white arrows)
are of heterogeneous high signal intensity. This was found to be an infected der-
moid tumor at surgery. The black arrowhead points to the sinus tract coursing
through subcutaneous fat.

MRI best demonstrates intramedullary dermoid and epidermoid nerve roots around any mass, giving a strong indication of its presence.
tumors (Figs. 9-13 and 9-14). T1-weighted images with wide window The use of strongly T1-weighted images (64), FLAIR images (156,157),
settings best show the subcutaneous portion of the sinus tracts; nar- or diffusion-weighted images (157,158) will help to identify intraspinal
row windows may make the tract impossible to see (Fig. 9-14). The extramedullary (epi)dermoids, which may have signal intensity identi-
intrathecal portions of the tracts are small and hypointense; therefore, cal to CSF on standard T1- and T2-weighted images. The administration
they are essentially invisible on noncontrast T1-weighted MR images of paramagnetic contrast (which results in enhancement of some sinus
unless fatty tissue is present (Fig. 9-12). Thin section T2-weighted tracts [60]) and use of fat-suppression techniques may help to identify
RARE (fast spin echo, turbo spin echo) images in the sagittal and axial the sinus tract if granulation tissue is present due to prior infection or
planes best show the sinus tract as a hypointense linear or curvilinear inflammation (Fig. 9-15). Carefully performed ultrasound can show
structure on MRI (Figs. 9-12 and 9-13) and will show the deviation of the intradural portion of the sinus in young infants. CT with intrathecal

Barkovich_Chap09.indd 873 5/6/2011 10:28:34 PM


874 Pediatric Neuroimaging

FIG. 9-15. Contrast enhancement of thoracic dermal sinus. A and B. Sagittal T2-weighted images show the hypointense dermal sinus (white arrows)
coursing through the subcutaneous fat and the distortion of the dorsal spinal cord (black arrows) where the sinus tract inserts. C. Sagittal postcontrast
T1-weighted image with fat suppression shows the enhancement of the deeper portion of the oblique sinus (white arrow). D and E. Axial T2-weighted
images show the lack of complete closure of the dorsal thoracic spinal cord just above the level of the tract, (black arrow in D) and the hypointense tract
through the subcutaneous fat (black arrows in E).

contrast best demonstrates the intraspinal portion of the dermal sinus may be intradural, extradural, or both. On T2-weighted MR images of
and small extramedullary (epi)dermoids in older children. Thus, in the spinal cord, abscesses appear as areas of high signal intensity, often
those rare cases in which the intraspinal anatomy is not clearly defined with a ring of low signal intensity. T1-weighted images show low signal
after high-quality MRI, a CT myelogram should be considered. intensity; these areas are usually poorly defined on precontrast images
As mentioned, extramedullary (epi)dermoids may be difficult to because of surrounding edema. Administration of paramagnetic con-
visualize with MRI using standard T1- and T2-weighted sequences; trast reveals ring-enhancing masses (159). Diffusion-weighted images
ruptured and infected dermoid and epidermoid tumors are even more show reduced diffusion within the abscess. Every attempt should be
difficult to identify, as no distinct mass is seen. Instead, the subarach- made to show the associated dermal sinus.
noid space has a “smudgy,” slightly heterogeneous appearance (Fig.
9-14) that cannot be differentiated from arachnoiditis by MR tech- Cervical Myelocystocele and Cervical Myelocele (may
niques. FLAIR and diffusion-weighted images can be very helpful in also considered as CSD with a subcutaneous mass)
this setting (156,157); they will help the surgeon know where to look to The myelocystocele is a malformation in which a dilated central canal
remove as much tumor as possible. protrudes dorsally through a bony spina bifida (Fig. 9-16). Although some
For detection of associated intraspinal abscesses, precontrast and authors consider these to be synonymous with cervical myelomeningo-
postcontrast MRI is the imaging study of choice (Fig. 9-14). The abscess celes (160) or to be part of a continuity with cervical meningocele in

Barkovich_Chap09.indd 874 5/6/2011 10:28:35 PM


Chapter 9 • Congenital Anomalies of the Spine 875

cord. Other authors differentiate cervical myelocystoceles from cervical


myeloceles, but consider the latter to be meningoceles (162). However,
meningoceles by definition do not contain neural tissue, and the cervical
myeloceles clearly have a stalk of neural tissue extending through the spi-
nal bifida, as shown by Rossi et al. (161). Cervical myelocystoceles should
also be differentiated from terminal myelocystoceles, which are anoma-
lies of the caudal cell mass and are located at the lumbosacral level. Ter-
minal myelocystoceles are discussed later in this chapter.
Patients with cervical myelocystoceles and those with cervical
myeloceles present as neonates with a dorsal midline cystic mass, usu-
ally at the cervical or cervicothoracic level. Most of the mass is covered
by full-thickness skin, whereas a tough, violaceous membrane covers
the apex. The neonates are typically alert and vigorous, with normal
neurologic exams and no evidence of extraneural congenital malfor-
mations (163). Occasionally, infants have mild abnormalities of muscle
strength or of tone (160,162). These deficits become more noticeable
during the first year of life, the patients becoming spastic or weak in
the arms or legs (164). By the end of the first decade, some sort of
motor deficit is usually present, often requiring orthopedic interven-
tion (164). The head size is often enlarged (162).
The diagnosis is made by identification of spinal cord tissue pro-
truding dorsally through a bony spina bifida into the dorsal subcutane-
ous soft tissues (Fig. 9-17). In a myelocystocele, the central canal of the
spinal cord is enlarged, and the area containing this syrinx protrudes
posteriorly through the spinal bifida. Fluid may not be continuous all
FIG. 9-16. Schematic of myelocystocele. This is an occult, skin-covered, the way into the subcutaneous region, but dysplastic neural tissue will
spinal dysraphism in which the spinal cord (which has a syringohydromy- be found within the leptomeninges in the subcutaneous tissue beneath
elia) and the arachnoid are herniated through a posterior spina bifida. The a thickened layer of squamous epithelium. The fluid in the enlarged
cyst is in continuity with the central canal of the spinal cord. Myelocysto- central canal may be multiloculated. In cervical myeloceles, soft tissue
celes can occur at any level. Localized expansion of the subarachnoid space and fluid may be present in the dorsal sac that protrudes through the
is not a necessary component and is uncommon in myelocystoceles that spina bifida, but no cyst is seen within the spinal cord (161,162). Care
occur at locations other than the cord terminus. should be taken to examine the entire spinal axis, as other malforma-
tions, such as dermal sinuses, SCMs, and Chiari II malformations may
which a fibrovascular stalk extends from a nearly normal-looking spinal be present (162,165). The diagnosis can be made by MR or by ultra-
cord through a spina bifida to form a skin-covered mass (161), we con- sound. CT myelography is less useful, because of its invasive nature and
sider the latter to be a true cervical myelocele and an entity distinct from radiation exposure and because it does not detect the focally enlarged
the myelocystocele. The latter, by definition should be an encysted spinal central canal.

FIG. 9-17. Cervical myelocystocele. A. Sagittal T1-weighted image through the cervical spine shows syringohydromyelia of the cord with multiple septa-
tions within the syrinx (white arrowheads). The ependyma-lined cyst is seen to extend posteriorly through a spina bifida into the subcutaneous tissues (black
arrowheads) forming a skin-covered dorsal mass (white arrows). B. Axial T1-weighted image at the lower level of the dorsal cyst shows the loculations within
the spinal cord and the cord extending dorsally through a bony spina bifida. The subarachnoid space ventral to the spinal cord is expanded.

Barkovich_Chap09.indd 875 5/6/2011 10:28:36 PM


876 Pediatric Neuroimaging

Disorders Resulting from Premature Intradural Lipomas (Also classified as a simple


Disjunction—Spinal Lipomas dysraphic state)
Intradural lipomas, which constitute slightly less than 1% of primary
Concepts of Lipomas
intraspinal tumors, are juxtamedullary masses that are totally enclosed
Spinal lipomas are masses of fat and connective tissue which appear in an intact dural sac. They are slightly more frequent in females.
at least partially encapsulated and which have a connection with Affected patients present at three age peaks: the first 5 years of life
the leptomeninges or the spinal cord (166). Grossly, the lipomas are (24%), the second and third decades (55%), and the fifth decade (16%)
homogeneous masses of mature fat cells that are separated into glob-
ules by strands of fibrous tissue. The proportion of fibrous tissue is
much greater near the interface of the cord and lipoma and consider-
ably less near the skin surface (151,167). Calcification and ossification
are sometimes seen (151,168), as are muscle fibers, nerves, glial tissue,
arachnoid, ependyma, and many other types of tissue (169).
Spinal lipomas can be divided into three principal groups: (a) intra-
dural lipomas (3%–5%); (b) lipomas with dorsal defect, encompassing
lipomyeloceles and lipomyelomeningoceles (75%–85%); and (c) lipomas
deriving from the caudal cell mass (10%–15%). Group 2 can be divided
into dorsal, caudal, and combined or “transitional” lipomyeloceles/lipo-
myelomeningoceles (170,171). Group 3 can be further divided into (a)
terminal lipomas, which are at the caudal part of the thecal sac and are
always associated with a thickened filum terminale, and (b) fibrolipo-
mas of the filum terminale. Both are almost always associated with teth-
ering of the spinal cord. Terminal lipomas and fibrolipomas of the filum
terminale are best classified as anomalies of the caudal cell mass. Termi-
nal lipomas are discussed in this section because their lobulated, fatty
appearance is so similar to that of intradural lipomas and lipomas with
dorsal defect. Fibrolipomas are discussed later in this chapter, in the sec-
tion on “Anomalies of the Caudal Cell Mass.” In contrast to the anoma-
lies of the caudal cell mass, intradural lipomas and lipomas with dorsal
defect are postulated to result from a premature separation of cutaneous
ectoderm from neural ectoderm during the process of neurulation (Fig.
9-2). This premature separation permits the surrounding mesenchyme
to enter the ependyma-lined central canal of the neural tube, which has
not yet closed. The presence of the mesenchyme impedes the closure
of the neural folds and results in an open neural placode at the site of
premature disjunction. Moreover, the mesenchyme that enters the cen-
tral canal differentiates into fat. This same mesenchyme, if exposed to
the exterior of the cord, differentiates into meningeal tissue, bone, and
paraspinous muscles. The junction between the internal and external
surfaces of the cord therefore determines the junction between the
meninges and the fat. The faulty disjunction between neural ectoderm
and cutaneous ectoderm that results in the formation of spinal lipomas
may also explain the frequent association of lipomas with dorsal dermal
sinuses, which result from a focal area of faulty disjunction.
An important concept in the imaging evaluation of spinal lipomas
is that these lipomas are largely but not exclusively composed of nor-
mal fat (169). More importantly, fat cells dramatically increase in size
during infancy (172). In fact, the proportion of body fat increases from
14% of body weight at birth to 25% at age 6 months (173). As a result, FIG. 9-18. Schematic illustrating spinal lipomas and lipomyelomenin-
lipomas may be missed or be considered inconsequential when imaging is goceles. A. Subpial-juxtamedullary lipoma. The spinal cord is open in the
performed in the fetal or neonatal period, only to be found much larger at midline dorsally with the lipoma situated between the nonapposed lips of
a subsequent imaging study (174). This potential for growth should be the placode. B. Lipomyelocele. This lesion is very similar to a myelocele with
kept in mind when deciding on the optimal time for imaging neonates two additional characteristics. The lipoma lies dorsal to and is attached to
with spinal anomalies. Another aspect of this concept is that a spinal the surface of the placode. This lipoma is continuous with subcutaneous
lipoma can diminish in size if the patient loses weight (175). There- fat. Equally important is the fact that an intact skin layer overlies the lesion,
making this an occult spinal dysraphism. C. Lipomyelomeningocele with
fore, control of body weight may, in some instances, be a conservative
rotation of the neural placode. When the lipoma is asymmetric, it extends
method of managing affected patients (175). into the spinal canal and causes the ventral meningocele to herniate pos-
MRI is the imaging modality of choice for the evaluation of teriorly and the dorsal surface of the placode to rotate to the side of the
patients with spinal lipomas. The short T1 relaxation time of fat results lipoma. This rotation brings the contralateral dorsal root (in this case the
in characteristic high signal intensity of the lipoma on T1-weighted right dorsal root) into the midline posteriorly, putting it at increased risk
sequences. MRI allows the full extent of the lipoma, and its relationship for surgical trauma. Moreover, the left roots are markedly shortened by this
to the neural placode, spinal cord, and roots of the cauda equina, to be rotation, limiting the mobility of the cord and impeding the neurosurgeon
fully evaluated. from completely untethering it.

Barkovich_Chap09.indd 876 5/6/2011 10:28:37 PM


Chapter 9 • Congenital Anomalies of the Spine 877

(176). Patients harboring cervical and thoracic intradural lipomas most


frequently present with a slow, ascending monoparesis or paraparesis,
spasticity, cutaneous sensory loss, and defective deep sensation. Radic-
ular pain is uncommon. Patients with lumbosacral intradural lipomas
may present with flaccid paralysis of the legs and sphincter dysfunction
(176) or predominately pain (177). Symptoms may be exacerbated by
pregnancy (178). The skin and the adjacent subcutaneous tissues over-
lying the lipoma are most often normal.
Intradural lipomas develop most commonly in the cervical and
thoracic spine (cervical 12%, cervicothoracic 24%, thoracic 30%) but
may develop anywhere in the spinal cord or cauda equina (171,179,180).
Most develop along the dorsal aspect of the cord, but 25% are lateral or
anterolateral (151,153,168,176,181,182). Hydromyelia and syringomy-
elia are present in approximately 2%.
Intradural lipomas are actually subpial in location (Fig. 9-18)
(176). The spinal cord is open in the midline dorsally with the lipoma
situated in the opening between the unopposed lips of the placode.
The lipoma fills the space between the central canal and the pia, which
is frequently lifted from the cord surface as the lipoma projects into the
subarachnoid space. In the 45% of patients in whom the lipoma is exo-
phytic, the exophytic component tends to be at the upper or lower pole
of the lipoma. Although “intramedullary” lipomas have been rarely FIG. 9-19. Intradural lipoma. Axial CT image shows a low-attenuation
reported, no lipoma has been described that is fully encompassed by mass (arrows) in the dorsal aspect of the spinal canal.
cord (20,176).
Although the bony spinal canal can be normal in patients with
intradural lipomas, focal enlargement of the spinal canal and, occa- on T1-weighted MR images by their lobulated shape and high signal
sionally, the adjacent neural foramina is more common (40). Some- intensity (Fig. 9-20). Their fatty nature can be confirmed, if necessary,
times, a localized, narrow spina bifida is observed at the level of the by use of fat-suppression pulses. MRI best identifies compression of
lipoma; however, the bony spinal canal is usually intact (178), aiding in the spinal cord.
the distinction from lipoma with dorsal defect. There are generally no
Lipoma with Dorsal Defect (Lipomyeloceles and
segmentation anomalies of the vertebral bodies.
Lipomyelomeningoceles)
Intradural lipomas appear on imaging studies as focal, round to
oval masses that most often lie dorsal to the spinal cord (Figs. 9-19 Description and Presentation
and 9-20) and may expand the spinal canal (Fig. 9-20). Lipomas have a Lipoma with dorsal defect occurs when a lipoma that is tightly attached
very low attenuation on CT and, therefore, can be detected on studies to the dorsal surface of a neural placode extends dorsally through a
without intrathecal contrast (Fig. 9-19). They are easily identified bony spina bifida to be continuous with subcutaneous fat (40,169,183).

FIG. 9-20. Intradural lipoma. Sag-


ittal (A) and axial (B) T1-weighted
images show the hyperintense lipoma
(arrows) dorsal to the compressed spi-
nal cord. The spinal canal is expanded
by the mass.

Barkovich_Chap09.indd 877 5/6/2011 10:28:38 PM


878 Pediatric Neuroimaging

Terminal lipomas are probably identical to caudal lipomyelocele; these as many as 88% of such children eventually develop progressive neu-
lipomas attach to the cord at the conus medullaris, which is almost rologic symptoms if untreated (169,171,186,187). There is no debate
invariably low in position due to tethering, and then extend dorsally that most symptomatic patients will have progression of symptoms if
through a sacral spina bifida. They constitute approximately 20% of untreated (169,171,186,187).
skin-covered lumbosacral masses and between 15% and 50% of cases Lipomas with dorsal defect usually occur in the lumbosacral region
of closed spinal dysraphism (31,184,185); they account for about 75% of the cord and tether the cord at that level. Anatomically, lipomyelom-
to 85% of spinal lipomas (31). Patients are typically female. Affected eningoceles and lipomyeloceles are very similar to myelomeningoceles
patients most commonly present with a rather soft lumbosacral mass, and myeloceles, respectively, with two important additional character-
less commonly with sensory loss in the sacral dermatomes, bladder istics: (a) a lipoma is attached to the dorsal surface of the placode, and
dysfunction, lower extremity weakness, orthopedic deformities of the (b) an intact skin layer overlies the lesion (closed spinal dysraphism).
foot, scoliosis, and/or leg pain (169,171,186,187). When a lumbosacral
mass is present, patients typically come to medical attention before the Imaging Characteristics
age of 6 months. If the mass is subtle, or no mass is present, clinical pre- Patients with lipomyeloceles have a normal-sized subarachnoid space
sentation is typically the result of neurologic or urologic deficits that ventral to the placode; the cord and the junction between the placode
are noticed between the ages of 5 and 10 years; patients may occasion- and the lipoma, therefore, are within the spinal canal (Figs. 9-21 and
ally go undetected into adulthood (171,187). As children with lum- 9-22). The lipoma extends dorsally through the spina bifida and is con-
bosacral masses are usually detected early, 40% to 45% of such children tinuous with subcutaneous fat. The focus of continuity with extraspinal
are neurologically normal on initial examination (40,169,183). It is not fat is sometimes quite small (Fig. 9-21) but is always present. The lipoma
clear how many of these asymptomatic patients will eventually become appears as an echogenic mass on ultrasound, as a mass of very low atten-
symptomatic, with different series reporting that as few as 16% and uation on CT, and as a mass with short T1 and T2 relaxation times on

FIG. 9-21. Dorsal lipomyelocele containing well-formed bone. A and B. Sagittal T1-weighted and T2-weighted images show a low-lying conus medullaris
and a lipoma (white arrows in A) dorsal to the spinal cord at the L-2 level. There is a dorsal spina bifida at L-2 to L-4 (white arrows in B). C and D. Axial SE
T1-weighted images show the lipoma (white arrows) lying immediately dorsal to the spinal cord, which is flattened in appearance. The black arrows in D
point to a bony structure better identified in E.

Barkovich_Chap09.indd 878 5/6/2011 10:28:39 PM


Chapter 9 • Congenital Anomalies of the Spine 879

MRI. The spinal cord can have a number of different shapes depending
upon the morphology of the lipoma. It is usually crescent-shaped, arch-
ing over the ventral surface of the lipoma. However, if the intracanalicu-
lar portion of the lipoma extends laterally on both sides of the cord, the
placode may appear pointed, with the tip of the point directed posteri-
orly between the lateral extensions of fat. Rarely, well-formed bones (Fig.
9-21) or hamartomatous masses (188) are found in the lipoma.
In patients with lipomyelomeningoceles the spinal subarachnoid
space is expanded ventrally, and the placode, cord, subarachnoid
space, and dura extend dorsally through the spina bifida (Figs. 9-23
and 9-24). Similar to lipomyeloceles, a lipoma is attached to the dorsal
aspect of the placode; this lipoma extends posteriorly to blend with the
subcutaneous fat. Depending upon the degree of posterior herniation
of the spinal cord, the junction between the placode and lipoma may
be entirely posterior to the spinal canal, or partly within and partly
posterior to the canal. The dura is deficient in the zone of the spina
bifida and does not pass posterior to the neural elements to form a
closed dural tube as it does elsewhere. Instead, the dura is attached to
the lateral border of the neural placode, posterior to the dorsal nerve
roots as they emerge from the cord (Fig. 9-18). A continuous layer of
pia-arachnoid is continuous superiorly and inferiorly with the suba-
rachnoid space, lining the deep surface of the dura and the ventral
surface of the placode (Fig. 9-18). The dorsal surface of nervous tissue
is, therefore, attached to the lipoma, external to the dural sac and sub-
arachnoid space (40,168,183). The dorsal and ventral nerve roots exit
FIG. 9-21. (continued) E. Lateral plain film of the spine shows a well- from the ventral surface of the placode (Fig. 9-18B). The nerve roots
formed bone (arrow; surgery showed it to be a digit) arising within the may course through fat as they course toward their neural foramina
lipoma, dorsal to the L-2 vertebra (white arrow). (169,171).

FIG. 9-22. Transitional lipomyelocele. A. Sagittal T1-weighted image shows a large fatty mass (arrowheads) over the lumbosacral spine and a large intraspi-
nal lipoma (L). The conus medullaris is low-lying, at the L-4 level. B–D. Axial T1-weighted images show the placode (white arrowheads in B) as a curvilinear
region of tissue ventral to the lipoma.

Barkovich_Chap09.indd 879 5/6/2011 10:28:40 PM


880 Pediatric Neuroimaging

FIG. 9-22. (Continued) In C, the lipoma (white arrows) is seen to herniate dorsally out of the spinal canal through the bony spina bifida. In D, the fat of
the lipoma merges with subcutaneous fat; the white arrows demarcate the point of merging.

FIG. 9-23. Transitional lipomyelomeningocele. A and B. Sagittal T1-weighted and T2-weighted images show the spinal cord tethered by a large lipoma
that extends from the subcutaneous fat through a spina bifida into the dorsal subarachnoid space. A portion of the dilated subarachnoid space can be seen
extending into the subcutaneous fat (arrows). Note the hydromyelia (black arrowheads in B), seen in about 25% of patients with tethered spinal cords. C and
D. Axial T1-weighted images show the subcutaneous fat (white arrows) herniating into the spinal canal through the spinal bifida. The placode (black arrows)
is rotated to the right, contralateral to the fat, which is invaginating on the left (white arrows in D). E. Axial T2-weighted imaging at a slightly more rostral
level shows the superior portion of the placode-lipoma interface, as well as dorsal and ventral nerve roots (black arrowheads) arising from the placode.

Barkovich_Chap09.indd 880 5/6/2011 10:28:41 PM


Chapter 9 • Congenital Anomalies of the Spine 881

FIG. 9-24. Lipomyelomeningocele. A and B. Sagittal T1- and T2-weighted


images show the tethered spinal cord which is attached to a dorsal lipoma
through a posterior osseous defect. The meningocele (m) can be seen extend-
ing into the subcutaneous fat. Fat extends into the spinal canal (black arrow in
A) and adheres to the dorsal surface of the cord. The central canal of the spinal
cord is dilated. C. Axial T1-weighted image rostral to the lipoma shows the
hydromyelia (arrow) within the spinal cord. D. The intraspinal lipoma (white
arrow) is present at this more caudal level. The dorsal and ventral nerve roots
are seen originating from the placode coursing ventrally toward their neural
foramina. The posterior elements are bifid at this level and the meningocele
(black arrow) extends in to the subcutaneous fat.

The dorsal surface of the placode, adjacent to the lipoma, has no meningocele to herniate posteriorly and the dorsal surface of the pla-
ependymal lining; instead it is covered by a relatively thick layer of con- code to rotate to the side of the lipoma (Fig. 9-23). Such rotation and
nective tissue mixed with islands of glial cells and smooth muscle fibers herniation brings the contralateral dorsal roots and dorsal root entry
(168). The lipoma lies immediately external to the connective tissue zones into the midline posteriorly, putting them at increased risk for
in the extradural space; it may be localized to the levels of the spina surgical trauma (Figs. 9-23 and 9-24). Moreover, the nerve roots are
bifida or may extend upward along the dorsal placode and underlie an unequal in length; those that originate posterior to the spinal canal
outwardly normal-appearing spinal canal. Moreover, the lipoma can are very long, whereas those that originate from the intracanalicular
enter the central canal of the spinal cord and pass upward within it portion of the placode are very short. These short nerve roots limit
to form an apparently isolated intramedullary lipoma at higher levels. the mobility of the cord and impede complete untethering of the cord
The lipoma can also extend upward within the extradural portion of (169,171).
the spinal canal, forming an apparent epidural lipoma. In both of these Some lipomas attach to the spinal cord at the level of the conus
situations, the use of MR, especially in the sagittal plane, helps to trace medullaris and course caudally through the lumbosacral spinal canal,
the lipoma to its caudal origin near the myeloschisis (Fig. 9-25). exiting through a bony spina bifida, usually at the lower lumbar or sacral
At the level of the placode, the spinal canal is generally large and level, to become continuous with subcutaneous fat (Fig. 9-26). The
is associated with a posterior osseous defect (189). Butterfly vertebrae conus is almost invariably abnormally low and the distal spinal cord is
and segmental anomalies of the vertebral bodies may be found in as almost invariably thin from being stretched inferiorly (Fig. 9-26). Rota-
many as 43% of affected patients, and abnormalities of the sacrum, tion of the spinal cord and anomalies of vertebral body segmentation
including confluent sacral foramina and partial sacral agenesis, are are uncommon in this malformation, which is often called a terminal
present in up to 50% (180,189). lipoma, or a caudal form of lipoma with dorsal defect. As with tethered
The lipoma is asymmetric in approximately 40% of patients. If spinal cords of all causes, the central canal of the spinal cord is dilated
the asymmetric lipoma extends into the spinal canal, it causes the in about 20% of affected patients (82,169).

Barkovich_Chap09.indd 881 5/6/2011 10:28:42 PM


882 Pediatric Neuroimaging

FIG. 9-25. Rostral extension of lipoma through the central canal of the spinal cord. A. Sagittal T1-weighted image of the cervical spine shows a lipoma (arrows)
within the spinal cord. B. Axial proton density image confirms the location of the lipoma (arrows) within the central canal of the spinal cord. C. T1-weighted
image of the lumbar spine shows a marked spinal deformity at the site of repair of a lipomyelomeningocele. The lipoma (arrows) entered the cord at the level of
the placode and extended upward through the central canal of the spinal cord, thereby mimicking the appearance of a purely intramedullary lipoma.

Associated Malformations hamartomas, angiomas, or arachnoid cysts may be present. Thus, it is


Lipomas with dorsal defect are commonly associated with other crucial to look for other anomalies whenever lipomyelomeningoceles and
anomalies; an active search for these anomalies should always be per- lipomyeloceles are identified.
formed when evaluating the imaging studies of affected patients. Sacral
hypogenesis is seen in about 25% of patients with lipomas affecting the
conus medullaris (169). Anorectal malformations (including imperfo-
ANOMALIES OF THE CAUDAL CELL MASS
rate anus, anorectal stenosis, and anal malposition) and genital and As discussed in the embryology section at the beginning of this chapter,
urinary tract anomalies are seen in 5% to 10% of affected patients. the lowest portion of the spinal cord (the conus medullaris), the filum
If anal or genitourinary anomalies are present, the incidence of terminale, and the lower lumbar and sacral nerve roots form from the
sacral anomalies increases to more than 90% (169); these patients are caudal cell mass by a process referred to as canalization and retrogres-
best classified as having anomalies of the caudal cell mass. Terminal sive differentiation. The nearby anorectal and lower genitourinary tract
SCM may accompany lumbosacral lipomas in up to 10% of affected structures form from the cloaca, which develops simultaneously and
patients (169). Less commonly, (epi)dermoid tumors, dermal sinuses, in close topological proximity to the caudal cell mass. As a result, the

FIG. 9-26. Terminal lipomas. A. Sagittal high-resolution


ultrasound image of the lumbosacral spine in an infant
where the echogenic fatty mass (white arrows) tethers the
low-lying spinal cord. Note the associated hydromyelia.
B. Different patient. Sagittal T1-weighted image shows a fatty
mass attached to the tip of a low-lying conus medullaris. The
most caudal three levels of the cord are abnormally thin.

Barkovich_Chap09.indd 882 5/6/2011 10:28:43 PM


Chapter 9 • Congenital Anomalies of the Spine 883

FIG. 9-26. (Continued) C. Axial T1-weighted image at the level of the sacrum shows the continuity of the lipoma (black arrows), through the bony spina
bifida and paraspinous muscles, with the subcutaneous fat. D. Different patient. This newborn was imaged because of the large subcutaneous lumbosacral
fatty mass. The terminal lipoma (black arrows) is seen caudal to the low-lying conus medullaris.

anomalies listed in this section are commonly associated with anorec- age of 3 months (68,89,193). In several series involving a total of more
tal and genitourinary anomalies (45). Anomalies of the caudal spine than 1,000 patients, the most caudal aspect of the conus was observed
should always be suspected in patients with anorectal and genitourinary above the L-2 to L-3 level in greater than 98% of cases and at the L-3
anomalies and vice versa. level in less than 2% (68,89,194,195). However, in more recent work,
where complete spine MRI was performed in asymptomatic oncology
patients, allowing counting from the craniocervical junction, no nor-
Normal Conus Medullaris and Filum Terminale
mal conus medullaris terminated below the level of the midbody of
Early in embryogenesis, the spinal cord extends to the caudal end of the L-2. Therefore, if the conus medullaris terminates below the level of L-2
spinal canal. At that time, each neural segment is at exactly the same inferior endplate, it should be considered abnormal, and a search should
level as the corresponding segment of spinal canal. Each nerve root be made for a tethering mass (usually lipoma), bony spur, or thick filum.
courses directly laterally towards its neural foramen. As the embryo The normal filum terminale measures 1 mm or less in diameter at
matures, the most distal portion of the cord undergoes retrogressive the L-5 to S-1 level (20); previous editions of this book have stated the
differentiation; moreover, the vertebral bodies grow more quickly than upper limit as 2 mm, but that was based on the poorer spatial resolu-
the spinal cord. This combination of factors produces a relative ascent tion of early MR scanners (82). Some even believe that if the filum can
of the spinal cord within the spinal canal. be seen at the L-5 to S-1 level on axial T2-weighted RARE sequences, it
The exact level of the conus medullaris at the time of birth is is abnormal, but this is certainly not a widely accepted belief.
debated; however, most recent evidence indicates that the conus should
be no lower than the L-3 level by 24 weeks of gestation (190) and at its
The Terminal Ventricle
normal (adult) level by 40 postconceptional weeks (191,192), that is, at
the time of birth. It has been well established that the conus medullaris During the development of the spinal cord, the central canal is widest
is usually positioned above the middle of the L-2 vertebral level by the at the level of the conus medullaris; this widening is referred to as the

Barkovich_Chap09.indd 883 5/6/2011 10:28:44 PM


884 Pediatric Neuroimaging

the symptoms limited to the lower extremities, urinary bladder, and


bowels. The clinical symptomatology is believed to be due to stretch-
ing of nerve fibers, resulting in abnormal oxidative metabolism in the
conus medullaris and nerve roots (204).
Small amounts of fat in the filum terminale may be asymptom-
atic. Emery and Lendon (166) found incidental fat (which they termed
fibrolipomas) in the filum terminale of 6% of autopsied patients with
presumably normal spines. Fibrolipomas are also sometimes seen inci-
dentally on imaging studies of both children and adults referred for
other reasons (88,205). However, patients with filum fibrolipomas may
become symptomatic at nearly any age (52,169) (indeed, at UCSF, we
have seen patients of all ages become symptomatic from tethered spi-
nal cords with one patient developing symptoms at age 70 years), and
the percentage of asymptomatic patients decreases rather dramatically
with increasing age (169). Thus many authorities currently believe
that fibrolipomas of the filum terminale are not a normal variant, that
patients with this finding should have a careful neurological examina-
tion and urodynamic testing, and, if abnormalities are found, should
be treated surgically (169,186). There is no information available con-
cerning long-term outcome in filar cysts.
FIG. 9-27. Terminal ventricle. A and B. Sagittal ultrasound and sagittal Plain radiographs may be normal, although they typically show a
T2-weighted image show a focal prominence (arrows) of the caudalmost small defect in the posterior elements; scoliosis is present in approxi-
portion of the central canal of the spinal cord, at the level of the conus mately 20% of affected patients (51,182,206,207). Filum fibrolipomas
medullaris. If cystic and asymptomatic, the lesion should be treated as an appear on CT as small foci of hypoattenuation and on T1-weighted
anatomic variant. MRI as areas of high signal intensity and low signal intensity on
T2-weighted images within an enlarged filum (Figs. 9-28 to 9-30). The
normal filum should measure 1 mm or less in diameter. Often, the
terminal ventricle or ventriculus terminalis. This feature may persist lipoma will be in the caudal portion of the filum. Therefore, if the patient
into infancy, childhood, or even adulthood; in one series it was found has any manifestations that raise the possibility of a tethered cord (atypi-
in 2.6% of children examined (196). Occasionally, the terminal ven- cal scoliosis, urologic abnormalities, signs or symptoms of lower extremity
tricle takes on a cystic appearance that can mimic a frank cyst or mass dysfunction), it is critical to acquire axial T1- and T2-weighted images all
(Fig. 9-27). Our experience and the cases reported in the literature the way from the conus medullaris to the bottom of the thecal sac to ensure
(196–202) indicate that these are incidental findings and are rarely, that a fibrolipoma is not missed. The conus medullaris is often, but not
if ever, associated with signs or symptoms. After confirmation of the always, low-lying, being tethered by the tight filum (71,82). It is impor-
purely cystic nature of the lesion by ultrasound (in infants) or MRI tant to remember that the conus may be at a normal level (L-2 inferior
(FLAIR and diffusion-weighted MRI are preferable), it should be con- endplate or above) in symptomatic patients (82). An area of prolonged
sidered as an anatomical variant. T1 and T2 relaxation, probably representing a dilated central canal, is
seen in the center of the spinal cord on T1-weighted MR images in
Anomalies of the Filum Terminale/Tight Filum approximately 20% to 25% of affected patients (Fig. 9-31). It is not
known whether this area represents a mild hydromyelia or myelomala-
Terminale (a simple spinal dysraphism) cia resulting from the metabolic abnormalities within the conus as a
The normal filum terminale is a long thin fibrous filament that is both result of the tethering (82,208). However, the fact that this finding may
intra- and extradural. It starts at the tip of the conus medullaris and disappear after release of the filum suggests that it represents hydromy-
extends caudally, through the bottom of the subarachnoid space and elia and is caused by the tethering (209).
the dura mater, to insert on the dorsal aspect of the first coccygeal ver- The dural sac is frequently widened and the dorsal dura may be
tebra (166). Fibrolipomas and cysts of the filum likely result from minor tense and tented posteriorly by the filum. The filum may be so closely
anomalies in the sequence of canalization and retrogressive differentia- applied to the dura posteriorly that it cannot be detected by myelogra-
tion; the pleuripotential caudal cell mass produces fat cells or does not phy (20). Therefore, MR with thin sagittal and, especially, axial sections
properly connect areas of cavitation to the central canal. The fibroli- is the study of choice. The thickness of the filum must be measured from
pomas may be limited to the intradural filum, may be limited to the axial images and should always be measured at the L-5 to S-1 level, as the
extradural filum, or may involve both portions. Cysts may be found by filum may be stretched, and therefore of normal thickness, at more rostral
ultrasound in up to 12% of young infants who are referred for spinal levels. In addition, the level of the conus medullaris is best determined
sonography (203); they are most commonly seen dorsally in the rostral from axial images, as the roots of the cauda equina may obscure the pre-
portion of the filum, just caudal to the tip of the conus medullaris. cise level of the conus when the sagittal images are viewed (82). In a series
They appear to be asymptomatic and are associated with normal devel- of 31 patients operated for the tethered cord syndrome, 55% had an
opment during the first postnatal year (203). obviously thick, fibrotic filum, 23% had a small fibrolipoma within the
It is likely that the tight filum terminale results from failure of invo- thickened filum, and 3% had a small filar cyst. In 13% of patients, no
lution (apoptosis and/or necrosis) of the terminal cord and/or failure distinct filum was seen; rather, the spinal cord was markedly elongated,
of lengthening of the nerve fibers that form the filum. When the filum extending downward to the lower end of the dural sac and ending in
terminale is too short, the tethering effect is transmitted to the spinal a small lipoma at the caudal end of the thecal sac (Fig. 9-31) (210).
cord. However, because the dentate ligaments secure the cord rostral to Increased awareness of the signs and symptoms of spinal cord tether-
T12, the effect of the tethering is limited to the conus medullaris and ing have prompted some neurosurgeons to section the filum terminale

Barkovich_Chap09.indd 884 5/6/2011 10:28:45 PM


Chapter 9 • Congenital Anomalies of the Spine 885

FIG. 9-28. Fibrolipoma of the filum


terminale in an adolescent. A. Sagittal
T1-weighted image shows a thickened
filum terminale (arrows) demonstrating a
short T1 relaxation time indicative of fat.
Note that the conus medullaris is at the
normal level. B. Axial T1-weighted image
through the filum shows a thickened, fatty
filum diagnostic of a fibrolipoma.

FIG. 9-29. Fibrolipoma of the filum terminale in an infant; importance of axial images through the L-5 to S-1 levels. A. Sagittal T1-weighted image shows
the conus terminating near the bottom of L-2. No fat is seen in the filum. B. Axial T1-weighted image at the L-3 to L-4 level shows normal cauda equina
without any abnormality of the filum terminale. C. Axial T1-weighted image at the L-5 level shows the thick, fatty filum (arrow) in the dorsal aspect of the
thecal sac. D. Different patient. Axial T2-weighted image at the L-5 level shows the enlargement and hypointensity (arrow) from fibrous thickening of the
filum terminale. If the filum terminale appears larger than the roots of the cauda equina on axial T2 images, it is probably too thick.

Barkovich_Chap09.indd 885 5/6/2011 10:28:45 PM


886 Pediatric Neuroimaging

FIG. 9-30. Fibrolipoma of the filum terminale; use of axial T2-weighted images. A. Sagittal T1-weighted image shows normal level of the conus medullaris.
B. Axial T1-weighted image shows a thick, fatty filum terminale (white arrow) at the sacral level. C. Axial T2-weighted image shows the filum (black arrow)
as abnormally thick.

FIG. 9-31. Tethered spinal cord (tight filum terminale syndrome). A and B. Sagittal T1- and T2-weighted images show a very thin, elongated spinal cord
extending all the way down into a terminal lipoma (white arrow in A). The thickness of the filum at the L-5 to S-1 level is always greater than 1 mm in these
patients. Note the dilated central canal (black arrows in B), present in 20% to 25% of patients with tethered spinal cord. C. Axial T2-weighted image at the
S-1 level shows nerve roots as linear hypointense structures exiting ventrally, indicating that this is spinal cord, not a thick filum. D–F. Tethered spinal cord
with a tail.

Barkovich_Chap09.indd 886 5/6/2011 10:28:47 PM


Chapter 9 • Congenital Anomalies of the Spine 887

FIG. 9-31. (Continued) D. Sagittal T2-weighted imaging shows the markedly elongated distal spinal cord, extending all the way to the abnormally low
termination of the thecal sac at the S-4 level (small white arrow). The normal thecal sac should terminate at the S-2 level. A portion of the 13 cm tail can be
seen behind the lumbar spine soft tissues (large white arrowhead). E and F. Axial T1-weighted images at the L-4 and S-3 levels show the low termination of
the spinal cord but no apparent fat deposition. The tail (arrows) can be seen posteriorly in both images.

more frequently, even in the setting of a normal MRI (55,211); the hypogenesis or anal atresia, are more common than the more severe
results of an ongoing prospective, randomized trial were not available forms. Approximately one sixth of infants with caudal regression syn-
at the time of this writing (212). drome have diabetic mothers (220). As would be expected from the
Phase-contrast MRI may be useful to detect diminished motion embryology, a high incidence of lumbosacral hypogenesis is found in
of the cervical spinal cord in patients with cord tethering (121,122). patients with anomalies of the lower gastrointestinal and genitouri-
The normal spinal cord moves caudally at a velocity of 1 cm/s during nary systems, particularly those with anal atresia (43,44). Moreover,
CSF diastole (at which time, CSF is moving rostrally at a velocity of up the higher the level of the anal atresia, the more severe is the lum-
to 2 cm/s) and moves rostrally during CSF systole (122). In patients bosacral hypogenesis (43,49,221). Therefore, the sacrum and lower spi-
with spinal cord tethering, this motion is damped and the cord veloc- nal cord should be carefully scrutinized in all patients with anomalies of
ity is reduced (121,122). After release of the tethering, normal motion the lower GI and GU systems. About 10% of patients with lumbosacral
returns in about one third of patients (122). hypogenesis have OEIS (concurrent omphalocele, cloacal exstrophy,
imperforate anus, and spinal deformities) and another 10% have the
Syndrome of Caudal Regression VACTERL syndrome (vertebral anomalies, anorectal malformations,
cardiac malformations, tracheoesophageal fistulae, renal anomalies,
(a complex spinal dysraphism)
and limb anomalies) (49).
The syndrome of caudal regression is composed of a spectrum of Clinical signs at presentation range from isolated deformities of the
anomalies, including sirenomelia (fusion of the lower extremities), feet or minor distal muscle weakness to complete sensorimotor paraly-
absence of the caudalmost vertebrae and spinal cord (lumbosacral sis of both lower extremities (49,218). Motor deficits, almost always
agenesis), anal atresia, malformed external genitalia, exstrophy of the more severe than the sensory loss, correspond to the level of vertebral
urinary bladder, renal aplasia or ectopia, and pulmonary hypopla- loss (within one level) unless an associated myelomeningocele is pres-
sia with Potter facies (213,214). The syndrome seems to result from ent. In most patients, sensation is intact caudally for several segments
disturbances of the caudal mesoderm, including the caudal cell mass below the vertebral loss; therefore, perineal sensation can be preserved
and cloaca, before the fourth week of gestation (213,214). Two cases despite severe leg weakness, sphincter disorder, and sacral agenesis.
have been described in association with deletion of the short arm of Almost all patients have a neurogenic bladder (49). The clinical sce-
chromosome 18 (215,216). The lack of formation of the caudal spi- nario of sphincter dysfunction and lower extremity motor deficits
nal cord and spinal column in patients with lumbosacral agenesis has results in a tentative clinical diagnosis of tethered spinal cord in many
been postulated to result from maldevelopment of the lower portions of the less severely affected patients. Indeed, as many as 60% of patients
of the spinal cord and notochord due to a toxic, infectious, or isch- with lumbosacral hypogenesis, particularly those with progressive neu-
emic insult. This insult presumably impairs the normal migrations of rologic symptoms, have cord tethering superimposed upon agenesis of
neurons, paraxial mesoderm cells (somites that form the vertebrae), the lower sacral nerve roots (49,221).
and the lateral mesoderm cells that form the lower digestive tract The degree of spinal agenesis is variable, ranging from a partial or
(39,217). Another possibility is that the notochord is injured, resulting total unilateral sacral agenesis (with an oblique lumbosacral joint) to
in abnormal neurulation and maldevelopment of the vertebral bodies. total agenesis of the lumbar and sacral spine (48). The caudalmost two
The abnormal neural tube subsequently causes abnormal development or three vertebral bodies are often fused and dysplastic (Figs. 9-32 and
of the vertebral arches (218). Other possibilities include abnormality 9-33). The bony canal immediately rostral to the last intact vertebra
of glucose transporter genes (219), mutation or lack of expression of may be severely narrowed as a result of bony excrescences originat-
the genes that code for the development of the lowest segments of the ing from the vertebral bodies, from fibrous bands connecting the bifid
spine, or induction of apoptosis in these segments for reasons that are spinous processes, or from stenosis of the dural tube. Surgical release
not currently understood. of such dural stenosis and duroplasty may achieve an improvement
The incidence of caudal regression syndrome is approximately of neurologic function (218). The major neural anomaly is hypopla-
1 in 7,500 births (214). The milder forms, including isolated sacral sia of the distal spinal cord, which is typically more severe ventrally

Barkovich_Chap09.indd 887 5/6/2011 10:28:48 PM


888 Pediatric Neuroimaging

FIG. 9-32. Severe caudal regression with VACTERL syndrome. A. Coronal T1-weighted image shows scoliosis, absence of the right kidney, and vertebral
anomalies at the apex of the spinal curvature (asterisk marks a hemivertebra). The sacrum is absent. White arrow points to the level of a neurenteric cyst.
B. Sagittal T1-weighted image of the cervical and upper thoracic spine shows a gap in the vertebral bodies (white arrows) at the level of the neurenteric cyst
and the cyst (white arrowheads). C. Sagittal T1-weighted image shows the blunted cord terminus (arrows) at the midthoracic level.

than dorsally, resulting in a characteristic blunted or “wedge” shape of show associated cord tethering or dural stenosis, if present. Sagittal and
the cord terminus (Figs. 9-32 and 9-34) (48). Such blunting is seen in coronal MR images are helpful in detecting hypogenesis or dysgenesis
essentially all children with partial or complete sacral agenesis and ter- of the caudal vertebral bodies (Figs. 9-32 and 9-33). Moreover, assess-
mination of the spinal cord above the level of the first lumbar vertebra ment of the level and shape of the conus medullaris will determine
(49). It is the result of diminished numbers of anterior horn cells in whether tethering is present. MRI reveals a characteristic blunted or,
the distal cord and abnormally small sacral nerve roots. Below the level often, “wedge-shaped” appearance of the terminus of the spinal cord
of the last intact vertebra, scattered nerve fibers pass through dense in those patients in whom the cord is not tethered; in these cases, the
fibrous tissue (151,222). cord terminus will generally be located above the L-1 level (Figs. 9-32
The spinal cord may be tethered in sacral agenesis; Pang et al. and 9-34) (44,48). When this appearance is seen on sagittal images,
(49) found tethering of the spinal cord in all patients with lumbosacral the sacrum should always be scrutinized to assess for the presence and
hypogenesis in whom the conus terminated below the L-1 level. They also degree of hypogenesis. As discussed above, the distal cord has a tapered
found a correlation with level of the conus and severity of the sacral appearance when tethered (Fig. 9-33); typically, the cord will terminate
hypogenesis. Termination of the conus above L-1 is highly correlated below the L-1 level in these patients. Axial T1- and T2-weighted images
with sacral malformations ending at S-1 or above, whereas conus ter- should be obtained through the levels immediately adjacent to the level
mination below L-1 is highly correlated with sacral malformations of regression to look for thickened or fatty filum terminale (Fig. 9-30),
ending at S-2 or below (49). Therefore, tethering is more common in a lipomyelocele, terminal hydromyelia (Fig. 9-34), lipoma, or the pres-
milder sacral agenesis. It is important to note the presence of spinal ence of bony spinal stenosis (44,48).
cord tethering in patients with lumbosacral hypogenesis, as release of
the tethered cord may result in improved urinary function, even if the Terminal Myelocystocele (a CSD with a
neurologic deficit remains static (223); at the very least, the release of
subcutaneous mass)
the tethered cord is likely to prevent neurological worsening (169).
Tethering may be associated with a thick filum terminale (65%), ter- The terminal myelocystocele (also known as syringocele) is the least
minal myelocystocele (15%), terminal hydromyelia (10%), or terminal common form of spinal dysraphism with posterior osseous defect
lipoma/lipomyelocele (10%) (49). When the spinal cord is tethered in (224). This anomaly is an occult spinal dysraphism in which the hydro-
lumbosacral hypogenesis, the cord terminus is not wedge-shaped, but myelic spinal cord and the arachnoid are herniated through a posterior
elongated, like a typical tethered spinal cord (Fig. 9-33). element defect, creating an unusual type of skin-covered myelomenin-
On imaging studies, the diagnosis of caudal regression is easily gocele (Fig. 9-35). Affected patients present with midline cystic masses
made. Plain films allow diagnosis of the bony hypogenesis but do not in the lumbosacral region. They have no bladder or bowel control, and

Barkovich_Chap09.indd 888 5/6/2011 10:28:49 PM


Chapter 9 • Congenital Anomalies of the Spine 889

FIG. 9-33. Caudal regression with a tethered cord.


A. Sagittal T1-weighted image shows a low-lying cord
terminus that is tapered, not blunted, as seen when
caudal regression is associated with tethering of the
spinal cord. Black arrows point to a terminal lipoma.
B. Coronal T1-weighted image shows a small, dysplas-
tic sacrum (black arrows), which is fused with the lower
lumbar vertebrae.

FIG. 9-34. Mild caudal regression. A. Sagittal T1-weighted


image shows that the cord terminus is blunted, ending at the
mid T11 level. The central canal appears dilated. The spinal col-
umn is normal except for the sacrum. S-2 is dysplastic and the
S-3, S-4, and S-5 segments and the coccyx are absent. B. Axial
T1-weighted image shows substantial dilatation of the central
canal of the spinal cord.

Barkovich_Chap09.indd 889 5/6/2011 10:28:50 PM


890 Pediatric Neuroimaging

lower extremity function is typically severely impaired (225). As with


other anomalies of the caudal cell mass, the terminal myelocystocele is
often associated with anomalies of the anorectal system, lower genito-
urinary system, and vertebrae, such as anal atresia, cloacal exstrophy,
lordosis, scoliosis, and partial sacral agenesis (45,225). Myelocystoceles
in other locations are more likely anomalies of disjunction and are dis-
cussed in that section, earlier in the chapter.
In the terminal myelocystocele, the spinal cord partially or com-
pletely traverses a meningocele and inserts within its lateral or poste-
rior wall (Fig. 9-35). At the junction line between the placode and the
meninges, the pia covering the spinal cord is reflected along the wall
of the meningocele, creating a closed cavity filled with cerebrospinal
fluid. In all forms of myelocystocele, a hydromyelic cavity lies within
the cord. At the level of the myelocystocele, this hydromyelic cavity
dilates into a large ependyma-lined cyst that protrudes beyond the
reflection of the pia in such a manner that it can be extra-arachnoidal
in location (Fig. 9-35). On occasion, the dorsal mesoderm is fatty in
these patients (169); in this situation, the malformation is termed a
lipomyelocystocele.
Imaging studies demonstrate direct continuity of the meningo-
cele with the subarachnoid space and the presence of a cyst in conti-
nuity with the central canal of the spinal cord (Fig. 9-36) (77,79,224).
Because the cyst does not communicate with the meningocele, myel-
ography will demonstrate only the meningocele (if present), which
is much smaller in volume and at a different level than the clinically
apparent mass. Imaging studies show that the clinically apparent mass
is a second cyst, which is thin-walled and has no internal structure
(Fig. 9-36) (77,79,80,224,226). The cyst may show delayed opacifi-
cation with water-soluble intrathecal contrast, similar to a hydromy-
FIG. 9-35. Schematic of terminal myelocystocele. Hydromyelia is always elia. The ependyma-lined (hydromyelia) cyst is frequently the larger
present in the spinal cord. At the cord terminus, the central canal opens of the two cysts; it is typically situated posteriorly and inferiorly to
into a large cyst (c). This cyst is below a region of bony spina bifida and the meningocele, but will occasionally extend rostrally outside the
expanded subarachnoid space (sas) that surrounds the distal spinal cord. meningocele.

FIG. 9-36. Terminal myelocystoceles; infants with sacral


masses. A. Sagittal T1-weighted image in a 2-day-old neonate
shows an expanded, syringohydromyelic spinal cord, which
appears to terminate at the midsacral level (white arrow). A
large cystic mass is seen caudal to the apparent cord termina-
tion (black arrows). At surgery, this terminal cyst was found to
communicate with the dilated central canal of the spinal cord,
resulting in the classification of this anomaly as a myelocysto-
cele. B. Sagittal T1-weighted image shows the myelocystocele
(c) in direct contact with the syringomyelic cavity (s) in the
distal spinal cord. The walls of the cavity (arrows) are seen to
separate as they lead into the cyst.

Barkovich_Chap09.indd 890 5/6/2011 10:28:51 PM


Chapter 9 • Congenital Anomalies of the Spine 891

Anterior Sacral Meningocele anus or anal stenosis, and a tethered spinal cord, has been described
(227). The sacral dural sac is usually widened; it communicates with
Anterior sacral meningoceles are anomalies characterized by focal the pelvic cyst through a (usually) narrow neck that is situated within
erosion or hypogenesis of sacral or coccygeal segments with herniation the sacral defect. The meningoceles may be large or small, simple or
of a cerebrospinal fluid-filled meningeal sac through the defect into multiloculated, devoid of neural tissue or traversed by nerve roots
the pelvis. They account for approximately 5% of retrorectal tumors within the wall or lumen of the sac (76,227–229). The identification
and are usually diagnosed in the second and third decades of life. In of traversing nerve roots is crucial, since the neck of the meningocele
children, males and females are equally affected (20,76). Symptoms cannot be simply ligated in their presence.
are produced as a result of pressure on the pelvic viscera, causing con- Bony anomalies consist of a widened sacral canal, scalloping of
stipation, urinary frequency and incontinence, dysmenorrhea, dys- the anterior wall of the sacrum, and sacral scoliosis (229). The curved
pareunia, or pain in the lower back or pelvis. Furthermore, pressure appearance of the residual sacrum, which is scalloped beneath the
may be exerted on nerve roots, resulting in sciatica, diminished rectal defect, gives the appearance of scimitar sacrum or a sickle-shaped
and detrusor tone, or numbness and paresthesia in the lower sacral sacrum on sagittal images; this appearance is highly suggestive of an
dermatomes. Finally, fluid shifts between the sac and the spinal suba- anterior sacral meningocele (228).
rachnoid space can cause intermittent high-pressure headache (when Imaging studies show a deficient sacrum and a variably sized cyst
supine), nausea, vomiting (especially during defecation), or low- extending into the pelvis through an enlarged sacral foramen. Conti-
pressure headache (while in the erect position). nuity of the cyst with the thecal sac must be demonstrated in order to
The embryogenesis of anterior sacral meningoceles is not deter- make the diagnosis. On MR studies, the continuity is best demonstrated
mined. A relatively simple form arises in patients with neurofibromatosis in the sagittal plane (Fig. 9-37) (76). If CT is used, contrast should be
Type 1 and in patients with Marfan syndrome. A more complex famil- injected intrathecally to optimally show the continuity of the meningo-
ial form, which is associated with a partial sacral agenesis, imperforate cele with the subarachnoid space. Whatever imaging technique is used,

FIG. 9-37. Anterior sacral meningocele. A and B. Lateral view and sagittal CT
reformation from a lumbosacral myelogram show the extension of the intrath-
ecal contrast through an anterior sacral osseous defect and into the presacral
space (arrows). C. Axial bone window from a CT myelogram shows the dysplas-
tic appearance of the left hemisacrum (h) and absence of the right hemisacrum
anteriorly. Note the contrast extension into the presacral space (white arrow). D
and E. Sagittal T1- and T2-weighted images show the anterior sacral meningocele
(arrows) coursing under the hypoplastic sacrum. It is important in these anoma-
lies to determine whether nerve roots of the cauda equina are floating within the
meningocele before the meningocele is ligated.

Barkovich_Chap09.indd 891 5/6/2011 10:28:52 PM


892 Pediatric Neuroimaging

the most important features to demonstrate are (a) tethering of the components, and calcification, the tumors are variably heterogeneous.
cord and the associated tethering lipoma or dermoid, if present, and Adequate CT evaluation necessitates opacification of the bowel, the ure-
(b) whether nerve roots traverse the sacral defect. ters, and bladder by the use of contrast agents to evaluate the relation-
ship of the tumor to the pelvic viscera. Contrast agents are not necessary
if MR is used. The MR appearance of the tumor is variable, depending
Sacrococcygeal Teratoma
upon its composition. Most often, the solid portions of these lesions are
Sacrococcygeal teratomas are rare congenital tumors that develop in extremely heterogeneous as a result of high signal from fat, intermedi-
the lower sacrum. They are thought to arise from rests of totipotential ate signal from soft tissue, and low signal from calcium on T1-weighted
cells in the caudal cell mass. Females are affected more commonly than images (Figs. 9-38 and 9-39). The tumors are usually lobulated and
males by a four to one preponderance. Sacrococcygeal teratomas may sharply demarcated. When they are mostly cystic (Fig. 9-39), intrave-
present as external masses protruding from the gluteal cleft or from nous contrast administration may be necessary to identify a solid com-
the perineum, or may intrude into the pelvis, causing radicular pain, ponent in the wall of the cyst.
constipation, or urinary frequency and incontinence. Histologically,
approximately two thirds are mature teratomas; the other one third ANOMALIES OF DEVELOPMENT
is split relatively evenly between immature teratomas and anaplastic OF THE NOTOCHORD
carcinomas (20). The vast majority of these teratomas are large, well-
encapsulated, lobulated masses, having both solid and cystic portions. The Split Notochord Syndrome Spectrum
Approximately 5% are predominantly cystic. (complex spinal dysraphisms)
Sacrococcygeal teratomas have been classified by their location
(230–232). Type I tumors (47%) are situated almost entirely exteriorly Embryology
and have only a minimal presacral portion. Type II tumors (35%) are A review of some embryology aids in the understanding of split
also largely exterior in location, but have a significant pelvic extension. notochord malformations. During the second week of gestation, the
Type III tumors (8%) are visible from the exterior as a mass but are embryo consists of a flat, two-layered disc. The developing ectoderm
mainly in the pelvis and abdomen. Type IV teratomas (10%) are entirely lies dorsally, in contact with the amniotic cavity. The primitive endo-
presacral without any external mass. The sacral canal is involved in 2% derm lies ventrally, in contact with the yolk sac. During the third
of affected patients. week of life, a thickening occurs in the dorsal midline as a result of a
If a sacral malformation is associated with a sacrococcygeal tera- focal rapid proliferation of cells; this area has been called the Hensen
toma, it suggests the rare familial form of tumor, which has an auto- node. Cells proliferate and migrate from Hensen node, separating the
somal dominant inheritance, a slight female preponderance, and ectoderm from the endoderm; these cells course in predefined routes
prominent sacrococcygeal defects. Anorectal stenosis, vesicoureteral lateral to the midline. Subsequently they will join in the midline to
reflux, and cutaneous stigmata are frequently present in children with form the notochordal process, which will eventually roll into a tube
the familial form. and separate from the endoderm to form the definitive notochord. If
The sporadic and familial forms are indistinguishable on imag- the ectoderm fails to completely separate from the endoderm (leav-
ing studies. Bony erosions in the sacrum, when present, outline ing a strand or adhesion), the notochord must either remain split
the entire soft tissue component, whether dorsal to the sacrum or around the adhesion or deviate to the left or right of it (31,233,234).
intrapelvic. Depending upon the percentage of solid components, cystic Moreover, the primitive neurenteric canal, formed by the process of

FIG. 9-38. Sacrococcygeal teratoma in a 27-week fetus. A. Sagittal imaging shows the heterogeneous mass (Type 3 teratoma) with the large intra-abdominal
and pelvic component and the smaller external component. This particular tumor was of an aggressive, immature histology and invaded the spinal canal
(arrows). B. Coronal image shows the widening of the pedicles by the invasive spinal component (arrows). C. A more anterior coronal image shows the
obstructive hydronephrosis and displacement of kidneys due to the large pelvic mass.

Barkovich_Chap09.indd 892 5/6/2011 10:28:53 PM


Chapter 9 • Congenital Anomalies of the Spine 893

FIG. 9-39. Cystic sacrococcygeal teratoma. A. Sagittal T1-weighted image shows a cystic mass (Type 3 teratoma, arrows) centered at the coccyx. B. Post-
contrast T1-weighted image with fat suppression shows a small solid portion of tumor (arrows). The high signal intensity focus in the tumor seen in C
disappears, proving that it represents fat. No enhancement is present. C. Precontrast T1-weighted image shows the presence of high signal intensity fat
within the solid portion of the tumor (arrows).

intercalation, may not regress, leaving a residual connection between or spherical diverticula that arise from the dorsal mesenteric border
endoderm and ectoderm. The mesoderm, which normally surrounds of the bowel. These diverticula represent a persistence of a portion of
the notochord to form the vertebral bodies, is split or deviated by the the tract between the gut and the vertebral column. Because the bowel
anomaly, resulting in a persistent connection between the dorsal sur- segments migrate inferiorly and rotate, it is not uncommon to trace
face of the gut and the dorsal, midline surface of the body. As the dorsal enteric diverticula far upward from the small bowel through
embryo grows and the gut migrates, the adhesion can become quite the mesentery and diaphragm into the mediastinum. Occasionally, the
long and the structures connected by it may become quite topo- diverticula will extend to the anterior surface of the vertebra or into
graphically distant. Because the notochord may remain split or devi- the spinal canal (235). If there is involution of the portion of the diver-
ate around the adhesion, the connection of the cyst to the vertebrae ticulum near the gut, a prevertebral dorsal enteric cyst is formed (236).
may be in the midline or paramedian. Moreover, because all or part Finally, in some patients, the dorsal enteric fistula may close at mul-
of the connection may involute, only portions of the connection may tiple sites. In these patients, one may find combined, noncommunicat-
be present (233,234). ing elements of the split notochord syndrome; for example intestinal
diverticula, mediastinal enteric cysts, and intraspinal enterogenous
Classification cysts may be present.
The term “split notochord syndrome” refers to a spectrum of anoma-
lies that are believed to result from splitting or deviation of the noto- Clinical and Imaging Characteristics
chord with a persistent connection between the ventrally located Patients with dorsal enteric fistula usually present as newborns with a
endoderm and the dorsally located ectoderm (235). The most severe bowel ostium or an exposed pad of mucous membrane in the dorsal
form of this syndrome is a fistula (the dorsal enteric fistula) through midline. These openings pass meconium and then feces. Associated con-
which the intestinal cavity communicates with the dorsal skin surface genital tumors have been reported (238). Patients with dorsal bowel her-
in the midline, traversing the prevertebral soft tissues, the vertebral niations present with large, midline, dorsal sacs that are partly covered
bodies, the spinal canal and its contents, and the posterior elements by skin and partly covered by mucous membrane; the vertebral column
of the spine (235–237). The most extreme form of the dorsal enteric around the sac is duplicated (Fig. 9-40) (237). Patients with extraspinal
fistula is a dorsal bowel hernia in which a portion of bowel is herniated enteric cysts present with masses either in the mediastinum or in the
into a skin- or membrane-covered dorsal sac after passing through a abdomen. Children with the above-mentioned lesions do not present
combined anterior and posterior spina bifida. Often, the bowel lumen with neurological problems as their chief problem and, therefore, will
is open to the skin surface. Because any portion of the dorsal enteric not be further discussed in this book. The reader is referred to standard
fistula may become obliterated or persist, apparently isolated diver- pediatrics and pediatric radiology texts for further information.
ticula, duplications, cysts, fibrous cords, and/or sinuses may be present Patients with dorsal enteric diverticula typically come to medical
at any point along the tract. The dorsal enteric sinus is the remnant of attention because of gastrointestinal complaints, but can present with
the posterior part of the tract and forms a blind tract with a midline neurologic problems if the diverticulum extends into the spinal canal.
opening to the dorsal external skin surface (235,236). Dorsal enteric The imaging clue in this diagnosis is the abnormalities of the vertebral
(enterogenous) cysts (which are derived from the intermediate part of bodies on plain films and the extension of intra-abdominal structures
the tract) are enteric-lined cysts which may be prevertebral, retrover- through the vertebral body defects on cross-sectional imaging studies
tebral, or intraspinal (235,236). Dorsal enteric diverticula are tubular (Fig. 9-41).

Barkovich_Chap09.indd 893 5/6/2011 10:28:54 PM


894 Pediatric Neuroimaging

FIG. 9-40. Dorsal enteric fistula. A. Coronal T1-weighted image shows a split spinal column and spinal cord (arrows) framing the bowel. B. Axial GE image
shows two spinal columns (open curved arrows), two spinal canals (open straight arrows), and the high intensity bowel extending dorsally between them to
open (solid arrows) at the skin surface. (This case courtesy Rosalind Dietrich, M.D. San Diego, CA).

FIG. 9-41. Dorsal enteric diverticulum. A. Coronal T1-weighted image shows


abnormal vertebrae (arrows) and abnormal narrowing of the spinal cord in the
lower thoracic region. B. Sagittal T1-weighted image shows a deficiency (arrows)
in the vertebral bodies in the lower thoracic region. C. Sagittal T2-weighted
image shows the dorsal enteric diverticulum (black arrows) coursing toward the
vertebral body deficiency. D. Axial T2-weighted image shows a cleft separating
the two halves of a vertebral body (v). The diverticulum (white arrows) courses
between the vertebral bodies. A small cyst (c) is present within the spinal canal,
dorsal to the diverticulum.

Barkovich_Chap09.indd 894 5/6/2011 10:28:55 PM


Chapter 9 • Congenital Anomalies of the Spine 895

Intraspinal Enteric Cysts (Neurenteric Cysts, cyst may lie dorsal to the cord (Fig. 9-44) or partially or completely
Endodermal Cysts) within the cord (Figs. 9-42 and 9-45); it is most important to define the
Intraspinal enteric cysts (also called neurenteric cysts and endodermal cyst and its relationship to the spinal cord. The cyst contents may be
cysts) infrequently cause signs or symptoms in newborn infants; when similar to CSF (Figs. 9-42 and 9-45) or may be proteinaceous, short-
neonates are symptomatic, the cyst is typically quite large. More com- ening the T1 relaxation time and resulting in relatively (compared
monly, affected patients present as adolescents or young adults with to CSF) high intensity on T1-weighted images (Figs. 9-43 and 9-44)
intermittent or progressive local and radicular pain that is worsened (246). If the cyst is creamy or has xanthogranulomatous changes, it
by elevating intraspinal pressure. Eventually, myelopathy ensues as spi- may have a heterogeneous appearance on both T1- and T2-weighted
nal cord compression causes both local and distal long tract symptoms images (239,247). Occasionally, this heterogeneity may mimic a nodule
(239). By the time these patients are imaged, marked compression of and suggest the presence of a neoplasm (248); in such cases, admin-
the spinal cord is usually evident (65,240). Acute presentation is rare, istering contrast will help in differentiating cyst (no enhancement of
and usually follows infection or illness (241). nodule) from tumor (nodule usually enhances). The identification of
Enteric cysts are usually single, smooth, and unilocular, occurring anomalies of the adjacent vertebral bodies aids in identification of the
primarily in the lower cervical and thoracic regions. Rarely, they occur abnormal level. The abnormal vertebrae and the cysts can be identified
in the lumbar spine or intracranially in the pons, prepontine cistern, by myelography. Similar information can be obtained from MRI. Cor-
or cerebellopontine angle (see Chapter 5) (242–244). The fluid within onal images through the vertebral bodies (Figs. 9-44 and 9-45) identify
them may be nearly identical to cerebrospinal fluid or may be milky, abnormal vertebrae. The cyst may be difficult to detect if it is small and
cream-colored, yellowish, or xanthochromic. Enteric cysts are usually its contents are very similar to cerebrospinal fluid; FLAIR imaging may
intradural, extramedullary, and situated ventral or ventrolateral to the be helpful. However, the displacement and distortion of the spinal cord
cord; less frequently, the cyst may be dorsal or dorsolateral to the cord by the cyst facilitates detection (Fig. 9-42). In addition to evaluating the
or within the cleft of a diastematomyelia (240,244). An intramedullary spine, it is important to look for associated fistulae and mediastinal or
component is present in 10% to 15% (245). The cord is usually nar- abdominal cysts.
rowed and displaced by the mass when symptoms develop.
Spine radiographs may show an enlarged spinal canal at the site Split Cord Malformation (Diastematomyelia and
of the enteric cyst. Newborns and infants who are symptomatic from Diplomyelia—complex spinal dysraphism)
intraspinal enteric cysts sometimes exhibit vertebral anomalies includ-
ing hemivertebrae, lack of segmentation, partial fusions, and sco- Definition
liosis in the region of the cyst (such changes are more common with The term diastematomyelia, a form of SCM, refers to a sagittal division
mediastinal or abdominal dorsal enteric cysts). Older patients usually of the spinal cord into two symmetric or asymmetric hemicords, each
exhibit no vertebral changes other than focal enlargement of the canal containing a central canal, one dorsal horn (giving rise to a dorsal nerve
resulting from local pressure effects (240). root), and one ventral horn (giving rise to a ventral nerve root). Each
Some neurosurgeons believe that both CT and MRI are required hemicord is invested by its own layer of pia (20,63,151,249–252). The
for proper preoperative imaging analysis of a neurenteric cyst (246). division may involve the entire thickness of the cord or may only affect
CT shows vertebral anomalies in about 50% of affected patients; verte- the anterior or posterior half of the cord (partial diastematomyelia or
bral body clefts, butterfly vertebrae, and anomalies of segmentation are “horseshoe cord”) (47,234,253). Partial division is frequently observed
most common (245). The spinal canal may be focally expanded. MRI, in the transitional zones superior and inferior to an area of complete
however, shows most of the vertebral anomalies and, more importantly, diastematomyelia (20,74,234,252). Although one can find a number of
shows the cysts and their relationship to the spinal cord (Figs. 9-42 to definitions of the rare diplomyelia in the medical literature, it is most
9-45). MRI typically shows a cyst lying ventral to and compressing the commonly used to refer to a duplication of the spinal cord, with each
adjacent spinal cord (Figs. 9-43 and 9-44) (65,240,244). However, the cord containing a central canal, two dorsal horns (giving rise to two

FIG. 9-42. Neurenteric cyst. A. Sagittal T1-weighted


image shows the characteristic anterior spinal defect
associated with the neurenteric cyst, as well as a T1-
hyperintense cyst (white arrows) in the dilated spinal
canal. Segmentation anomalies of the vertebral bodies
and posterior elements are also present at this level. B and
C. Coronal T2-weighted imaging demonstrates the ante-
rior osseous defect (d) and the hyperintense cyst (arrow
in C) resulting in lateral deviation of the spinal cord.

Barkovich_Chap09.indd 895 5/6/2011 10:28:56 PM


896 Pediatric Neuroimaging

FIG. 9-43. Neurenteric cyst associated with SCM. A and B. Sagittal T1-weighted images show a hyperintense neurenteric cyst (small solid white arrows)
ventral to a dysplastic upper cervical spinal cord. The cyst points toward a space (small open white arrows) between the dysplastic upper cervical vertebrae.
C. Axial T1-weighted image shows a dysplastic vertebral body and a nearly complete sagittal cleft (solid white arrow) of the spinal cord. The cyst (open white
arrows) presumably contains proteinaceous, possibly inspissated, fluid. (This case courtesy of Dr. Michael Brant-Zawadzki).

dorsal nerve roots), and two ventral horns (giving rise to two ventral and is a common reason for clinical presentation in these age groups,
nerve roots). As diastematomyelia and diplomyelia cannot always be along with low back pain, sciatica, and perianal dysesthesias (255,258).
distinguished from one another and may be incorrectly used as syn- SCM is responsible for approximately 5% of congenital scolioses (254).
onyms, the term SCM is preferred (234). Neurologic symptoms are nonspecific and may be indistinguishable
from other causes of cord tethering (20,74,117,234,254,258–260).
Clinical Presentation
The signs and symptoms of SCM may appear at any time of life. Females Embryology
are affected much more commonly than males (74,117,254). Cutane- SCM most likely develops as a result of splitting of the notochord
ous stigmata such as hairy patches (hypertrichosis, the most common (234,253). If the cells forming the notochord, in the course of their
stigma), nevi, lipomas, dimples, and hemangiomas overlie the spine in migration from Hensen node, encounter an obstacle such as an adhe-
more than half of cases; these signs often lead to diagnosis in infancy sion between ectoderm and endoderm, the notochordal cells must
(255). Half of affected patients manifest orthopedic problems of the course laterally around the obstacle on one side or split and go around
feet, particularly clubfoot. A specific neuro-orthopedic syndrome (251), on both sides at the same time. As a consequence of this detour, the
consisting of weakness and muscle wasting in one leg, often associated notochord develops either a lateral notch or a central cleft. Because
with an ipsilateral clubfoot, is seen in about half of patients with lum- the notochord influences the development of the vertebral bodies,
bar SCM (256). Scoliosis is common in older children and adults (257) any alteration of the notochord could result in an abnormality of the

FIG. 9-44. Dorsal intraspinal enteric cyst. A. Sagittal


T1-weighted image shows a mass (arrows) dorsally in the
cervical subarachnoid space compressing the spinal cord.
B. Coronal T1-weighted image shows several hemiverte-
brae (arrows) at the level of the cyst.

Barkovich_Chap09.indd 896 5/6/2011 10:28:57 PM


Chapter 9 • Congenital Anomalies of the Spine 897

FIG. 9-45. Intramedullary neurenteric cyst. A. Sagittal T2-weighted image shows an apparent cyst (solid black arrow) within the dorsal aspect of the spinal
cord. The upper cervical vertebral bodies (open white arrows) appear small and possibly dysplastic. B. Axial T1-weighted image shows the cyst (solid white
arrows) within the dorsal spinal cord. C. Coronal T1-weighted image shows the multiple malsegmented vertebrae (white arrows) of the cervical and upper
thoracic spine.

vertebral bodies such as a hemivertebra (if the notochord were notched) usually reunite caudal to the cleft (74). Occasionally, however, the cleft
or a butterfly vertebra (if the notochord had a cleft). Similarly, a split will extend unusually low and the hemicords remain distinct with two
notochord could induce the formation of two neural plates, which may separate coni medullaris and two fila terminale.
go on to form two hemicords. Surrounding mesenchyme can migrate In 40% to 70% of patients, the arachnoid and the dura split into
into the space between the hemicords, forming a fibrous, cartilaginous, two separate arachnoid and dural tubes, each surrounding the corres-
or bony spur. Moreover, one can imagine in this setting that the distal ponding hemicord (258). Thus, each hemicord has its own pial, arach-
notochord could be split, resulting in two separate foci of canalization noid, and dural sheath for several spinal sections; this is referred to as
and retrogressive differentiation; this sequence of events could explain Type I SCM (234,251,263). Patients with two separate dural sheaths
the rare cases of SCM affecting only the conus medullaris and filum nearly always have a bony or cartilaginous spur at the most inferior
terminale. portion of the cleft; this is often better seen on axial and coronal images
than on sagittal images (Fig. 9-47). The spur may originate from a lam-
Pathology and Imaging ina or from the vertebral body (Figs. 9-46 and 9-47). If the myelogram
Imaging of SCM can be difficult, because patients commonly have or imaging study appears to show the spur at a more rostral level, a sec-
severe scoliosis, often with a rotatory component. MR is the best tech- ond spur, present in approximately 5% of patients with SCM, is likely
nique if 3DFT volumetric techniques are used or if oblique images to be present (Fig. 9-46) (84,234,251,263). The bony spur forms from
are acquired in planes parallel and perpendicular to each relatively cartilage and has multiple ossification centers. Therefore, depending
straight segment of spine. These techniques are described in Chapter 1. upon the age of the patient and the number of ossification centers,
T1-weighted images are optimal to visualize the spinal cord and to the spur can be nonossified cartilage, cartilaginous with multiple small
look for fibrolipomas of the filum terminale. Exiting nerve roots and ossification centers arranged linearly between the two hemicords, a
thickening of the filum terminale are best imaged by axial T2-weighted bony strut attached to the wall of the spinal canal by a synchondrosis,
RARE images. Bony and cartilaginous spurs are most easily identified or a complete osseous bridge joining the vertebral body with the pos-
on axial T2- or T2*-weighted images or on CT. However, if the patient terior elements (252). The spur may be isointense or slightly hyperin-
has difficulty in holding still during the MR exam or if the scoliosis tense compared to CSF on T1-weighted images if nonossified; it will
is particularly severe, CT myelography with reformations in multiple be hyperintense on T1-weighted images if ossified (Fig. 9-46) because
planes, parallel and perpendicular to the course of the spinal cord, may of the high signal from the marrow. Bony, cartilaginous, and fibrous
be extremely useful to assess the course of the spinal cord and the pres- spurs all appear hypointense on T2-weighted spin-echo and gradient-
ence or absence of a fibrolipoma of the filum. echo (Fig. 9-46) images. The CT attenuation will vary, depending upon
The level of the cleft is variable but it is most commonly (more than how much of the spur is bone, how much is cartilage, and how much
80%) in the lumbar region (258). Upper thoracic clefts are unusual and is fibrous tissue; the appearance of the bony/cartilaginous section will
cervical clefts are rare (117,258,261,262). It is possible that cervical and vary with the stage of ossification.
upper thoracic SCM are more common than is generally accepted but Usually, the bony spur is in the midline sagittal plane and divides
remain asymptomatic because they are less likely to cause symptom- the canal into two fairly equal halves. However, scoliosis may rotate
atic tethering of the cord than are clefts that are located more caudally. the spur such that the two hemicanals are oriented more anteriorly
(Tethering is less likely because the segmental level of the spinal cord and posteriorly (Fig. 9-48) (251). Occasionally, the spur may cross the
much more closely coincides with the segmental level of the adjacent spinal canal obliquely and insert into an externally rotated lamina or
vertebrae in the cervical and upper thoracic levels.) The two hemicords a pedicle creating a significant asymmetry in the two hemicanals; in

Barkovich_Chap09.indd 897 5/6/2011 10:28:58 PM


898 Pediatric Neuroimaging

FIG. 9-46. SCM with spurs at two levels. A. Sagittal T1-weighted image shows a spur (curved arrow) at the midthoracic level. This spur is several levels
rostral to the lowest level of the split spinal cord, suggesting that a second spur is present. B. Adjacent image shows a possible spur (curved arrow) at the
lower thoracic level. C. Axial T1-weighted image at the level of the upper spur shows the two hemicords (arrows) separated by a poorly defined spur. D. The
spur (arrows) is better demonstrated by this GE image. E. Axial T1-weighted image shows the lower spur (arrows). Note that the posterior part of the spur
is fibrous and, therefore, poorly seen by MR.

these patients, the larger canal is usually posterior. MRI is especially facilitate identification of bone) in the axial plane or with CT. Resec-
helpful when patients have rotoscoliosis (Fig. 9-48). An initial coronal tion of the spur is critical for proper untethering of the cord (117).
image is obtained. Oblique sagittal and oblique axial planes of imaging In 30% to 60% of patients with diastematomyelia, the two hemi-
are then defined parallel and perpendicular, respectively, to the straight cords, each invested by an intact layer of pia, travel through a single
segments of spine, as described in Chapter 1. Alternatively, a volume subarachnoid space surrounded by a single dural sac; this malforma-
image may be obtained and reformatted in multiple planes. In our tion is referred to as Type II SCM (117,257,258,263). Each hemicord
experience, more artifact is present on volume images because of more has its own anterior spinal artery. This form of SCM is not accompa-
motion (due to greater time for the sequence) and the presence of two nied by a bony spur but always has a fibrous band coursing through the
phase encoding gradients. most inferior portion of the cleft, inserting in the dura (234). Patients
It must be emphasized that even osseous spurs can be missed with this type of SCM are rarely symptomatic unless hydromyelia or
on T1-weighted spin-echo MR images. Therefore, patients with two tethering (Figs. 9-49 and 9-50) is present (117,194,234). Imaging stud-
hemicords should be imaged using T2- or T2*-weighted images (which ies in these patients show the split cord and the vertebral anomalies, if

Barkovich_Chap09.indd 898 5/6/2011 10:28:59 PM


Chapter 9 • Congenital Anomalies of the Spine 899

FIG. 9-47. SCM with bony spur, scoliosis, and vertebral anomalies.
A. Coronal T1-weighted image shows thoracic dextroscoliosis with multiple
anomalies of vertebral segmentation (asterisks) including hemivertebra and
lack of segmentation. B. Coronal T1-weighted image adjacent to (A) shows
the hyperintense spur (white arrows) between the two hemicords. C. Sagit-
tal T1-weighted image shows the segmentation anomalies (white arrowheads)
and two areas of hydromyelia (arrows) but does not show the spur. D. Axial
T1-weighted image shows the hyperintense spur (white arrowheads) coursing
between the two hemicords.

Barkovich_Chap09.indd 899 5/6/2011 10:29:00 PM


900 Pediatric Neuroimaging

FIG. 9-48. SCM with rotoscoliosis. A. Coronal image showing the oblique plane prescribed for axial images. B and C. Coronal T1-weighted images show
vertebral anomalies (arrows) at the level of the most severe spinal curve. D and E. Images posterior to those in (B) and (C) show the spinal cord splitting
into two hemicords (arrows). F and G. Oblique axial T1-weighted images show the hemicords (curved arrows) aligned anterior and posterior to each other,
split around a spur (small white arrows) that is nearly horizontal. Note the markedly thick laminae (straight black arrows).

present, but rarely show the fibrous band (117). In the authors’ experi- is that of a small band of tissue extending from the hemicord to the
ence, the fibrous band is most often seen on sagittal T2-weighted RARE dorsal dura and mimicking a nerve root; only a portion of the band
images (Fig. 9-49). may be visible on the imaging study (117,265). This band of tissue can
Associated spinal anomalies are very common, being present in cause persistent tethering of the cord after resection of the spur and
up to 85% of affected patients (257). The conus medullaris is situ- transection of the filum, so its identification is important. Hydromyelia
ated below the L-2 level and the filum thickened in more than 75% is found in about half of patients with diastematomyelia. When pres-
of patients with SCM (Figs. 9-49 and 9-50) (117,264). Myeloceles/ ent, the hydromyelic cavity may extend from the spinal cord above the
myelomeningoceles (of the undivided spinal cord) are present in 15% cleft into one or both hemicords (264). The importance of the high
to 25%, whereas hemimyeloceles (myeloceles/myelomeningoceles of incidence of associated anomalies is that the radiologist cannot be sat-
one of the hemicords) are present in 15% to 20% of affected patients isfied with identification of the diastematomyelia, but must scrutinize
(117). Lipomas, dermal sinuses, (epi)dermoid tumors, and tethering the imaging examination in an attempt to identify other lesions that
adhesions (meningocele manqué) are reported commonly, but in less may lead to neurologic deterioration in the patient.
than 20% of patients (117,255). The meningocele manqué (Fig. 9-50) The spinal column is nearly always abnormal in patients with
is a very subtle finding on imaging studies and will only be seen if the SCM (Figs. 9-47 and 9-49) (74,194). The laminae are often thick and
radiologist is actively looking for it (117,260,265,266). The appearance may be fused with the ipsilateral or contralateral laminae of adjacent

Barkovich_Chap09.indd 900 5/6/2011 10:29:01 PM


Chapter 9 • Congenital Anomalies of the Spine 901

FIG. 9-49. SCM with fibrous spur. Value of sagittal T2 image and CT. A. Coronal T1-weighted image shows dextroscoliosis and shows the cleft (white
arrows) between the two hemicords. B. Sagittal T2-weighted image shows the fibrous spur (white arrowhead) coursing obliquely upward from the dorsal to
the ventral aspect of the spinal canal, through the tethered spinal cord. C. Axial T2-weighted image shows the bony prominence (white arrow) from which
the fibrous spur arises. The cord is not split at this level. D. The fibrous band is not seen on this axial image at the proper level because of the oblique course
of the band. E. Axial T1-weighted image at the sacral level shows a fibrolipoma of the filum terminale (arrowhead).

Barkovich_Chap09.indd 901 5/6/2011 10:29:03 PM


902 Pediatric Neuroimaging

FIG. 9-49. (Continued) F. Coro-


nal reformation of a noncontrast
CT scan shows interlaminar fusion
(white arrows), a common find-
ing in SCM. G. Axial image at the
same level as (C) shows the bony
prominence (black arrow) from
which the fibrous band arises. The
band was not seen on CT.

FIG. 9-50. SCM with single dural tube and meningocele manqué. A. Sagittal T1-weighted image shows an abrupt kyphosis at the (T-8) level of the SCM
(large white arrowhead). Focal hydromyelia (small white arrows) is seen two levels higher. B. Axial T1-weighted image shows the hydromyelia above the level
of the split cord.

Barkovich_Chap09.indd 902 5/6/2011 10:29:04 PM


Chapter 9 • Congenital Anomalies of the Spine 903

FIG. 9-50. (Continued) C. Axial GE image shows the two hemicords with small fibrous bands (meningocele manqué, black arrows) coursing posteromedi-
ally from each hemicord. These bands can cause local tethering of the spinal cord(s). D. Axial T1-weighted image shows a fibrolipoma (white arrow) of the
filum terminale at the sacral level.

vertebrae (Fig. 9-49); fusion with contralateral laminae of the adjacent


level is known as intersegmental laminar fusion. Spina bifida is almost
MALFORMATIONS OF UNKNOWN ORIGIN
always present. The association of an intersegmental laminar fusion All of the anomalies described in this chapter are actually of unknown
with spina bifida is seen in approximately 60% of patients with SCM origin. Those anomalies discussed up to this point can be explained
and is essentially pathognomonic for this anomaly. The intersegmen- by reasonable theories, permitting a classification based upon their
tal laminar fusion is almost always at the level of the SCM (74,250). embryogenesis. At this time, however, no reasonable theory has been
Anomalies of the vertebral bodies (Fig. 9-47) including hemiverte- proposed that explains segmental spinal dysgeneses, simple meningo-
brae, butterfly vertebrae, block vertebrae, and decreased disc height celes, and lateral meningoceles, which can develop anywhere along the
are observed in a high proportion of cases. Kyphoscoliosis (Figs. 9-48 spinal canal.
and 9-50) resulting from the osseous anomalies is seen in more than
half of patients. Segmental Spinal Dysgenesis
Another anomaly of the spinal column that is associated with par-
(a complex spinal dysraphism)
tial or complete SCM is the Klippel-Feil anomaly, a condition defined
as two or more consecutive unsegmented vertebrae at the cervical Segmental spinal dysgenesis is a malformation that is probably the
level (267,268). The Klippel-Feil anomaly has been divided into three result of embryonic segmental malformation (272,273) or focal injury
categories: Group 1 in which patients have a short, webbed neck with to the developing spine in utero (274). In utero detection is possible;
low hairline and complete lack of cervical segmentation; Group 2 in however, affected children are typically identified at the time of birth
which the lack of segmentation (most commonly called “fusion”) is by a kyphotic deformity (often sharply angled), anomalies of the lower
isolated and which most commonly involves C-2 to C-3 or C-5 to C-6; extremities (usually equinovarus deformities of the feet and flexion
and Group 3, in which patients have separate thoracic or lumbar levels contractures of the hips and knees), hyperreflexia of the lower extremi-
of nonsegmentation in addition to cervical involvement (47,267,269). ties, and bladder dysfunction. The skin overlying the deformity may
Neurologic impairment can result from several aspects of the Klippel- be bluish (273–275). One reported patient had dextrocardia and situs
Feil malformation. Probably the most common cause of neurologic inversus (274).
dysfunction or neck pain is accelerated disc degeneration resulting Imaging reveals a marked focal hypoplasia of the vertebral column,
from the restricted motion at the level of the unsegmented vertebrae thecal sac, and spinal cord, typically at the thoracolumbar junction.
(267). A second cause is malformation of the spinal cord; up to 50% Plain films show a kyphotic deformity with hypoplasia or absence of
of patients with the Klippel-Feil anomaly have at least partial dorsal one or more vertebral bodies. Myelography shows smooth tapering of
splitting of the cord (47,267,269). In such patients, anomalies have the thecal sac and spinal cord with a complete or nearly complete block
been identified in the decussation of the corticospinal tracts (270), of contrast at the most severely affected level(s). Caudal to the narrow-
and mirror movements may be detected on physical exam (267). ing, a round or oval intradural mass represents the lower segments of
A third cause is associated bony abnormalities of the craniocervical the spinal cord. CT myelography shows marked bony narrowing of the
junction (271). Other causes of neurologic dysfunction are associ- spinal canal with the small remnant of subarachnoid space running
ated CNS anomalies, which are reported to include occipital encepha- through the extraspinal soft tissues. MR may show a variable degree of
loceles, Chiari malformations, Dandy-Walker malformations, Duane tapering of the spinal cord to a point of marked narrowing or complete
syndrome, nasofrontal and posterior fossa (epi)dermoids, and syrin- focal absence of the cord. However, in most cases, the termination of
gohydromyelia. Associated visceral anomalies include Sprengel defor- the upper segment of the spinal cord is abrupt (Figs. 9-51 and 9-52),
mity (congenital elevation of the scapula, often associated with an and in some cases it appears quite blunted (Fig. 9-52). Both of these
omovertebral bone), cervical ribs, supranumerary digits, tracheal and appearances are similar to the appearance of the spinal cord in patients
proximal bronchial stenosis, sickle sacrum, cleft palate, and various with caudal regression/lumbosacral hypogenesis. (Indeed, this blunt-
renal anomalies (47,267,269,271). ing is one reason that Tortori-Donati et al. (276) have postulated that

Barkovich_Chap09.indd 903 5/6/2011 10:29:05 PM


904 Pediatric Neuroimaging

FIG. 9-51. Segmental spinal dysgenesis. A. Sagit-


tal T1-weighted image shows a high termination of
the spinal cord (white arrow) at the T-2 to T-3 level.
Note how the spinal canal and thecal sac taper rap-
idly (black arrowheads) caudal to the cord termina-
tion. B. Sagittal T1-weighted image in the lower half
of the spine shows a normal-appearing segment of
spinal cord (arrows) in the midlumbar region below
a level where the spinal canal appears again.

segmental spinal dysgenesis and lumbosacral hypogenesis have similar By definition, meningoceles are lined with arachnoid. Arachnoi-
causes; see the section on “Syndrome of Caudal Regression (a complex dal adhesions occasionally occur within the sac and, when unusually
spinal dysraphism)” earlier in this chapter.) Below the segmental agen- thick, may partially obliterate the neck of the sac (277). Because the sac
esis, the bony spinal canal, thecal sac, and spinal cord resume a nor- communicates with the subarachnoid space, it may change in size with
mal appearance (Figs. 9-51 and 9-52). Evaluation of the entire spinal patient position or with Valsalva maneuver.
column is essential, as affected patients may have associated lipomas, The bony abnormalities accompanying meningoceles are usually
dermal sinuses, or hydromyelia (276). limited. They may range from absence of a single spinous process to a
Surgery to decompress the bony narrowing at the site of dysgenesis localized spina bifida or to a multisegmental spina bifida with enlarge-
does not seem to help the patient regain any neurological or urological ment of the spinal canal.
function, but may prevent further neurological deterioration (273–275). The purpose of imaging studies in patients with meningoceles is
(a) to detect the meningocele, (b) to determine its shape, (c) to define
Dorsal Meningocele (a CSD with a subcutaneous associated anomalies of the spinal cord and the bony spinal canal,
(d) to determine the presence or absence of nervous tissue within the
mass)
sac, and (e) to assess the relationship of the sac to the conus med-
Dorsal meningoceles are dorsal herniations of dura, arachnoid, and ullaris and the filum terminale. Although sonography demonstrates
cerebrospinal fluid that extend into the subcutaneous tissues of the most of these details, MR and CT myelography give the most infor-
back. The overlying skin is intact unless secondary skin ulcerations mation. Because it is noninvasive, MRI is the imaging procedure of
arise. By definition, the simple meningocele does not contain nervous choice in the evaluation of these patients. High-resolution images
tissue, although, occasionally, a nerve root may enter the sac before with white CSF, either by T2 weighting or steady-state techniques,
leaving it to reenter the spinal canal en route to its respective neural are ideal to show the location of nerve roots. MR of patients with
foramen (151). Rarely, a nerve root may become adherent to the wall of simple dorsal meningoceles shows an empty meningocele sac, a nor-
the sac. The conus medullaris tends to be in its normal position within mal spinal cord, a normal or nearly normal spinal column, a normal
the spinal canal. The filum terminale occasionally extends into the neck termination of the conus medullaris, and a normal filum terminale
of the sac. Complex meningoceles differ from simple meningoceles by (Fig. 9-53). MR of patients with complex dorsal meningoceles shows
the presence of significant associated anomalies of the spine and, usu- the presence of anomalies within the spinal cord, spinal canal, or
ally, spinal column. spinal column (Fig. 9-54).

Barkovich_Chap09.indd 904 5/6/2011 10:29:06 PM


Chapter 9 • Congenital Anomalies of the Spine 905

FIG. 9-52. Segmental spinal dysgenesis with blunted


termination of the upper segment of the spinal cord:
MR and CT appearances. A. Sagittal T1-weighted
image of the spine shows a blunted cord terminus
(white arrows) in the thoracolumbar region. B. Sagit-
tal T2-weighted image shows the marked narrowing
of the spinal canal (white arrows) and vertebral body
dysgeneses and fusions caudal to the termination of
the upper spinal cord segment. A distal cord segment
(asterisk) is seen below the region of abnormal spi-
nal column. C. Sagittal reformation of a noncontrast
CT scan shows vertebral body dysgenesis and lack of
segmentation with narrowing of the spinal canal and
offset of the upper and lower vertebrae in the lower
lumbar region. D. Axial noncontrast CT image shows
the dysplastic vertebral body and very narrow spinal
canal secondary to dysplastic posterior elements in
the dysgenetic segment of spine.

Barkovich_Chap09.indd 905 5/6/2011 10:29:07 PM


906 Pediatric Neuroimaging

FIG. 9-53. Simple dorsal meningocele.


A. Sagittal T1-weighted image shows a large
ovoid dorsal meningocele (arrows) in the
midthoracic region. The vertebral column
and spinal cord appear normal. B. Axial
T1-weighted image shows absence of neural
tissue within the meningocele sac.

Lateral Meningocele slight weakness. Type 1 neurofibromatosis is present in approximately


85% of patients with lateral thoracic meningoceles (277,278). The
Thoracic Lateral Meningocele preferential thoracic location has been postulated to result from lesser
Thoracic lateral meningoceles are cerebrospinal fluid-filled protrusions development of the paraspinal musculature at this level and the rela-
of dura and arachnoid that extend laterally through an enlarged neu- tively high pressure differential between the negative thoracic pressure
ral foramen. They then course anteriorly through the adjacent inter- and the pressure of the CSF in the thoracic subarachnoid space (279).
costal space into the extrapleural aspect of the thoracic gutter. Males Less common predisposing conditions include Marfan and Ehlers-
and females are equally affected and most commonly present during Danlos syndromes.
the fourth and fifth decades of life. Although usually asymptomatic, Patients with lateral thoracic meningoceles may have a sharply
affected patients may have pain, vague sensory deficits, hyperreflexia, or angled scoliosis in the upper thoracic spine, convex toward the

FIG. 9-54. Complex dorsal meningocele. A and B. Sagittal T1-weighted images show a lumbar dorsal meningocele (large white arrows). The conus med-
ullaris is abnormally low. A cyst (small white arrow) is present in the cord just above the meningocele. C and D. Coronal T1-weighted images show the
intramedullary cyst (small white arrows in C) and multiple lumbar vertebral anomalies (large black arrows).

Barkovich_Chap09.indd 906 5/6/2011 10:29:08 PM


Chapter 9 • Congenital Anomalies of the Spine 907

meningocele, which is located near the apex of the curvature (278). neoplasms constitute 0.15% of all intraspinal tumors (20). Males and
Scalloping of the pedicles, laminae, and vertebral bodies adjacent to the females are affected equally, usually presenting with pain and myelopa-
meningocele often result in an enlarged spinal canal (see Fig. 6-22). thy. All ages are affected.
The size of the meningocele varies markedly from small, nearly Two basic theories have been proposed for the pathogenesis
undetectable meningeal protrusions to enormous cystic masses. Large of teratomas. They may arise by multiplication of displaced germ
thoracic meningoceles may fill an entire hemithorax and compromise cells or of cells left behind during the migration of the Hensen
ventilation, particularly in neonates. Most remain static in size, but node that can give rise to any of the three germinal layers of tis-
occasionally they show slow growth. The meningocele may disappear sue (185,281,282). Alternatively, teratomas might arise from rests
after shunting of hydrocephalus (280). of tissues that have escaped the control of those factors in early
The position of the cord with respect to the meningocele sac is embryonic development that determine cell line. Tissue growth
variable. When scoliosis is present, the cord tends to be situated on the therefore produces diverse tissues that can be more or less well dif-
side opposite the sac. More rarely, the cord may be pulled toward the ferentiated (283).
sac or into the sac by nerve roots at that level (20). The appearance of spinal teratomas is extremely variable. The
tumor may be solid, partially or wholly cystic, unilocular, or multiloc-
Lumbar Lateral Meningocele ular. In general, the more immature the histology, the more the lesion
Lumbar lateral meningoceles are protrusions of dura and arachnoid resembles astrocytomas and ependymomas, with fairly homogeneous,
through one or several enlarged lumbar neural foramina into the lum- enhancing soft tissue and associated cysts (284) (see description of
bar subcutaneous tissue and retroperitoneum. They often occur in spinal cord tumors in Chapter 10). Mature tumors show evidence of
a setting of Marfan syndrome or type 1 neurofibromatosis, but may fat (hyperintense on T1-weighted images) and calcification; bone and
be seen as isolated anomalies. The meningoceles may be unilateral or cartilage may be present, sometimes in the form of a well-defined bone
bilateral, may involve a single neural foramen or multiple adjacent or tooth. When cysts are present, they may be thin or thick walled, and
neural foramina, and may displace adjacent structures (Fig. 9-55). the fluid within them may be clear, milky, or dark. Teratomas may be
Lumbar meningoceles are similar to those in the thorax in that they intramedullary or extramedullary; whatever their location, they usu-
are commonly associated with an expanded spinal canal, erosion of the ally fill the spinal canal at the time of presentation and will produce a
posterior surface of the vertebral bodies, thinning of the neural arches, complete block at myelography. When extramedullary, the tumors are
and enlargement of the neural foramina (Fig. 9-55). often closely adherent to the cord with a poorly defined cleavage plane
formed by tumor, connective tissue, and reactive astrogliosis. In such
cases, it is difficult to differentiate the extramedullary tumor from an
CONGENITAL TUMORS OF THE SPINE intramedullary one. When teratomas occur in the lumbar region, the
roots of the cauda equina frequently adhere to the walls of the tumor
Teratoma
as they are draped over it. Syringomyelia may appear above the level
Teratomas are neoplasms containing tissues belonging to all three ger- of the tumor secondary to restricted CSF flow. Malignant teratomas
minal layers at sites where these tissues do not normally occur. Exclud- are rare and cannot be differentiated from the more common benign
ing sacrococcygeal teratomas (discussed earlier in the chapter), these variant by imaging.

FIG. 9-55. Lumbar lateral meningoceles in a patient with Marfan syndrome. A and B. Coronal T2-weighted images show multiple bilateral hyperintense
meningoceles extending laterally into the pelvis and lumbar paraspinous muscles through enlarged neural foramina. C. Axial T1-weighted image shows the
bilateral meningoceles (arrows) extending laterally through expanded neural foramina. The spinal canal is enlarged.

Barkovich_Chap09.indd 907 5/6/2011 10:29:10 PM


908 Pediatric Neuroimaging

The spinal canal may be normal or may be focally widened by erosion infrequent in the cervical and thoracic regions (153). Sixty percent of
of the pedicles and laminae. Other bony anomalies are uncommon. epidermoids are extramedullary and 40% intramedullary (154).
Although they range in size from tiny subpial growths to huge
mass lesions, at the time of clinical presentation, dermoid and epi-
Dermoid and Epidermoid Tumors
dermoid tumors almost always cause a complete spinal block to
Dermoids are round, ovoid, or multilobular tumors, sometimes cystic, intrathecal contrast when myelography is performed (154). On
that are lined by a squamous epithelium containing skin appendages, imaging studies, dermoids almost always have the appearance of fat
such as hair follicles, sweat glands, and sebaceous glands. Epidermoids on CT, showing low attenuation. However, the appearance is vari-
are tumors lined by a membrane composed of only the superficial (epi- able on MRI, sometimes showing high intensity on T1-weighted
dermal) elements of the skin (151). Dermoids usually cause symptoms images, but more commonly having low to intermediate intensity
before the age of twenty; they occur equally in males and females. Epi- on T1-weighted images and high intensity on T2-weighted images
dermoids develop slightly more slowly, begin to manifest symptoms in (see Fig. 9-13 in section on “Dosal Dermal Sinuses” in this chap-
early adulthood (third to fifth decade), and are more common in males ter). The lack of a fatty signal may be a result of secretions from
(153). Together, dermoid and epidermoid tumors constitute approxi- sweat glands within the tumor causing increased water content.
mately 1% to 2% of spinal cord tumors at all ages and 10% of spinal Epidermoids also have variable signal intensity on MRI (Fig. 9-56),
cord tumors below the age of fifteen. Approximately 20% develop in most commonly isointense to CSF but sometimes hyperintense or
association with dermal sinuses; of those not associated with dermal hypointense; this variability is similar to that seen in intracranial
sinuses, epidermoids are slightly more common than dermoids (153). epidermoids, as described in Chapter 7. Small epidermoids are dif-
When not associated with dorsal dermal sinuses, dermoids and epider- ficult to diagnose on both MRI and CT because they are difficult to
moids may present as a slowly progressive myelopathy or as an acute differentiate from surrounding cerebrospinal fluid on both imag-
onset of a chemical meningitis secondary to rupture of the inflam- ing modalities. Using MRI, larger extramedullary epidermoids can
matory cholesterol crystals from the cyst into the cerebrospinal fluid be identified by slightly altered signal intensity and the displace-
(148,153). ment of the spinal cord or nerve roots by the mass (Fig. 9-56F–H).
Dermoid and epidermoid tumors most commonly arise from con- It can be confirmed by FLAIR or diffusion sequences (156–158).
genital dermal or epidermal rests or from focal expansion of a der- FLAIR will demonstrate the epidermoid as a hyperintense mass sur-
mal sinus. They may also be acquired as iatrogenic lesions resulting rounded by hypointense CSF, while diffusion images will show soft
from implantation of viable dermal and epidermal elements by spinal tissue, not fluid, characteristics of water motion (Fig. 9-56D and E)
needles not provided with trocars (124,285) or as a result of surgery (156–158,286). If FLAIR or diffusion sequences are available, CT
(119,126). myelography should no longer be necessary to detect these tumors.
Epidermoids are distributed fairly uniformly along the spine (upper Neither dermoids nor epidermoids enhance after administration of
thoracic 17%, lower thoracic 26%, lumbosacral 22%, and cauda equina intravenous contrast unless they are or have been infected. Infection
35%) (153). Dermoids, on the other hand, are much more common is much more common if the tumors have developed in association
in the lumbosacral area (60%) and cauda equina (20%), but are quite with dermal sinuses.

FIG. 9-56. Intraspinal epidermoid and dermoid. A–E. Recurrence 12 years after resection of an epidermoid and dorsal dermal sinus. A and B. Sagittal
T2-weighted images from the initial presentation show the focal tethering of the spinal cord by a hypointense band (white arrows) which traverses the tho-
racic posterior osseous defect and communicates with the hypointense tract through the subcutaneous tissues. The associated epidermoid (black arrow) is
slightly more hyperintense than CSF. C–E. Sagittal T2-weighted imaging (C), diffusion-weighted imaging (D), and the apparent diffusion coefficient map
(E) performed 12 years later show the recurrent epidermoid (e) with its central T2 prolongation, peripheral T2 shortening, and reduced diffusion. Adjacent
spinal cord edema is also present.

Barkovich_Chap09.indd 908 5/6/2011 10:29:11 PM


Chapter 9 • Congenital Anomalies of the Spine 909

FIG. 9-56. (Continued) F–H. Dermoid associated


with fibrofatty infiltration of the filum terminale.
F and G. Sagittal and coronal T1-weighted images
show the large dermoid (d) at L-3 to L-4 with a
hyperintense proximal and distal filum terminale
(white arrows). H. Sagittal T2-weighted imaging
shows slightly heterogeneous T2 prolongation in the
dermoid and the hypointense proximal and distal
filum terminale.

Hamartoma of the central canal, exclusive of terminal ventricles (see section “The
Terminal Ventricle” of this chapter). It was located at the junction of the
Hamartomas are malformations composed of an abnormal mixture of ventral 1/3 and dorsal 2/3 of the cord, except when located at the level
those tissue elements that normally occur at the tumor site. Because of the lumbar enlargement, when it was located centrally. The cavities
they grow and develop at the same rate as normal tissues, hamartomas were most typically located at the lower cervical or midthoracic level.
are unlikely to result in compression of adjacent parenchyma; there- The authors recommend that such cavities, when found incidentally,
fore, neurologic deficits and hydrocephalus are usually not present do not require follow-up. We have had similar experiences and concur,
(20,287). although we typically perform a single follow-up examination with
Hamartomas are composed of mesodermal elements such as contrast at 12 months after discovery of an asymptomatic dilation of
bone, cartilage, fat, and muscle. They appear as midline, dorsal, skin- greater than 2 mm, to ensure that this is not the earliest phase of a cord
covered masses, usually found at birth, that are located in the midtho- neoplasm or syrinx. We also make sure that a craniovertebral or spinal
racic, thoracolumbar, or lumbar regions and may be associated with anomaly that might predispose to syrinx formation is not present.
cutaneous angiomas (288). Posterior element deficiency is present in
approximately 60% of patients and the spinal canal is widened in 80%.
Heterotopic bone formation is seen in about half of cases (288). On Clinical Presentation and Course
MRI, they are reportedly isointense with spinal cord on both T1- and
Patients with syringomyelia are uncommonly symptomatic from their
T2-weighted images (289). Rarely, hamartomas may contain function-
syrinx during childhood. Indeed, many childhood cases of syringomy-
ing choroid plexus and present with syringohydromyelia (Fig. 9-57).
elia are discovered due to the progressive scoliosis found in about 30%
of such children (294). The presence of progressive scoliosis, levosco-
SYRINGOHYDROMYELIA liosis (a curve convex to the left), or back pain suggests an underlying
abnormality such as syringomyelia, tumor, or dysraphism (295) and
Definition
are indications for an MRI study. Syringes discovered in children are
Although syringohydromyelia is usually a disease of adults, it can most commonly enlarged central canals of the spinal cord (true hydro-
occur in children and, furthermore, is often associated with (and likely myelia) and associated with postnatally repaired Chiari II (more com-
the result of) a congenital anomaly of the spine or the cervicomedul- monly) or Chiari I (less commonly) malformations (129). In patients
lary junction. Therefore, we have included the topic in this chapter. who are symptomatic, presentation is similar to that in adults, with
Syringohydromyelia is characterized by the presence of longitudinally anesthesia and weakness of the upper limbs, scoliosis, and unsteadiness
oriented cerebrospinal fluid-filled cavities and astrogliosis within the of gait (129,296). Children may also develop slightly dilated central
spinal cord. When the cavity is a dilated central canal of the spinal cord, canals in association with the tight filum terminale syndrome; how-
the term hydromyelia has been applied, reserving the term syringomy- ever, the central canal does not seem to be under increased pressure in
elia for spinal cord cavities extending lateral to, or independent of, the this group of patients and, therefore, the term syringomyelia does not
central canal. On detailed pathologic or radiologic exam, most cavi- seem appropriate. Children with spinal cord tumors can develop asso-
ties are found to involve both parenchyma and central canal. The term ciated syringomyelia or hydromyelia, not to be mistaken for tumoral or
syringohydromyelia reflects this difficulty in classification. Some have peritumoral cysts; typically, the syrinx will resolve after resection of the
used the terms syringomyelia and syrinx in a general manner to repre- tumor. Spinal cord tumors of childhood are discussed in Chapter 10.
sent any spinal cord cyst. We shall use these terms in the same general Most commonly, symptoms of syringomyelia begin in late ado-
sense in this chapter, recognizing that in some patients the term is not lescence or early adulthood and progress irregularly with long inter-
a precisely correct one (85,290–292). vening periods of stability. Sometimes, the syrinx may be picked up
An important point is that not all patients with dilation of the central incidentally; such cases present a therapeutic dilemma, as such patients
canal of the spinal cord have a pathologic condition. Petit-Lacour et al. may remain asymptomatic for 10 years or more (297). In traumatic
(293) found that 12/794 patients (1.5%) had a focal filiform enlargement syringomyelia, a latent period of 20 to 30 years is common before

Barkovich_Chap09.indd 909 5/6/2011 10:29:12 PM


910 Pediatric Neuroimaging

FIG. 9-57. Spinal cord hamartoma with associated syringomyelia. A and B. Coronal T1-weighted images show a dilated spinal cord with CSF intensity
within it (open arrows), both rostral and caudal to a focal heterogeneous narrowing of the cord at the midthoracic level (solid white arrow). The hamartoma
was located at the site of the narrowing. C. Axial T1-weighted image at the level of the hamartoma shows heterogeneity of signal and a bilobed appearance
to the cord resulting from the tumor and the associated syringomyelia. This hamartoma, which contained functioning choroid plexus, is believed to be the
cause of the severe syringomyelia. D. Axial T1-weighted image in the lower thoracic region. The ventral aspect of the spinal cord is markedly thinned and
expanded by the syrinx cavity (open arrows). The dorsal cord (solid arrows) is compressed and displaced posteriorly by the syrinx.

the onset of symptoms is noted (298). Signs and symptoms are vari- subarachnoid space is nearly always found in patients with syringo-
able, depending upon the cross-sectional and vertical extent of cord hydromyelia. The narrowing is most commonly found at the level of
destruction. The classical presentation is of segmental weakness with the foramen magnum. The Chiari I malformation, arachnoiditis, bony
atrophy of the hands and arms, loss of tendon reflexes, and segmen- narrowing of the foramen magnum or spinal canal, intramedullary
tal anesthesia of the dissociative type (loss of pain and temperature tumors, and extramedullary tumors can all be found in patients with
sense with preservation of the sense of touch) over the neck, shoul- syringomyelia (301,303–308).
ders, and arms (299). Pain is often severe and may be most intense Occasionally, syringomyelia undergoes spontaneous decompres-
in an analgesic limb (anesthesia dolorosa). In practice, symptoms are sion, with the appearance of the spinal cord returning to normal
often unilateral, confined to the lower extremities, or absent (300,301). (309–311). The reasons for this spontaneous cure are unknown; per-
Dysesthesias may be present; they are almost always the result of exten- haps spontaneous myelotomy occurs, with drainage of the syrinx into
sion of the syrinx into the dorsolateral quadrant of the spinal cord the subarachnoid space or perhaps the abnormal fluid dynamics (see
(302). The clinical course is unpredictable. Long periods of stability of below) revert to normal. Whatever the cause of the regression, it does
symptoms can be followed by acute deterioration. Narrowing of the not appear to lessen the symptoms of the patient (309,310).

Barkovich_Chap09.indd 910 5/6/2011 10:29:12 PM


Chapter 9 • Congenital Anomalies of the Spine 911

Classification and Causes rhage, and transverse myelitis. Clinically, patients have various com-
binations of long tract and segmental signs that can be related to the
Syringomyelia has many causes, just as cystic spaces in the brain may be level, side, and specific quadrant of spinal cord cavitation (313,314).
of diverse origin. The shape and structure of the spinal cord and spinal Another classification system is based upon the way the syrinx com-
canal have a strong influence on the appearance of the syrinx, making municates with the central canal (315). Communicating syringomyelia
the pathological (and imaging) appearance of the cavities and the clini- is postulated to communicate with the fourth ventricle via the obex at
cal signs similar, regardless of the underlying cause (85,312). some time during the development of the spinal cord cyst, allowing
Milhorat et al. (313) has proposed a classification system for syrin- fluid transmission to the syrinx cavity via this communication. Thus,
gohydromyelia based upon histopathology. These different types of communicating syringes should be centrally located; however, in real-
syringomyelia can be differentiated by axial T1-weighted MR images ity, these hydromyelic cavities often dissect into, and are thus eccentric
(314), making this a useful classification for radiologists. In type 1 within, the surrounding spinal cord tissue (85,291). Communicating
syringes, the dilated central canal is in direct communication with the syringomyelia is the form that is associated with the Chiari malforma-
fourth ventricle. Type 1 syringes are found in association with hydro- tions (Fig. 9-58) and with syndromes with decreased size of the foramen
cephalus; patients are asymptomatic or have nonspecific neurologic magnum, such as achondroplasia (Fig. 9-59) (316,317). Dandy-Walker
signs. Type 2 syringes are dilations of the central canal below a syrinx- malformations (see Chapter 5) can also be associated with syringo-
free segment of spinal cord. Type 2 syringes are found in patients who hydromyelia, in particular when the posterior fossa cyst enlarges and
have alterations of cerebrospinal fluid dynamics in the subarachnoid interferes with flow of CSF through the foramen magnum (318).
space, such as Chiari I malformations and arachnoiditis. (Note that the
foramen magnum can also be blocked by tumors, cysts, or tonsils that
Theories of Pathogenesis
are pushed downward due to chronic downward pressure from hydro-
cephalus, cyst, or mass. Similarly, if the tonsils are pulled downward by Gardner (319,320) has postulated that continued communication
a CSF leak, blockage of the foramen by the cerebellar tonsils can occur; between the fourth ventricle and central canal is the result of a failure of
see Chapter 5, section on “Anomalies of the craniocervical junction.”) the foramina of the fourth ventricle to open at the eighth or ninth week
Type 2 syringes are more likely than type 1 to dissect into parenchyma of fetal life. He describes a “water hammer effect” (caused by cerebro-
outside of the central canal of the spinal cord. These dissections most spinal fluid pulsations) that is transmitted to the central canal through
commonly extend into the posterolateral quadrant of the spinal cord; the obex, causing the canal to dilate. Rupture of the ependymal lin-
their development may correlate with onset of clinical symptoms, most ing results in cyst extension into the substance of the spinal cord (Fig.
commonly dysesthesias. Type 3 syringes are extracanalicular cysts that 9-60B). Gardner’s theory was modified by Williams (321,322), who
arise in the spinal cord parenchyma and do not communicate with the favored a pressure differential between the intracranial and spinal cere-
central canal. Type 3 syringes are typically found in the watershed area brospinal fluid as the cause for formation of the syrinx (Fig. 9-60C).
of the cord (in the central gray matter, dorsal and lateral to the central Williams speculated that coughing and other maneuvers that produce
canal, between the anterior and posterior spinal artery distributions). increased intrathoracic and intra-abdominal pressure result in spinal
Associated myelomalacia is commonly present. The most common epidural venous distention. Distention of the veins within the confined
causes of Type 3 syringes are trauma, infarction, spontaneous hemor- space of the spinal canal causes a rapid rostral displacement of spi-

FIG. 9-58. Syringohydromyelia second-


ary to Chiari I malformation. A. Sagittal
T2-weighted image shows a well-defined,
multiloculated segment of hyperintensity
(white arrowheads), representing a syrinx
in the center of the dilated spinal cord.
The intermediate intensity between the
syrinx and normal spinal cord likely rep-
resents astrogliosis or interstitial edema.
The patient has a Chiari I malformation
with the cerebellar tonsils (white arrows)
extending below the foramen magnum.
B. Axial T2-weighted image through the
cervical spine shows the hyperintense
syringohydromyelic cavity causing expan-
sion of the cervical spinal cord.

Barkovich_Chap09.indd 911 5/6/2011 10:29:14 PM


912 Pediatric Neuroimaging

FIG. 9-59. Syringomyelia secondary to


achondroplasia. A. Sagittal T1-weighted
image shows small clivus, long infundibu-
lum, and big ventricles as described in
Chapter 8. A focal well-defined region of
hypointensity (arrows) is seen extending
from the foramen magnum to the bottom
of C-2. The craniocervical junction is com-
pressed by the narrow foramen magnum.
B. Axial T2-weighted image confirms the
intramedullary cyst (arrow) at the cranio-
cervical junction.

nal fluid. In patients with the Chiari I malformation, or other causes More recently, Heiss et al. (327) have emphasized the importance
of obstruction within the cerebrospinal fluid spaces, the initial surge of the ectopic, compressed cerebellar tonsils in the progression of
forces cerebrospinal fluid into the intracranial space; however, the cere- syringomyelia. They suggest that the tonsils partially occlude the fora-
brospinal fluid does not drain out immediately because of a ball-valve men magnum, then act as pistons to create large cervical subarachnoid
effect resulting from the obstructing lesion. The result of the fluid shift pressure waves that compress the spinal cord and drive the syrinx fluid
is craniospinal pressure dissociation. According to Williams, the higher caudally with each heartbeat. Heiss et al. do not speculate on how the
intracranial pressure forces fluid through the obex into the central syrinx is initially formed.
canal of the spinal cord (321). Furthermore, Williams speculated that The other forms of syringomyelia that have been described are trau-
the engorgement of the epidural veins by increased thoracoabdominal matic syringomyelia, tumor-associated syringomyelia, syringomyelia sec-
pressure compresses the subarachnoid space and spinal cord, forcing ondary to arachnoiditis, and “idiopathic” syringomyelia (315). Clearly,
fluid upward within the syrinx cavity (Fig. 9-60C). If enough fluid is communication with the fourth ventricle via the obex is not the underly-
forced upward with sufficient force, the syrinx cavity will be extended. ing mechanism of the formation of the syrinx in all cases. However, exten-
A rapid upward acceleration of contrast within syrinx cavities and in sion of the cavity within the cord (or even into the brainstem or thalami, a
the subarachnoid space has been demonstrated with coughing, Valsalva, condition known as syringobulbia [328,329]) may well occur via the same
and respiration (321,323). mechanism once the cavity is established. Williams’ proposed mechanism
Ball and Dayan (324) have speculated that cerebrospinal fluid of (a) an obstructing lesion causing pressure dissociation in the subarach-
enters the central canal via the perivascular spaces. They postulated that noid spaces above and below it, and (b) extension of the cavities as a result
increased pressure in the spinal subarachnoid space is generated by an of CSF accelerations from increased thoracoabdominal pressure and dis-
obstructing lesion (such as the ectopic cerebellar tonsils in the Chiari I tention of the epidural venous plexus (Fig. 9-60C) explains the clinical
malformation) acting as a one-way valve, allowing cerebrospinal fluid observation that these cavities frequently extend after severe coughing,
to escape from the basal cisterns into the spinal canal, but blocking straining, or sneezing. This mechanism might also explain the astrogliosis
its return. They proposed that the increased intraspinal pressure then seen in the spinal cord at the caudal and rostral ends of syringes and the
drives cerebrospinal fluid into the central canal (Fig. 9-60D), resulting edema seen in cord at the rostral end of the syrinx (Figs. 9-61 and 9-62)
in an increased intraspinal CSF pressure compared to the brain. The before the syrinx extends rostrally (85) (the “pre-syrinx” [330]). More-
finding of increased pressure within syrinx cavities in a large series of over, the observation that foramen magnum decompression often cures
patients by Milhorat et al. (312) supports this concept, as does animal the syringomyelia without any other therapy supports the theory that
work showing the development of intramedullary edema and subse- syringes are primarily caused by disturbed CSF dynamics.
quent cyst formation after constriction of the thecal sac (325). Supporting these observations, Koyanagi and Houkin (331) have
Aboulker (326) proposed a theory somewhat analogous to that proposed that reduced compliance of the posterior spinal veins due
of Ball and Dayan. Referring to animal experiments demonstrating to blocking of the CSF pulse wave after cardiac systole (see the hydro-
that 30% of cerebrospinal fluid is produced in the central canal of the dynamic theory of hydrocephalus in Chapter 8) results in reduced
cord, he postulated that stenosis at the foramen magnum or elsewhere absorption of CSF via the intramedullary venous channels and that
in the canal inhibits cerebrospinal fluid flow toward intracranial areas this lack of resorption plays an important part in the development of
of resorption and causes increased cerebrospinal fluid pressure in cord edema (presyrinx [330]) and subsequent syrinx formation.
the spinal canal. The cerebrospinal fluid, driven by high intraspinal
pressure, then filters into the spinal cord, either through the paren-
Imaging
chyma or along the posterior nerve roots. Long-standing spinal cord
edema may eventually cause cavitation within the cord parenchyma MR is unquestionably the diagnostic modality of choice in the diagno-
(Fig. 9-60D). sis of and the evaluation of treatment of syringohydromyelia and syrin-

Barkovich_Chap09.indd 912 5/6/2011 10:29:15 PM


Chapter 9 • Congenital Anomalies of the Spine 913

FIG. 9-60. Diagrammatic representations of


pathophysiologic theories concerning syringohydro-
myelia. A. In the normal situation, CSF flows from the
aqueduct into the fourth ventricle, out the fourth ven-
tricular foramina and into the cisterna magna, basilar
cisterns and spinal subarachnoid space. The central
canal of the spinal cord is usually incompletely pat-
ent in older children and adults. B. Gardner’s theory
(319,320) suggests that there is a lack of perforation of
the foramen of Magendie (blocked arrow in inset) that
forces CSF through the obex into the central canal of
the spinal cord. C. Williams (321) suggests that there is
CSF flow cephalad from the spinal subarachnoid space
into the intracranial cisterns but that flow is partially
blocked in the cephalocaudal direction. He suggests
that this cranial-spinal pressure association leads to CSF
being “sucked” into the central canal of the cord from
the fourth ventricle, initiating the syrinx. He further
proposes that the fluid within the syrinx moves both
rostrally and caudally as a result of changes in epidural
venous pressure; when engorged, these veins compress
the subarachnoid space and spinal cord, forcing fluid
upward within the syrinx cavity. Rapid accelerations
result in extension of the syrinx cavity. D. Both Ball
and Dayan and Alboulker believe that the craniospinal
pressure dissociation is present but reversed in direc-
tion from that proposed by Williams. These authors
believe that increased CSF pressure in the spinal CSF
space results in CSF filtering from the subarachnoid
space into the central canal through the spinal cord.

Barkovich_Chap09.indd 913 5/6/2011 10:29:15 PM


914 Pediatric Neuroimaging

FIG. 9-61. Holocord syringohydromyelia with septations due to a Chiari I malformation. A–C. Sagittal T1 FLAIR images of the cervical, thoracic, and
lumbar spine performed at 3T show the enlargement of the entire spinal cord and a large syrinx with multiple septations. D. Axial T2-weighted image shows
increased signal intensity (white arrow) within the syrinx cavity, probably secondary to pulsations of the fluid within the cavity.

FIG. 9-62. Syringomyelia resulting from arachnoiditis. A. Sagittal T1-weighted image shows that the ventral thoracic subarachnoid space (black arrow-
heads) is enlarged. In the kyphotic thoracic region, the dorsal subarachnoid space is usually enlarged and the ventral subarachnoid space is small. The lower
cervical/upper thoracic spinal cord (large white arrows) is enlarged and hypointense, suggesting a “presyrinx” state. B and C. Sagittal T1-weighted (B) and
T2-weighted images a year later show a frank syrinx cavity (s) with an intermediate signal intensity area (presyrinx, p) rostral and caudal to the cavity. In
addition, there has been development of a small syrinx in the midthoracic spinal cord (white arrowhead). The arachnoid loculation ventral to the thoracic
spinal cord can still be seen and appears to have grown.

Barkovich_Chap09.indd 914 5/6/2011 10:29:17 PM


Chapter 9 • Congenital Anomalies of the Spine 915

is diagnosed. The process usually enlarges the affected region of spinal


cord or medulla. Often, there will be multiple incomplete septations
within the syrinx cavity, giving a “beaded” appearance (Figs. 9-61 and
9-63). If T2-weighted sequences are used, increased signal intensity
may be seen within the parenchyma at the rostral or caudal end of
the cavity (Figs. 9-61 and 9-62). This high signal intensity should not
be misinterpreted as tumor; the prolonged T2 relaxation time results
from microcystic or astrogliotic changes at the end of the syrinx cav-
ity, presumably caused by the pulsations of the cyst upon the adjacent
cord. The edema is reversible, so this appearance is sometimes referred
to as a “presyrinx” (330). If flow compensation techniques are not
used, the syrinx cavity may be hypointense on T2-weighted sequences
(Fig. 9-61) as a result of CSF pulsations within the cavity (85,332).
Cine high-resolution imaging techniques, particularly with high field
strength systems, have been reported to increase the visualization of
arachnoidal adhesions (333).
If MR is unavailable or contraindicated, CT myelography can
be used to evaluate the spinal cord; this technique, however, requires
acquisition of delayed images to visualize the cavity. The patient should
be scanned 4 hours after infusion of the water-soluble contrast. After
4 hours, contrast will usually have diffused into the cavity. If a syrinx
is suspected clinically, and it is not detected on the initial scan or the
4-hour delayed scan, a 12-hour delayed or even a 24-hour delayed scan
should be obtained. Even with delayed imaging, however, CT myelog-
raphy is much less sensitive to syringomyelia, does not show the rostral
and caudal extent as well as MR, and is insensitive to associated spinal
cord pathology.
Syringes can be eccentric within the spinal cord, even exophytic,
FIG. 9-63. Posttraumatic syringomyelia. Sagittal T1-weighted image in all forms of syringomyelia (Fig. 9-57) (73). When very eccentric, a
shows a holocord syrinx with multiple septa. The spinal canal is narrowed syrinx may be difficult to differentiate from an extramedullary (arach-
in the upper lumbar region by a fractured vertebral body (arrows) from a noid) cyst. In less severe cases, examining consecutive axial images can
motor vehicle in the past. differentiate the lesions. In an intramedullary cyst (a syrinx) the rostral
and caudal portions of the cyst are clearly intramedullary. When very
eccentric or very large and rounded, the remnant of compressed spinal
gobulbia (85,328). In order to properly assess syringes by standard MRI, cord peripheral to the syrinx cavity may be impossible to see on an MR
it is necessary to obtain thin (≤3 mm) sagittal T1-weighted images, fol- scan and, therefore, the syrinx may be impossible to differentiate from
lowed by axial T1-weighted images. The syrinx cavity can be missed if an arachnoid cyst. In practice, the differentiation is not important for
only sagittal images are obtained. The syrinx appears on MR images the treating physician, however.
as a CSF-intensity collection within the cord (Figs. 9-58, 9-62, and Techniques have been developed that allow assessment of CSF flow
9-63). If the cavity extends into the medulla (Fig. 9-64), syringobulbia in the cerebral ventricles and at the foramen magnum by phase-contrast

FIG. 9-64. Syringobulbia and presyrinx. A and B. Sagittal T2-(A) and T1-(B) weighted images of the brainstem and cervical spine show syringohydro-
myelia in the cervical spinal cord, extending to the medulla (syringobulbia, white arrowheads). Regions of T2 prolongation—interstitial edema—are seen
in the upper medulla and upper thoracic spine (white arrows in A). These areas of “presyrinx” indicate potential extension of the cavity. Artifact from
hardware placed during previous foramen magnum decompression for Chiari I malformation is present posteriorly. C. Coronal T2-weighted imaging of
the brainstem shows the presyrinx (white arrows) above the syringobulbia. D. Axial FLAIR image through the medulla shows the area of presyrinx (white
arrows) within the upper medulla.

Barkovich_Chap09.indd 915 5/6/2011 10:29:19 PM


916 Pediatric Neuroimaging

MR (334–336). The technique used at UCSF is described in Chapter 1 If patients do not have a known history of prior severe trauma and
and illustrated in Chapter 5 (in the section on “Chiari I Malforma- an obstructing lesion cannot be demonstrated, they should be studied
tions”). Using this and similar techniques, flow dynamics of CSF by MR with intravenous contrast. Some patients previously diagnosed
through the foramen magnum or past an obstructive lesion within the with “idiopathic” syringomyelia harbor small spinal cord neoplasms
spinal canal can be evaluated and quantified (337). This technique is that can be shown by contrast-enhanced MR. Moreover, a small but
most useful if a syrinx is not already present, as it helps to identify significant number of patients with syringomyelia and cerebellar ton-
which patients with Chiari I malformations have altered CSF dynamics sillar ectopia (the Chiari I malformation) have spinal cord neoplasms
at the foramen magnum and are, therefore, at risk to develop syrin- as well (85). Such patients will not improve unless the neoplasm, which
gomyelia (338). It has been demonstrated that the foramen magnum is the primary cause of their syrinx, is treated. If the patient has not
obstruction in Chiari I malformations may be reduced when the head improved after foramen magnum decompression, paramagnetic con-
is in extension (339). We have also seen patients in whom CSF flow trast should always be given on the follow-up MR scan.
improves with flexion. Therefore, in patients in whom a Chiari I mal-
formation is present and CSF flow at the craniocervical junction is
normal in the neutral position, additional sequences with the patient’s REFERENCES
head in flexion and extension may be valuable.
Gated phase-contrast CSF flow studies are also useful when syringes 1. Bainter JJ, Boos A, Kroll KL. Neural induction takes a transcriptional twist.
Dev Dyn 2001;222:315–327.
are idiopathic or associated with arachnoidal scarring. Such scarring may
2. Copp AJ, Greene ND, Murdoch JN. The genetic basis of neurulation. Nat
be the result of prior trauma, infection, surgery, or hemorrhage; as dis- Rev Genet 2003;4:784–793.
cussed above, scarring probably causes syrinx formation by altering CSF 3. Kibar Z, Capra V, Gros P. Toward understanding the genetic basis of neural
flow dynamics. Impaired CSF flow appears as lack of pulsatile CSF flow tube defects. Clin Genet 2007;71:295–310.
in the affected region. If MR is contraindicated or is difficult because of 4. Keller R. Shaping the vertebrate body plan by polarized embryonic cell
extensive metallic instrumentation of the spine, myelography can be used movements. Science 2002;298:1950–1954.
to demonstrate the scarring, which usually appears as a myelographic 5. Wallingford JB, Fraser SE, Harland RM. Convergent extension: the molecu-
block. The location of the scarring is important, as adequate treatment lar control of polarized cell movements during embryonic development.
requires microsurgical dissection of the arachnoid scar and decompres- Dev Cell 2002;2:695–706.
sion of the subarachnoid space with a fascia lata graft (340). 6. Colas JF, Schoenwolf GC. Towards a cellular and molecular understanding
of neurulation. Dev Dyn 2001;221:117–145.
Static MRI and MR CSF flow studies can also be used to assess
7. Ybot-Gonzalez P, Cogram P, Gerrelli D, et al. Sonic hedgehog and the
the foramen magnum after decompression. The MR appearance of the molecular regulation of mouse neural tube closure. Development 2002;129:
decompressed foramen magnum is that of a widened foramen; the pos- 2507–2517.
terior margin of the foramen is located high above its normal position 8. Ybot-Gonzalez P, Gaston-Massuet C, Girdler G, et al. Neural plate mor-
secondary to removal of bone. Some “kinking” of the cervicomedullary phogenesis during mouse neurulation is regulated by antagonism of Bmp
junction and “slumping” of the cerebellum into the enlarged foramen signaling. Development 2007;134:3203–3211.
magnum is commonly seen (341,342). Some CSF should be seen ante- 9. Wallingford JB. Neural tube closure and neural tube defects: studies in ani-
rior to the cervicomedullary junction and posterior to the cerebellum mal models reveal known knowns and known unknowns. Am J Med Genet
at all levels. The CSF flow study should show unimpeded flow through C Semin Med Genet 2005;135:59–68.
the foramen with the head in the flexed position. 10. Van Allen M, Kalousek D, Chernoff G, et al. Evidence for multi-site closure
of the neural tube in humans. Am J Med Genet 1993;47:723–743.
In all patients in whom a syrinx is discovered radiologically, a
11. Seller MJ. Further evidence for an intermittent pattern of neural tube
search should be made for the underlying cause of the syrinx. First, the closure in humans. J Med Genet 1995;32:205–207.
foramen magnum should be evaluated; the position of the cerebellar 12. Nakatsu T, Uwabe C, Shiota K. Neural tube closure in humans initiates at
tonsils should be noted. The tonsils should have a rounded appear- multiple sites: evidence from human embryos and implications foor the
ance and should lie no more than 6 mm below the level of the fora- pathogenesis of neural tube defects. Anat Embryol 2000;201:455–466.
men magnum as measured on sagittal images (343,344). MR of the 13. Golden J, Chernoff G. Intermittent pattern of neural tube closure in two
craniocervical junction may also show stenosis of the foramen mag- strains of mice. Teratology 1993;47:73–80.
num, as in achondroplasia, masses at the level of the foramen mag- 14. Copp AJ, Greene ND. Genetics and development of neural tube defects.
num, such as schwannomas and meningiomas, cysts, herniated tonsils, J Pathol 2010;220:217–230.
or arachnoidal loculations. Any mass or narrowing at the foramen mag- 15. Müller F, O’Rahilly R. The first appearance of the human nervous system at
stage 8. Anat Embryol 1981;163:1–13.
num can cause craniospinal pressure dissociation. If arachnoiditis, either
16. Müller F, O’Rahilly R. The first appearance of the major divisions of the
from previous trauma or infection, is the cause of syringohydromyelia, human brain at stage 9. Anat Embryol 1983;168:419–432.
arachnoid loculations can usually be identified at some point within 17. Müller F, O’Rahilly R. The first appearance of the neural tube and optic
the spinal canal. Although the loculations are isointense with free- primordium in the human embryo at stage 10. Anat Embryol 1985;172:
flowing cerebrospinal fluid, they can be identified by their mass effect 157–169.
upon the spinal cord (Fig. 9-62). The cord will be slightly displaced and 18. Müller F, O’Rahilly R. The development of the human brain and the clo-
deformed by adjacent loculations. sure of the rostral neuropore at stage 11. Anat Embryol 1986;175:205–222.
As mentioned earlier in this section, MRI shows mild dilatation 19. Müller F, O’Rahilly R. The development of the human brain, the closure of the
of the central canal in up to 25% of patients with a tethered spinal caudal neuropore, and the beginning of secondary neurulation at stage 12.
cord (Fig. 9-31) (82). The exact cause of this dilatation is not known. Anat Embryol 1987;176:413–430.
20. Naidich TP, McLone DG, Harwood-Nash DC. Spinal dysraphism. In: New-
However, the mild dilatation of the central canal in these patients is
ton TH, Potts DG, eds. Modern Neuroradiology: Computed Tomography of
not felt to be significant, and the cause of the tethering should be dealt the Spine and Spinal Cord, Vol. 1. San Anselmo: Clavadel, 1983:299–353.
with primarily. Indeed, the central canal typically returns to normal 21. Nievelstein RAJ, Hartwig NG, Vermeij-Keers C, Valk J. Embryonic develop-
once the cord is untethered (209). Only if symptoms persist after the ment of the mammalian caudal neural tube. Teratology 1993;48:21–31.
release of the tethered cord, should the dilated central canal be evalu- 22. Chung YN, Lee DH, Yang HJ, et al. Expression of neuronal markers in the
ated further. secondary neurulation of chick embryos. Childs Nerv Syst 2008;24:105–110.

Barkovich_Chap09.indd 916 5/6/2011 10:29:20 PM


Chapter 9 • Congenital Anomalies of the Spine 917

23. Sapunar D, Vilovic K, England M, Saraga-Babic M. Morphological diversity 51. Holtzman RN, Stein BM. The Tethered Spinal Cord. New York:
of dying cells during regression of the human tail. Ann Anat 2001;183: Thieme-Stratton, 1985.
217–222. 52. Pang D, Wilberger JE, Jr. Tethered cord syndrome in adults. J Neurosurg
24. Müller F, O’Rahilly R. The development of the human brain from a closed 1982;57:32–37.
neural tube at stage 13. Anat Embryol 1988;177:203–224. 53. Pang D. Tethered cord syndrome in adults. In: Holtzman RNN, Stein BM,
25. Müller F, O’Rahilly R, Benson DR. The early origin of vertebral anomalies, eds. The Tethered Spinal Cord. New York: Thieme-Stratton, 1985:99–115.
as illustrated by a ‘butterfly vertebra’. J Anat 1986;149:157–169. 54. Golonka N, Haga L, Keating R, et al. Routine MRI evaluation of low imper-
26. Watanabe Y, Duprez D, Monsoro-Burq A-H, Vincent C, Le Douarin NM. forate anus reveals unexpected high incidence of tethered spinal chord.
Two domains in vertebral development: antagonistic regulation by SHH J Pediatr Surg 2002;37:966–969.
and BMP4 proteins. Development 1998;125:2631–2639. 55. Umur AS, Selcuki M, Selcuki D, Beduk A, Doganay L. Adult tethered cord
27. Dias MS. Normal and abnormal development of the spine. Neurosurg Clin syndrome mimicking lumbar disc disease. Childs Nerv Syst 2008;24:841–
N Am 2007;18:415–429. 844.
28. Moe JH, Winter RB, Bradford DS, et al. Scoliosis and Other Spinal Deformi- 56. Schut L, Pizzi FJ, Bruce DA. Occult spinal dysraphism. In: McLaurin RL, ed.
ties. Philadelphia: Saunders, 1978. Myelomeningocele. New York: Grune & Stratton, 1977:349–368.
29. Sensenig EC. The early development of the meninges of the spinal cord in 57. French BN. The embryology of spinal dysraphism. Clin Neurosurg
human embryos. Contrib Embryol 1951;34:147–157. 1983;30:295–340.
30. Della Rovere D. Due casi di lipoma della pia meninge. Studio anatomico 58. Anderson FM. Occult spinal dysraphism: diagnosis and management.
e critico. Clin Med Ital 1902;41:129–143. J Pediatr 1968;73:163–178.
31. Tortori-Donati P, Rossi A, Cama A. Spinal dysraphism: a review of neurora- 59. Korsvik HE, Keller MS. Sonography of occult dysraphism in neonates and
diological features and embryological correlations and proposal for a new infants with MR imaging correlation. Radiographics 1992;12:297–306.
classification. Neuroradiology 2000;42:471–491. 60. Algra PR, Hageman LM. Gadopeneteate dimeglumine-enhanced MR imag-
32. Nait-Oumesmar B, Stecca B, Fatterpekar G, Naidich T, Corbin J, Lazzarini ing of spinal dermal sinus tract. Am J Neuroradiol 1991;12:1025–1026.
R. Ectopic expression of Gcm1 induces congenital spinal cord abnormali- 61. Altman NR, Altman DH. MR imaging of spinal dysraphism. Am J Neurora-
ties. Development 2002;129:3957–3964. diol 1987;8:533–538.
33. Karabagli H, Karabagli P, Ladher R, Schoenwolf G. Comparison of the 62. Aoki S, Machida T, Sasaki Y, et al. Enterogenous cyst of cervical spine:
expression patterns of several fibroblast growth factors during chick gas- clinical and radiological aspects (including CT and MR). Neuroradiology
trulation and neurulation. Anat Embryol 2002;205:365–370. 1987;29:291–293.
34. Wallingford J, Harland R. Neural tube closure requires Dishevelled- 63. Arredondo F, Haughton VM, Hemmy DC, Zeleya B, Williams AL. The
dependent convergent extension of the midline. Development 2002;129: computed tomography appearance of diastematomyelia. Radiology
5815–5825. 1980;136:685–688.
35. Finnell R, Junker W, Wadman L, Cabrera R. Gene expression profiling 64. Barkovich AJ, Edwards MSB, Cogen PH. MR evaluation of spinal dermal
within the developing neural tube. Neurochem Res 2002;27:1165–1180. sinus tracts in children. Am J Neuroradiol 1991;12:123–129.
36. Catala M. Genetic control of caudal development. Clin Genet 65. Brooks BS, Duvall ER, El Gammal T, Garcia JH, Gupta KL, Kapila A. Neu-
2002;61:89–96. roimaging features of neurenteric cysts: analysis of nine cases and review of
37. Bassuk AG, Kibar Z. Genetic basis of neural tube defects. Semin Pediatr the literature. Am J Neurorad 1993;14:735–746.
Neurol 2009;16:101–110. 66. Brunberg JA, Di Pietro MA, Venes JL, et al. Intramedullary lesions of the
38. McLone DG. The subarachnoid space: a review. Child’s Brain 1980;6: pediatric spinal cord: correlation of findings from MR imaging, intraopera-
113–130. tive sonography, surgery, and histologic study. Radiology 1991;181:573–579.
39. Catala M. Embryogenesis: why do we need a new explanation for the emer- 67. Davis PC, Hoffman JC, Jr, Ball TI, et al. Spinal abnormalities in pediatric
gence of spinal bifida with lipoma. Childs Nerv Syst 1997;13:336–340. patients: MR imaging findings compared with clinical, myelographic, and
40. Naidich TP, Mclone DG, Mutleur S. A new understanding of dorsal surgical findings. Radiology 1988;166:679–685.
dysraphism with lipoma (lipomyeloschisis): radiological evaluation and 68. DiPietro MA. The conus medullaris: normal US findings throughout child-
surgical correction. Am J Neuroradiol 1983;4:103–116. hood. Radiology 1993;188:149–153.
41. Li YC, Shin SH, Cho BK, et al. Pathogenesis of lumbosacral lipoma: a test of 69. Gryspeerdt GL. Myelographic assessment of occult forms of spinal dysra-
the “premature dysjunction” theory. Pediatr Neurosurg 2001;34:124–130. phism. Acta Radiol 1963;1:702–717.
42. van Aalst J, Beuls EA, Cornips EM, et al. The spinal dermal-sinus-like stalk. 70. Gusnard DA, Naidich TP, Yousefzadeh DK, Haughton VM. Ultrasonic
Childs Nerv Syst 2009;25:191–197. anatomy of the normal neonatal and infant spine: correlation with cryo-
43. Long FR, Hunter JV, Mahboubi S, Kalmus A, Templeton JM, Jr. Tethered microtome sections and CT. Neuroradiology 1986;28:493–511.
cord and associated vertebral anomalies in children and infants with imper- 71. Hall WA, Albright AL, Brunberg JA. Diagnosis of tethered cords by mag-
forate anus: evaluation with MR imaging and plain radiolgraphy. Radiology netic resonance imaging. Surg Neurol 1988;30:60–64.
1996;200:377–382. 72. Harwood-Nash DC, Fitz CR, Resjo M, Chuang S. Congenital spinal and
44. Nievelstein RAJ, Valk J, Smit ME, Vermeij-Keers C. MR of the caudal cord lesions in children and computed tomographic metrizamide myelog-
regression syndrome: embryologic implications. Am J Neuroradiol 1994;15: raphy. Neuroradiology 1978;16:69–70.
1021–1029. 73. Heinz ER, Curnes J, Friedman A, Oakes J. Exophytic syrinx, an extreme
45. Warf BC, Scott RM, Barnes PD, Hendren WH, III. Tethered spinal cord in form of syringomyelia: CT, myelographic, and MR imaging features. Radi-
patients with anorectal and urogenital malformations. Pediatr Neurosurg ology 1992;183:243–246.
1993;19:25–30. 74. Hilal SK, Marton D, Pollack E. Diastematomyelia in children: radiographic
46. Chestnut R, James HE, Jones KL. The VATER association and spinal dys- study of 34 cases. Radiology 1974;112:609–621.
raphia. Pediatr Neurosurg 1992;18:144–148. 75. Lantos F, Epstein F, Kory LA. MR imaging of intradural spinal lipoma. Neu-
47. Ulmer JL, Elster AD, Ginsberg LE, Williams DW, III. Klippel-Feil syndrome: rosurgery 1987;20:469–472.
CT and MR of acquired and congenital abnormalities of cervical spine and 76. Lee K, Gower DJ, McWhorter JM, Albertson DA. The role of MR imaging
cord. J Comput Assist Tomogr 1993;17:215–224. in the diagnosis and treatment of anterior sacral meningocele. J Neurosurg
48. Barkovich AJ, Raghavan N, Chuang SH. MR of lumbosacral agenesis. 1988;69:628–631.
Am J Neuroradiol 1989;10:1223–1231. 77. Naidich TP, Fernbeck SK, McLone DG, Shkolnik A. John Caffey Award.
49. Pang D. Sacral agenesis and caudal spinal cord malformations. Neurosur- Sonography of the caudal spine and back: congenital anomalies in children.
gery 1993;32:755–779. Am J Neuroradiol 1984;5:221–234.
50. Klekamp J, Raimondi AJ, Samii M. Occult dysraphism in adulthood: clini- 78. Naidich TP, Doundoulakis SH, Poznanski AK. Intraspinal masses: efficacy
cal course and management. Childs Nerv Syst 1994;10:312–320. of plain spine radiography. Pediatr Neurosci 1985/86;12:10–17.

Barkovich_Chap09.indd 917 5/6/2011 10:29:20 PM


918 Pediatric Neuroimaging

79. Naidich TP, Radkowsky MA, Britton J. Real time sonographic display of 106. Anthony TE, Heintz N. The folate metabolic enzyme ALDH1L1 is
caudal spine anomalies. Neuroradiology 1986;28:512–527. restricted to the midline of the early CNS, suggesting a role in human
80. Peacock WJ, Murovic JA. Magnetic resonance imaging in myelocystoceles: neural tube defects. J Comp Neurol 2007;500:368–383.
report of two cases. J Neurosurg 1989;70:804–807. 107. Czeizel AE, Dudás I. Prevention of the first occurrence of neural-tube
81. Quencer RM, Montalvo BM, Naidich TP, Donovan Post MJ. Intraop- defects by periconceptional vitamin supplementation. N Engl J Med
erative sonography in spinal dysraphism and syringohydromyleia. Am J 1992;327:1832–1835.
Neuroradiol 1987;8:329–337. 108. Group MVR. Prevention of neural tube defects: results of the medical
82. Raghavan N, Barkovich AJ, Edwards MS, Norman D. MR imaging in the research council vitamin study. Lancet 1991;338:131–137.
tethered cord syndrome. Am J Neuroradiol 1989;10:27–36. 109. (CDC) CfDCaP. Spina bifida and anencephaly before and after folic acid
83. Samuelsson L, Bergstrom K, Thomas KA, Hemmingson A, Wallensten R. mandate–United States, 1995–1996 and 1999–2000. MMWR Morb Mortal
MR imaging of syringohydromyelia and Chiari malformations in myelom- Wkly Rep 2004;53:362–365.
eningocele patients with scoliosis. Am J Neuroradiol 1987;8:539–546. 110. Shaw GM, Velie EM, Schaffer D. Risk of neural tube defect-affected preg-
84. Scotti G, Musgrave MA, Harwood-Nash DC, Fitz CR, Chuang SH. nancies among obese women. J Am Med Assn 1996;275:1093–1096.
Diastematomyelia in children: metrizamide and CT metrizamide myelog- 111. Werler MM, Louik C, shapiro S, Mitchell AA. Prepregnant weight in rela-
raphy. Am J Neuroradiol 1980;1:403–410. tion to risk of neural tube defects. J Am Med Assn 1996;275:1089–1092.
85. Sherman JL, Barkovich AJ, Citrin CM. The MR appearance of syringohy- 112. Mojtabai R. Body mass index and serum folate in childbearing age women.
dromyelia: new observations. Am J Neuroradiol 1986;7:985–995. Eur J Epidemiol 2004;19:1029–1036.
86. Sigal R, Denys A, Halimi P, Shapeero L, Doyon D, Boudghene F. Ventricu- 113. Pigott H. The natural history of scoliosis in myelodysplasia. J Bone Joint
lus terminalis of the conus medullaris: MR imaging in four patients with Surg [Br] 1980;62B:54–58.
congenital dilatation. Am J Neuroradiol 1991;12:733–737. 114. Hoppenfeld S. Congenital kyphosis in myelomeningocele. J Bone Joint
87. Tadmore R, Davis KR, Roberson G, Chapman PH. The diagnosis of Surg [Br] 1967;49:276–280.
diastematomyelia by computed tomography. Surg Neurol 1977;8:434–436. 115. Cameron AH. The Arnold-Chiari and other neuroanatomical malforma-
88. Uchino A, Mori T, Ohno M. Thickened fatty filum terminale: MR imag- tions associated with spina bifida. J Pathol Bacteriol 1957;73:195–211.
ing. Neuroradiology 1991;33:331–333. 116. Emery JL, Lendon RG. The local cord lesion in neurospinal dysraphism
89. Wilson DA, Prince JR. MR imaging determination of the location of (meningomyelocele). J Pathol 1973;110:83–96.
the normal conus medullaris throughout childhood. Am J Roentgenol 117. Pang D. Split cord malformation: part II: clinical syndrome. Neurosurgery
1989;152:1029–1032. 1992;31:481–500.
90. Rohrschneider WK, Forsting M, Darge K, Tröger J. Diagnostic value of 118. Duckworth T, Sharrard WJ, Lister J, Seymour N. Hemimyelocele. Dev Med
spinal US: comparative study with MR imaging in pediatric patients. Child Neurol 1968;10(Suppl 16):69–75.
Radiology 1996;200:383–388. 119. McLone DG, Dias MS. Complications of myelomeningocele closure. Pedi-
91. Nakahara D, Yonezawa I, Kobanawa K, et al. Magnetic resonance imaging atr Neurosurg 1991–92;17:267–273.
evaluation of patients with idiopathic scoliosis: a prospective study of four 120. McLone DG, Herman JM, Gabrieli AP, Dias L. Tethered cord as a cause of
hundred seventy-two outpatients. Spine 2010 Epub 2010 May 14. scoliosis in children with a myelomeningocele. Pediatr Neurosurg 1990–91;
92. Morcuende JA, Dolan LA, Vazquez JD, Jirasirakul A, Weinstein SL. A prog- 16:8–13.
nostic model for the presence of neurogenic lesions in atypical idiopathic 121. Levy LM, DiChiro G, McCullough DC, Dwyer AJ, Johnson DL, Yang
scoliosis. Spine (Phila Pa 1976) 2004;29:51–58. SSL. Fixed spinal cord: diagnosis with MR imaging. Radiology 1988;169:
93. Winter R, Lonstein J, Heithoff K, Kirkham J. Magnetic resonance imaging 773–778.
evaluation of the adolescent patient with idiopathic scoliosis before spinal 122. Brunelle F, Sebag G, Baraton J, Carteret M, Martinat P, Pierre-Kahn A.
instrumentation and fusion. Spine 1997;22:855–858. Lumbar spinal cord motion measurement with phase contrast MR imag-
94. Freund M, Hähnel S, Thomsen M, Sartor K. Treatment planning in severe ing in normal children and in children with spinal lipomas. Pediatr Radiol
scoliosis: the role of MRI. Neuroradiology 2001;43:481–484. 1996;26:265–270.
95. McLone DG, Knepper PA. Role of complex carbohydrates and neurula- 123. Chadduck WM, Roloson GJ. Dermoid of the filum terminale of a new-
tion. Pediat Neurosci 1985–86;12:2–9. born with myelomeningocele. Pediatr Neurosurg 1993;19:81–83.
96. Copp AJ. Neural tube defects. Trends Neurosci 1993;16:381–383. 124. Halcrow SJ, Crawford PJ, Craft AW. Epidermoid spinal cord tumor after
97. Meuli M, Meuli-Simmen C, Hutchens GM, et al. In utero surgery res- lumbar puncture. Arch Dis Child 1985;60:978–979.
cues neurological function at birth in sheep with spina bifida. Nat Med 125. Kirsch WM, Hodges FJ, III. An intramedullary epidermal inclusion cyst
1995;1:342–347. of the thoracic cord associated with a previously repaired meningocele.
98. Hutchins GM, Meuli M, Meuli-Simmen C, Jordan MA, Heffez DS, J Neurosurg 1966;24:1018–1020.
Blakemore KJ. Acquired spinal cord injury in human fetuses with myelom- 126. Scott RM, Wolpert SM, Bartoshesky LE, Zimbler S, Klauber GT. Dermoid
eningocele. Pediatr Pathol Lab Med 1996;16:701–712. tumors occurring at the site of previous myelomeningocele repair. J Neu-
99. Bouchard S, Davey M, Rintoul N, Walsh D, Rorke L, Adzick N. Correction rosurg 1986;65:779–783.
of hindbrain herniation and anatomy of the vermis after in utero repair of 127. Awwad EE, Backer R, Archer CR. The imaging of an intraspinal cervi-
myelomeningocele in sheep. J Pediatr Surg 2003;38:451–458. cal dermoid tumor by MR, CT, and sonography. Comput Radiol 1987;11:
100. Tulipan N, Sutton L, Bruner J, Cohen B, Johnson M, Adzick N. The effect 169–173.
of intrauterine myelomeningocele repair of the incidence of shunt- 128. Vion-Drury J, Vincentelli F, Jiddane M, et al. MR imaging of epidermoid
dependent hydrocephalus. Pediatr Neurosurg 2003;38:27–33. cysts. Neuroradiology 1987;29:333–338.
101. Danzer E, Adzick NS, Rintoul NE, et al. Intradural inclusion cysts fol- 129. Hoffman HJ, Neil J, Crone KR, et al. Hydrosyringomyelia and its manage-
lowing in utero closure of myelomeningocele: clinical implications and ment in childhood. Neurosurgery 1987;21:347–351.
follow-up findings. J Neurosurg Pediatr 2008;2:406–413. 130. Hall PV, Lindseth R, Campbell R, Kalsbeck JE, Desousa A. Scoliosis and
102. Danzer E, Finkel RS, Rintoul NE, et al. Reversal of hindbrain herniation hydrocephalus in myelocele patients; the effects of ventricular shunting.
after maternal-fetal surgery for myelomeningocele subsequently impacts J Neurosurg 1979;50:174–178.
on brain stem function. Neuropediatrics 2008;39:359–362. 131. Hall PV, Campbell RL, Kalsbeck JE. Meningomyelocele and progres-
103. Lynch SA. Non-multifactorial neural tube defects. Am J Med Genet C sive hydromyelia: progressive paresis in myelodysplasia. J Neurosurg
Semin Med Genet 2005;135:69–76. 1975;43:457–463.
104. Mitchell LE. Epidemiology of neural tube defects. Am J Med Genet C 132. Batnitzky S, Hall PV, Lindseth RE, Wellman HN. Meningomyelocele and
Semin Med Genet 2005;135. syringohydromyelia: some radiologic aspects. Radiology 1976;120:351–357.
105. Fleming A, Copp AJ. Embryonic folate metabolism and mouse neural tube 133. Barnes PD, Brody JD, Jaramillo D, Akbar JU, Emans JB. Atypical idiopathic
defects. Science 1998;280:2107–2109. scoliosis: MR imaging evaluation. Radiology 1993;186:247–253.

Barkovich_Chap09.indd 918 5/6/2011 10:29:20 PM


Chapter 9 • Congenital Anomalies of the Spine 919

134. Nokes SR, Murtaugh FR, Jones JD, III, et al. Childhood scoliosis: MR 162. Andronikou S, Wieselthaler N, Fieggen GG. Cervical spinal bifida
imaging. Radiology 1987;164:791–797. cystica: MRI differenitation of the subtypes in children. Childs Nerv Syst
135. McLone DG, Knepper PA. The cause of Chiari II malformation: a unified 2006;22:379–384.
theory. Pediatr Neurosci 1989;15:1–12. 163. Steinbok P, Cochran DD. The nature of congenital posterior cervical or
136. MacKenzie NG, Emery JL. Deformities of the cervical cord in children with cervicothoracic midline cutaneous mass lesions. Report of 8 cases. J Neu-
neurospinal dysraphism. Dev Med Child Neurol 1971;25(Suppl):58–67. rosurg 1991;75:206–212.
137. Naidich TP, McLone DG, Fulling KH. The Chiari II malformation: Part IV. 164. Sun JCL, Steinbok P, Cochrane DD. Cervical myelocystoceles and menin-
The hindbrain deformity. Neuroradiology 1983;25:179–197. goceles: long term follow-up. Pediatr Neurosurg 2000;33:118–122.
138. Wolpert SM, Anderson M, Scott RM, Kwan ES, Runge VM. The Chiari II 165. Feltes CH, Fountas KN, Dimopoulos VG, et al. Cervical meningocele in
malformation: MR imaging evaluation. Am J Neuroradiol 1987;8: association with spinal abnormalities. Childs Nerv Syst 2004;20:357–361.
783–791. 166. Emery JL, Lendon RG. Lipomas of the cauda equina and other fatty
139. Variend S, Emery JL. Cervical dislocation of the cerebellum in children tumors related to neurospinal dysraphism. Dev Med Child Neurol 1969;11
with meningomyelocele. Teratology 1976;13:281–290. (Suppl 20):62–70.
140. Emery JL, MacKenzie N. Medullo-cervical dislocation deformity (Chiari II 167. Ehni G, Love JG. Intraspinal lipomas: report of cases, review of the
deformity) related to neurospinal dysraphism (meningomyelocele). Brain literature, and clinical and pathologic study. Arch Neurol Psychiatr
1973;96:155–164. 1945;53:1–28.
141. Daniel PM, Strich SJ. Some observations on the congenital deformity of 168. Leveuf JB. Spina bifida avec tumeur. In: Leveuf J, ed. Études sur le spina
the central nervous system known as the Arnold-Chiari malformation. bifida. Paris: Masson, 1937:93–106.
J Neuropath Exp Neurol 1958;17:256–266. 169. Pierre-Kahn A, Zerah M, Renier D, et al. Congenital lumbosacral lipomas.
142. Wright RL. Congenital dermal sinuses. Prog Neurol Surg 1971;4:175–191. Childs Nerv Syst 1997;13:298–335.
143. Haworth JC, Zachary RB. Congenital dermal sinuses in children: their 170. Arai H, Sato K, Okuda O, et al. Surgical experience of 120 patients with
relation to pilonidal sinuses. Lancet 1955;2:10–14. lumbosacral lipomas. Acta Neurochirurgica 2001;143:857–864.
144. Walker AE, Bucy PC. Congenital dermal sinuses: source of spinal menin- 171. Chapman PH. Congenital intraspinal lipomas: anatomic considerations
geal infection and subdural abscesses. Brain 1934;57:401–421. and surgical treatment. Childs Brain 1982;9:37–47.
145. Mount LA. Congenital dermal sinuses as a cause of meningitis, intraspinal 172. Knittle J, Timmers K, Ginsberg-Fellner F. The growth of adipose tissue
abscess and intracrania abscess. J Am Med Assn 1949;139:1263–1268. in children and adolescents. Cross sectional and longitudinal studies of
146. Bean JR, Walsh JW, Blacker HM. Cervical dermal sinus and intramedul- adipose cell number and size. J Clin Invest 1979;63:239–246.
lary spinal cord abscess: case report. Neurosurgery 1979;5:60–62. 173. Rolland-Cachera MF, Bellisle F, Fricker J. Obésité. In: Ricour C, Ghisolfi J,
147. Aryan H, Jandial R, Farin A, Chen J, Granville R, Levy M. Intradural cra- Putet G, eds. Traité de nutrition pédiatrique. Paris: Monine, 1993.
nial congenital dermal sinuses: diagnosis and management. Childs Nerv 174. Leung E, Sgouros S, Williams S, Johnson K. Spinal lipoma misinterpreted
Syst 2006;22:243–247. as a meningomyelocele on antenatal MRI scan in a baby girl. Childs Nerv
148. Scotti G, Harwood-Nash DC, Hoffman HJ. Congenital thoracic dermal Syst 2002;18:361–363.
sinus: diagnosis by computer assisted metrizamide myelography. J Com- 175. Endoh M, Iwasaki Y, Koyanagi I, Hida K, Abe H. Spontaneous shrinkage
put Assist Tomogr 1980;4:675–677. of lumbosacral lipoma in conjunction with a general decrease in body fat:
149. Schwartz HG. Congential tumors of the sponal cord in infants. Ann Surg case report. Neurosurgery 1998;43:150–152.
1952;136:183–192. 176. Guiffre R. Intradural spinal lipomas: review of the literature (99 cases) and
150. Matson DD. Neurosurgery of Infancy and Childhood, 2nd ed. Springfield: report of an additional one. Acta Neurochir 1966;14:69–95.
Charles C. Thomas, 1969. 177. Kabir SM, Thompson D, Rezajooi K, Casey AT. Non-dysraphic intra-
151. Friede RL. Developmental Neuropathology, 2nd ed. Berlin: Springer-Verlag, dural spinal cord lipoma: case series, literature review and guidelines for
1989. management. Acta Neurochir (Wien) 2010;152:1139–1144.
152. Harwood-Nash DC, Fitz CR. Normal spine, abnormal spine, myelog- 178. Fujiwara F, Tamaki N, Nagashima T, Nakamura M. Intradural spinal
raphy, mass lesions of the spinal canal. In: Harwood-Nash DC, Fitz CR, lipomas not associated with spinal dysraphism: a report of four cases.
eds. Neuroradiology in Infants and Children, vol 3. St Louis: Mosby, 1976: Neurosurgery 1995;37:1212–1215.
1054–1227. 179. Knierim DS, Wacker M, Peckham N, Bedros AA. Lumbosacral intramed-
153. List CF. Intraspinal epidermoids, dermoids, and dermal sinuses. Surg ullary myolipoma. J Neurosurg 1987;66:457–459.
Gynecol Obstet 1941;73:525–538. 180. Dubowitz V, Lorber J, Zachary RB. Lipoma of the cauda equina. Arch Dis
154. Guidetti B, Gagliardo FM. Epidermoid and dermopid cysts: clinical evalu- Child 1965;40:207–213.
ation and late surgical results. J Neurosurg 1977;47:12–18. 181. Schroeder S, Lackner K, Weiand G. Lumbosacral intradural lipoma.
155. Martinez-Lage JF, Esteban JA, Poza M, Casas C. Congenital dermal J Comput Assist Tomogr 1981;5:274.
sinus associated with an abscessed intramedullary epidermoid cyst in a 182. Jones PH, Love JG. Tight filum terminale. Arch Surg 1956;73:556–566.
child: case report and review of the literature. Childs Nerv Syst 1995;11: 183. McLone DG, Mutleur S, Naidich TP. Lipomeningoceles of the conus med-
301–305. ullaris. In: American Society for Pediatric Neurosurgeons, ed. Concepts in
156. Avellino A, Wang P, Miller N, Herkovits E. FLAIR magnetic resonance image Pediatric Neurosurgery, vol. 3. Basel: Karger, 1983:170–177.
of a pediatric spinal epidermoid cyst. Pediatr Neurosurg 2002;36:220–222. 184. Villarejo FJ, Blazquez MG, Gutierrez-Diaz JA. Intraspinal lipomas in chil-
157. Chen S, Ikawa F, Kurisu K, Arita K, Takaba J, Kanou Y. Quantitative MR dren. Childs Brain 1976;2:361–370.
evaluation of intracranial epidermoid tumors by fast fluid-attenuated 185. Lemire RJ, Graham CB, Beckwith JB. Skin-covered sacrococcygeal masses
inversion recovery imaging and echo-planar diffusion-weighted imaging. in infants and children. J Pediatr 1971;79:948–954.
Am J Neuroradiol 2001;22:1089–1096. 186. La Marca F, Grant JA, Tomita T, McLone DG. Spinal lipomas in children:
158. Tsuruda J, Chew W, Moseley M, Norman D. Diffusion-weighted MR outcome of 270 procedures. Pediatr Neurosurg 1997;26:8–16.
imaging of the brain: value of differentiating between extraaxial cysts and 187. Bruce DA, Schut L. Spinal lipomas in infancy and childhood. Childs Brain
epidermoid tumors. Am J Neuroradiol 1990;11:925–931. 1979;5:192–203.
159. Dev R, Husain M, Gupta A, Gupta RK. MR of multiple intraspinal 188. Takeyama J, Hayashi T, Saito M, et al. Spinal hamartoma associated with
abscesses associated with congenital dermal sinus. Am J Neuroradiol spinal dysraphism. Childs Nerv Syst 2006;22:1098–1102
1997;18:742–743. 189. Gold LH, Kieffer SA, Peterson HO. Lipomatous invasion of the spinal cord
160. Pang D, Dias MS. Cervical myelomeningoceles. Neurosurgery 1993;33: associated with spinal dysraphism: myelographic evaluation. Am J Roent-
363–373. genol 1969;107:479–485.
161. Rossi A, Cama A, Consales A, et al. Spectrum of nonterminal myelocysto- 190. Perlitz Y, Izhaki I, Ben-Ami M. Sonographic evaluation of the fetal conus
celes. Neurosurgery 2006;58:509–515. medullaris at 20 to 24 weeks’ gestation. Prenat Diagn 2010;30:862-864.

Barkovich_Chap09.indd 919 5/6/2011 10:29:20 PM


920 Pediatric Neuroimaging

191. Beek FJA, de Vries LS, Gerards LJ, Mali WPTM. Sonographic determination 217. Catala M, Teillet M, De Robertis EM, Le Douarin NM. A spinal cord fate
of the position of the conus medullaris in premature and term infants. map in the avian embryo: while regressing, Hensen’s node lays down the
Neuroradiology 1996;38:S174–S177. notochord and floor plate thus joining the spinal cord lateral walls. Devel-
192. Zalel Y, Lehavi O, Aizenstein O, Achiron R. Development of the fetal spinal opment 1996;122:1599–1610.
cord: time of ascendance of the normal conus medullaris as detected by 218. Pang D, Hoffman HJ. Sacral agenesis with progressive neurological deficit.
sonography. J Ultrasound Med 2006;25:1397–1401. Neurosurgery 1980;7:118–126.
193. Barson AJ. The vertebral level of termination of the spinal cord during 219. Heilig CW, Saunders T, Brosius FC, III, et al. Glucose transporter-1-
normal and abnormal development. J Anat 1970;106:489–497. deficient mice exhibit impaired development and deformities that are
194. James CCM, Lassman LP. Spinal Dysraphism: Spina Bifida Occulta. New similar to diabetic embryopathy. Proc Natl Acad Sci USA 2003;100:
York: Appleton-Century-Crofts, 1972. 15613–15618.
195. Sevinc O, Barut C, Eryoruk N, Kiran S, Arifoglu Y. MRI determination of 220. Passarge E. Congenital malformations and maternal diabetes. Lancet
conus medullaris level in and adult population in Turkey. Neuroradiol J 1965;1:324–325.
2006;19:375–378. 221. Heij HA, Nievelstein RAJ, de Zwart I, Berbeeten BWJM, Valk J, Vos A.
196. Coleman L, Zimmerman R, Rorke L. Ventriculus terminalis of the Abnormal anatomy of the lumbosacral region imaged by magnetic
conus medullaris: MR findings in children. Am J Neuroradiol 1995;16: resonance in children with anorectal malformations. Arch Dis Child
1421–1426. 1996;74:441–444.
197. Kriss V, Kriss T, Babcock D. The ventriculus terminalis of the spinal 222. Rusnak SL, Driscoll SG. Congenital spinal anomalies in infants of diabetic
cord in the neonate: a normal variant on sonography. Am J Roentgenol mothers. Pediatrics 1965;35:989–995.
1995;165:1491–1493. 223. Muthukumar N. Surgical treatment of nonprogressive neurological defi-
198. Kriss V, Kriss T, Coleman R. Sonographic appearance of the ventriculus cits in children with sacral agenesis. Neurosurgery 1996;38:1133–1138.
terminalis cyst in the neonatal spinal cord. J Ultrasound Med 2000;19: 224. McLone DG, Naidich TP. Terminal myelocystocele. Neurosurgery
207–209. 1985;16:36–43.
199. Matsubayashi R, Uchino A, Kato A, Kudo S, Sakai S, Murata S. Cys- 225. Choi S, McComb JG. Long term outocme of terminal myelocystocele
tic dilatation of ventriculus terminalis in adults: MRI. Neuroradiology patients. Pediatr Neurosurg 2000;32:86–91.
1998;40:45–47. 226. Vade A, Kennard D. Lipomyelomeningocystocele. Am J Neuroradiol
200. Unsinn K, Geley T, Freund M, Gassner I. US of the spinal cord in new- 1987;8:375–377.
borns: spectrum of normal findings, variants, congenital anomalies, and 227. Naidich TP, McLone DG. Congenital pathology of the spine and spinal
acquired diseases. Radiographics 2000;20:923–938. cord. In: Taveras JM, Ferrucci JT, eds. Radiology, vol 3. Philadelphia:
201. Dullerud R, Server A, Berg-Johnsen J. MR imaging of ventriculus termina- Lippincott, 1986:1–23.
lis of the conus medullaris. A report of two operated patients and a review 228. Sherman RM, Caylor HD, Long L. Anterior sacral meningocele. Am J Surg
of the literature. Acta Radiol 2003;44:444–446. 1950;79:743–747.
202. Celli P, D’Andrea G, Trillo G, Roperto R, Acqui M, Ferrante L. Cyst of the 229. Dyck P, Wilson CB. Anterior sacral meningocele: case report. J Neurosurg
medullary conus: malformative persistence of terminal ventricle or com- 1980;53:548–552.
pressive dilation. Neurosurg Rev 2002;25:103–106. 230. Schey WL, Shkolnik A, White H. Clinical and radiographic considerations
203. Irani N, Goud AR, Lowe LH. Isolated filar cyst on lumbar spine sonogra- of scarococcygeal teratomas. An analysis of 26 new cases and review of the
phy in infants: a case-control study. Pediatr Radiol 2006;36:1283–1288. literature. Radiology 1977;125:189–195.
204. Yamada S, Won DJ, Pezeshkpour G, et al. Pathophysiology of tethered cord 231. Noseworthy J, Luck EE, Kozakewich HPW. Sacrococcygeal germ cell
syndrome and similar complex disorders. Neurosurg Focus 2007;23:1–10. tumor in childhood: an updated experience with 113 patients. J Pediatr
205. McLendon RE, Oakes WJ, Heinz ER, Yeates AE, Burger PC. Adipose tissue Surg 1981;16:358–364.
in the filum terminale: a CT finding that may indicate tethering of the 232. Altman RP, Randolph JG, Lilly JR. Sacrococcygeal teratoma. J Pediatr Surg
spinal cord. Neurosurgery 1988;22:873–876. 1973;9:389–398.
206. Love JG, Daly DD, Harris LE. Tight filum terminale: report of condition in 233. Beardmore HE, Wigglesworth FW. Vertebral anomalies and alimentary
three siblings. J Am Med Assoc 1961;176:31–33. duplications: clinical and embryological aspects. Pediatr Clin North Am
207. Hendrick EB, Hoffman HJ, Humphreys RP. The tethered spinal cord. Clin 1958;5:457–473.
Neurosurg 1983;30:457–463. 234. Pang D, Dias MS, Ahab-Barmada M. Split cord malformation: part I: a
208. Tani S, Yamada S, Knighton RS. Extensibility of the lumbar and sacral unified theory of embryogenesis for double spinal cord malformations.
cord. Pathophysiology of the tethered spinal cord in cats. J Neurosurg Neurosurgery 1992;31:451–480.
1987;66:116–123. 235. Burrows FGO, Sutcliffe J. The split notochord syndrome. Brit J Radiol
209. Ng WH, Seow WT. Tethered cord syndrome preceding syrinx formation - 1968;41:844–847.
serial radiologica documentation. Childs Nerv Syst 2001;17:494–496. 236. Bentley JFR, Smith JR. Developmental posterior enteric remnants and
210. Hendrick EB, Hoffman HJ, Humphreys RP. Tethered cord syndrome. spinal malformations: the split notochord syndrome. Arch Dis Child
In: McLaurin RL, ed. Myelomeningocele. New York: Grune & Stratton, 1960;35:76–86.
1977:369–376. 237. Hoffman CH, Dietrich RB, Pais MJ, Demos DS, Pribram HFW. The
211. Steinbok P, Kariyattil R, MacNeily AE. Comparison of section of filum split notochord syndrome with dorsal enteric fistula. Am J Neuroradiol
terminale and non-neurosurgical management for urinary incontinence 1993;14:622–627.
in patients with normal conus position and possible occult tethered cord 238. Razack N, Page LK. Split notochord syndrome: case report. Neurosurgery
syndrome. Neurosurgery 2007;61:550–555; discussion 555–556. 1995;37:1006–1008.
212. Steinbok P, MacNeily AE. Section of the terminal filum for occult tethered 239. Kim C, Wang K, Choe G, et al. Neurenteric cyst: its various presentations.
cord syndrome: toward a scientific answer. Neurosurg Focus 2007;23:1–4. Childs Nerv Syst 1999;15:333–341.
213. Duhamel B. From the mermaid to anal imporforation: the syndrome of 240. D’Almeida AC, Steward DJ, Jr. Neurenteric cyst: case report and literature
caudal regression. Arch Dis Child 1961;36:152–155. review. Neurosurgery 1981;8:596–599.
214. Kallen B, Winberg J. Caudal mesoderm pattern of anomalies: from renal 241. Rizk T, Lahoud G, Maarrawi J, et al. Acute paraplegia revealing an intraspi-
agenesis to sirenomelia. Teratology 1974;9:99–112. nal neurenteric cyst in a child. Childs Nerv Syst 2001;17:754–757.
215. Anderson-Shotwell S, Wilson WG. 18p- syndrome with partial sacral 242. Bejjani GK, Wright CD, Schessel D, Sekhar LN. Endodermal cysts of the
agenesis. J Med Genet 1989;26:70–71. posterior fossa. J Neurosurg 1998;89:326–335.
216. Makinski M, Jaworek M, Skowronek-Bala B. Caudal regression syndrome 243. Harris CP, Dias MS, Brockmeyer DL, Townsend JJ, Willis BK, Apfelbaum
associated with white matter lesions and chromosome 18p11.2 deletion. RI. Neurenteric cysts of the posterior fossa: recognition, management, and
Brain Dev 2007;29:164–166. embryogenesis. Neurosurgery 1991;29:893–897.

Barkovich_Chap09.indd 920 5/6/2011 10:29:20 PM


Chapter 9 • Congenital Anomalies of the Spine 921

244. LeDoux MS, Faye-Petersen OM, Aronin PA, Vaid YN, Pitts RM. 272. López BC, Dávid KM, Crockard HA. Inadequate PAX-1 gene expression as
Lumbosacral neurenteric cyst in an infant. J Neurosurg 1993;78: a cause of agenesis of the thoracolumbar spine with failure of segmenta-
821–825. tion. J Neurosurg 1997;86:1018–1021.
245. French BN. Midline fusion defects of formation. In: Youman JR, ed. Neu- 273. Scott RM, Wolpert SM, Bartoshesky LE, Zimbler S, Karlin L. Segmental
rological Surgery. Philadelphia: Saunders, 1990:1201–1204. spinal dysgenesis. Neurosurgery 1988;22:739–743.
246. Menezes AH, Ryken TC. Craniocervical intradural neurenteric cysts. Pedi- 274. Mamelak AN, Cogen PH, Barkovich AJ. The “filum intermedium” sign:
atr Neurosurg 1995;22:88–95. focal in utero spinal cord infarct and extraspinal thecal sac. J Neurosurg
247. Shin J, Byun B, Kim D, Choi D. Neurenteric cyst in the cerebellopontine 1994;81:941–946.
angle with xanthogranulomatous changes: serial MR findings with patho- 275. Faciszewski T, Winter RB, Lonstein JE, Sane S, Erickson D. Segmental spi-
logic correlation. Am J Neuroradiol 2002;23:663–665. nal dysgenesis. J Bone Joint Surg [Am] 1995;77A:530–537.
248. Paolini S, Ciappetta P, Domenicucci M, Guiducci A. Intramedullary 276. Tortori-Donati P, Fondelli MP, Rossi A, Raybaud CA, Cama A, Capra V.
neurenteric cyst with a false mural nodule: case report. Neurosurgery Segmental spinal dysgenesis: neuroradiologic findings with clinical and
2003;52:243–245. embryologic correlation. Am J Neuroradiol 1999;20:445–456.
249. Han JS, Benson JE, Kaufman B, et al. Demonstration of diastematomy- 277. Erkulvrawatr S, El Gammal T, Hawkins J, Green JB, Srinivasan G.
elia and associated abnormalities with MR imaging. Am J Neuroradiol Intrathoracic meningoceles and neurofibromatosis. Arch Neurol 1979;36:
1985;6:215–219. 557–559.
250. James CCM, Lassman LP. Diastematomyelia: a critical survey of 24 cases 278. Bunner R. Lateral intrathoracic meningocele. Acta Radiol 1959;51:1–9.
submitted to laminectomy. Arch Dis Child 1964;39:125–130. 279. Reis C, Carneiro E, Fonseca J, et al. Epitheloid hemangioendothelioma and
251. Guthkelch AN. Diastematomyelia with median septum. Brain 1974;97: multiple thoracolumbar lateral meningoceles: two rare pathological enti-
729–742. ties in a patient with NF1. Neuroradiology 2005;47:165–169.
252. Cohen J, Sledge CB. Diastematomyelia: an embryological interpretation 280. Mahboubi S, Schut L. Decrease in size of intrathoracic meningocele after
with report of a case. Am J Dis Child 1960;100:257–263. insertion of a ventriculovenous shunt. Pediatr Radiol 1977;5:178–180.
253. Rilleit B, Schowing J, Berney J. Pathogenesis of diastematomyelia: can a 281. Rosenbaum TJ, Loule EH, Onofrio BM. Teratomatous cyst of the spinal
surgical model in the chick embryo give some clues about the human mal- canal. J Neurosurg 1978;49:292–297.
formation? Childs Nerv Syst 1992;8:310–316. 282. Lemire RJ, Loeser JD, Leech RW, Alvord EC. Normal and Abnormal Devel-
254. Keim HA, Greene AF. Diastematomyelia and scoliosis. J Bone Joint Surg opment of the Human Nervous System. Hagarstown: Harper and Row,
[Am] 1973;55A:1425–1435. 1975.
255. Schijman E. Split spinal cord malformations: report of 22 cases and review 283. Pickens JM, Wilson J, Myers GG, Frunnet MD. Teratoma of the spinal cord:
of the literature. Childs Nerv Syst 2003;19:96–103. case report and review of the literature. Arch Pathol 1975;99:446–448.
256. Andar UB, Haarkness WFJ, Hayward RD. Split cord malformations of the 284. Seol H, Wang K, Kim S, et al. Intramedullary immature teratoma in a
lumbar region. Pediatr Neurosurg 1997;26:17–24. young infant involving a long segment of the spinal cord. Childs Nerv Syst
257. Ersahin Y, Mutluer S, Kocaman S, Demirtas E. Split spinal cord malforma- 2001;17:758–761.
tions in children. J Neurosurg 1998;88:57–65. 285. Van Gilder JC, Schwartz HG. Growth of dermoids from skin implants to
258. Sinha S, Argarwal D, Mahapatra AK. Split cord malformations: an experi- the nervous system and surrounding spaces of the newborn rat. J Neuro-
ence of 203 cases. Childs Nerv Syst 2006;22:3-7. surg 1967;26:14–20.
259. Gower DJ, Curling OD, Kelly DL, Jr, Alexander E, Jr. Diastematomyelia - a 286. Kukreja K, Manzano G, Ragheb J, Medina LS. Differentiation between
40 year experience. Pediatr Neurosci 1988;14:90–96. pediatric spinal arachnoid and epidermoid-dermoid cysts: is diffusion-
260. Mathieu JP, Dube J. Diastematomyelia and diplomyelia. In: Myriantho- weighted MRI useful? Pediatr Radiol 2007;37:556–560.
poulos NC, ed. Malformations, vol 6(50). Amsterdam: Elsevier, 1987: 287. Tibbs PA, James HE, Rorke LB, Schut L, Bruce DA. Midline hamartomas
435–442. masquerading as meningomyeloceles or teratomas in the newborn infant.
261. Wolf AL, Tubman DE, Seljeskog EL. Diastematomyelia of the cervical spi- J Pediatr 1976;89:928–933.
nal cord with tehtering in an adult. Neurosurgery 1987;21:94–97. 288. Morris G, Murphy K, Rorke L, James H. Spinal hamartomas: a distinct
262. Beyerl BD, Ojemann RG, Davis KR, Hedley-Whyte ET, Mayberg MR. Cervical clinical entity. J Neurosurg 1998;88:954–957.
diastematomyelia presenting in adulthood. J Neurosurg 1985;62:449–453. 289. Castillo M, Smith M, Armao D. Midline spinal cord hamartomas:
263. Naidich TP, Harwood-Nash DC. Diastematomyelia: hemicord and men- MR imaging features of two patients. Am J Neuroradiol 1999;20:
ingeal sheaths; single and double arachnoid and dural tubes. Am J Neuro- 1169–1171.
radiol 1983;4:633–636. 290. Peerless SJ, Durward QU. Management of syringomyelia: a pathophysi-
264. Schlesinger AE, Naidich TP, Quencer RM. Concurrent hydromyelia and ologic approach. Clin Neurosurg 1983;30:531–576.
diastematomyelia. Am J Neuroradiol 1986;7:473–477. 291. Larroche J-C. Malformations of the nervous system. In: Adams JH,
265. Kaffenberger DA, Heinz ER, Oakes JW, Boyko O. Meningocele man- Corsellis J, Duchin EW, eds. Greenfield’s Neuropathology. New York: Wiley
qué: radiologic findings with clinical correlation. Am J Neuroradiol & Sons, 1984:396–402.
1992;13:1083–1088. 292. Harwood-Nash DC, Fitz CR. Myelography and syringohydromyelia in
266. Lassman LP, James CCM. Meningocele manqué. Childs Brain 1977;3:1–11. infancy and childhood. Radiology 1974;113:661–669.
267. Royal S, Tubbs R, D’Antonio M, Rauzzino M, Oakes W. Investigations 293. Petit-Lacour MC, Lasjaunias P, Iffenecker C, et al. Visibility of the central
into the association between cervicomedullary neuroschisis and mirror canal on MRI. Neuroradiology 2000;42:756–761.
movements in patients with Klippel-Feil syndrome. Am J Neuroradiol 294. Hanieh A, Sutherland A, Foster B, Cundy P. Syringomyelia in children with
2002;23:724–729. primary scoliosis. Childs Nerv Syst 2000;16:200–202.
268. Klippel M, Feil A. Un cas d’absence des vetebres cervicales avec cage 295. Ramirez N, Johnston CE, Browne RH. The prevalence of back pain in chil-
thoracic remontant jusqu ‘a’ base du craine. N Iconog de la Salpetriére dren who have idiopathic scoliosis. J Bone Joint Surg 1997;79:364–368.
1912;25:223–250. 296. Steinbok P. Clinical features of Chiari I malformations. Childs Nerv Syst
269. Cochrane D, Haslam R, Myles S. Cervical neuroschisis and meningocoele 2004;20:329–331.
manque in type I (no neck) Klippel-Feil syndrome. Pediatr Neurosurg 297. Nishizawa S, Yokoyama T, Yokota N, Tokuyama T, Ohta S. Incidentally
1990–1991;16:174–178. identified syringomyelia associated with Chiari I malformations: is early
270. Avery L, Rentfro C. Klippel-Feil syndrome. Arch Neurol Psychiatry interventional surgery necessary? Neurosurgery 2001;49:637–640.
1936;36:1068–1076. 298. Rossier AB, Foo D, Shillito J, Dyro FM. Posttraumatic cervical syringomy-
271. Chandra PS, Gupta A, Mishra NK, Mehta VS. Association of craniover- elia. Brain 1985;108:439–461.
tebral and upper cervical anomalies with dermoid and epidermoid cysts: 299. Adams RD, Victor M. Principles of Neurology. New York: McGraw-Hill,
report of four cases. Neurosurgery 2005;56:E1155. 1981.

Barkovich_Chap09.indd 921 5/6/2011 10:29:20 PM


922 Pediatric Neuroimaging

300. Schlesinger EB, Antunes JL, Michelsen WJ, Louis KM. Hydromyelia: 322. Williams B. The distending force in the production of “communication
clinical presentation and comparison of modalities of treatment. Neuro- syringomyelia”. Lancet 1969;2:189–193.
surgery 1981;9:356–365. 323. DuBoulay GH. Pulsatile movements of the CSF pathways. Br J Radiol
301. Logue V, Edwards MR. Syringomyelia and its surgical treatment–an analy- 1966;19:255–262.
sis of 75 patients. J Neurol Neurosurg Psychiatry 1981;44:273–284. 324. Ball MJ, Dayan AD. Pathogenesis of syringomyelia. Lancet 1972;2:799–801.
302. Milhorat TH, Kotzen RM, Mu HTM, Capocelli AL, Milhorat RH. Dys- 325. Josephson A, Greitz D, Klason T, Olson L, Spenger C. A spinal thecal sac
esthetic pain in patients with syringomyelia. Neurosurgery 1996;38: constriction model supports the theory that induced pressure gradients in
940–947. the cord cause edema and cyst formation. Neurosurgery 2001;48:636–645.
303. Tubbs RS, Wellons JC, III, Blound JP, Grabb PA, Oakes WJ. Inclination of 326. Aboulker J. La syringomyelie et les liquides intrarachidiens. Neurochirur-
the odontoid process in the pediatric Chiari I malformation. J Neurosurg gie 1979;24(Suppl 2):1–44.
2003;98 (Suppl 1):43–49. 327. Heiss J, Patronas N, DeVroom H, et al. Elucidating the pathophysiology of
304. Greenlee J, Donovan K, Hasan D, Menezes A. Chiari I malformation in syringomyelia. J Neurosurg 1999;91:553–562.
the very young child: the spectrum of presentations and experience in 31 328. Sherman JL, Citrin CM, Barkovich AJ. MR imaging of syringobulbia.
children under age 6 years. Pediatrics 2002;110:1212–1219. J Comput Assist Tomogr 1987;11:407–411.
305. Wu Y, Chin C, Chan K, Barkovich A, Ferriero D. Pediatric Chiari I mal- 329. Valentini MC, Forni C, Bracchi M. Syringobulbia extending to the basal
formations: do clinical and radiologic features correlate? Neurology ganglia. Am J Neurorad 1988;9:205–207.
1999;53:1271–1276. 330. Fischbein N, Dillon W, Cobbs C, Weinstein P. the “presyrinx” state: a
306. Nishimura G, Aoki K, Haga N, Hasegawa T. Syringohydromyelia in Hajdu- reversible myelopathic condition that may precede syringomyelia. Am
Cheney syndrome. Pediatr Radiol 1996;26:59–61. J Neuroradiol 1999;20:7–20.
307. Boltshauser E, Schmitt B, Wichmann W, Valavanis A, Sailer H, Yonekawa 331. Koyanagi I, Houkin K. Pathogenesis of syringomyelia associated with
Y. Cerebellomedullary compression in recessive craniometaphyseal dys- Chiari type 1 malformation: review of evidences and proposal of a new
plasia. Neuroradiology 1996;38:S193–S195. hypothesis. Neurosurg Rev 2010;33:271–285.
308. Saez RJ, Onofrio BM, Yanigihara T. Experience with the Arnold Chiari 332. Sherman JL, Citrin CM. Magnetic resonance of normal CSF flow. Am
malformation 1960–1970. J Neurosurg 1976;45:416–422. J Neuroradiol 1986;7:3–6.
309. Jack CR, Kokmen E, Onofrio BM. Spontaneous decompression of syrin- 333. Gottschalk A, Schmitz B, Mauer UM, et al. Dynamic visualization of
gomyelia: magnetic resonance imaging findings. Case report. J Neurosurg arachnoid adhesions in a patient with idiopathic syringomyelia using
1991;74:283–286. high-resolution cine magnetic resonance imaging at 3T. J Magn Reson
310. Kastrup A, Nägele T, Topka H. Spontaneous resolution of idiopathic Imaging 2010;32:218–222.
syringomyelia. Neurology 2001;57:1519–1520. 334. Enzmann DR, Pelc NJ. Normal flow patterns of intracranial and spinal
311. Sun JC, Steinbok P, Cochrane DD. Spontaneous resolution and recurrence cerebrospinal fluid defined with phase-contrast cine MR imaging. Radiol-
of a Chiari I malformation and associated syringomyelia. Case report. ogy 1991;178:467–474.
J Neurosurg 2000;92(Suppl 2):207–210. 335. Levy LM, Di Chiro G. MR phase imaging and cerebrospinal fluid flow in
312. Milhorat TH, Capocelli AL, Jr, Kotzen RM, Bolognese P, Heger IM, the head and spine. Neuroradiology 1990;32:399–406.
Cottrell JE. Intramedullary pressure in syringomyelia: clinical and 336. Quencer RM, Post MJD, Hinks RS. Cine MR in the evaluation of normal
pathophysiological correlates of syrinx distension. Neurosurgery and abnormal CSF flow: intracranial and intraspinal studies. Neuroradiol-
1997;41:1102–1110. ogy 1990;32:371–391.
313. Milhorat TH, capocelli ALJ, Anzil AP, Kotzen RM, Milhorat RH. Patho- 337. Hentschel S, Mardal KA, Lovgren AE, Linge S, Haughton V. Characteriza-
logical basis of spinal cord cavitation in syringomyelia: analysis of 105 tion of cyclic CSF flow in the foramen magnum and upper cervical spi-
autopsy cases. J Neurosurg 1995;82:802–812. nal canal with MR flow imaging and computational fluid dynamics. Am J
314. Milhorat TH, Johnson RW, Milhorat RH, Capocelli ALJ, Pevsner PH. Neuroradiol 2010;31:997–1002.
Clinicopathological correlations in syringomyelia using axial magnetic 338. Ventureyra E, Aziz H, Vassilyadi M. The role of cine flow MRI in children
resonance imaging. Neurosurgery 1995;37:206–213. with Chiari I malformation. Childs Nerv Syst 2003;19:109–113.
315. Barnett MJM. The epilogue. In: Barnett MJM, Foster JB, Hudgson P, eds. 339. Tachibana S, Iida H, Yada K. Significance of positive Queckenstedt test
Syringomyelia London: Saunders, 1973:302–313. in patients with syringomyelia associated with Arnold-Chiari malforma-
316. Hecht JT, Butler IJ. Neurologic morbidity associated with achondoplasia. tions. J Neurosurg 1992;76:67–71.
J Child Neurol 1990;5:84–97. 340. Klekamp J, Batzdorf U, Samii M, Bothe HW. Treatment of syringomyelia
317. Kao SCS, Waziri MH, Smith WL, Sato Y, Yuh WTC, Franken EA, Jr. MR associated with arachnoid scarring caused by arachnoiditis or trauma.
imaging of the craniovertebral junction, cranium, and brain in children J Neurosurg 1997;86:233–240.
with achondoplasia. Am J Roentgenol 1989;153:565–569. 341. Vaquero J, Martinez R, Arias A. Syringomyelia-Chiari complex: magnetic
318. Cinalli G, Vinikoff L, Zerah M, Renier D, Pierre-Kahn A. Dandy-Walker resonance imaging and clinical evaluation of surgical treatment. J Neuro-
malformation associated with syringomyelia. J Neurosurg 1997;86:571. surg 1990;73:64–68.
319. Gardner WJ. Hydrodynamic mechanism of syringomyelia: its relationship 342. Barkovich AJ, Sherman JL, Citrin CM, Wippold FJ, II. MR of postoperative
to myelocele. J Neurol Neurosurg Psychiatry 1965;28:247–259. syringomyelia. Am J Neuroradiol 1987;8:319–327.
320. Gardner W, Angel J. The mechanism of syringomyelia and its surgical cor- 343. Elster AD, Chen MYM. Chiari I malformations: clinical and radiologic
rection. Clin Neurosurg 1975;6:131–140. reappraisal. Radiology 1992;183:347–353.
321. Williams B. On the pathogenesis of syringomyelia: a review. J Royal Soc 344. Barkovich AJ, Wippold FJ, Sherman JL, Citrin CM. Significance of cerebel-
Med 1980;73:798–806. lar tonsillar ectopia on MR. Am J Neuroradiol 1986;7:795–799.

Barkovich_Chap09.indd 922 5/6/2011 10:29:20 PM


CHAPTER

Neoplasms of the Spine


10 A. JAMES BARKOVICH

Spinal tumors—introduction

You might also like