You are on page 1of 301

Clinical Brain Mapping

Notice

Medicine is an ever-changing science. As new research and clinical experience


broaden our knowledge, changes in treatment and drug therapy are required.
The authors and the publisher of this work have checked with sources believed
to be reliable in their efforts to provide information that is complete and gener-
ally in accord with the standards accepted at the time of publication. However,
in view of the possibility of human error or changes in medical sciences, neither
the authors nor the publisher nor any other party who has been involved in the
preparation or publication of this work warrants that the information contained
herein is in every respect accurate or complete, and they disclaim all responsi-
bility for any errors or omissions or for the results obtained from use of the infor-
mation contained in this work. Readers are encouraged to confirm the informa-
tion contained herein with other sources. For example and in particular, readers
are advised to check the product information sheet included in the package of
each drug they plan to administer to be certain that the information contained in
this work is accurate and that changes have not been made in the recommended
dose or in the contraindications for administration. This recommendation is
of particular importance in connection with new or infrequently used drugs.
Clinical Brain Mapping
Daniel Yoshor, MD
Associate Professor
Department of Neurosurgery
Baylor College of Medicine
Chief of Neurosurgery
St. Luke’s Episcopal Hospital
Houston, Texas

Eli M. Mizrahi, MD
Chair, Department of Neurology
Professor of Neurology and Pediatrics
Baylor College of Medicine
Chief of Neurophysiology
St. Luke’s Episcopal Hospital
Houston, Texas

New York Chicago San Francisco Lisbon London Madrid Mexico City
Milan New Delhi San Juan Seoul Singapore Sydney Toronto
Copyright © 2012 by The McGraw-Hill Companies, Inc. All rights reserved. Except as permitted under the United States Copyright Act of 1976, no part of this
publication may be reproduced or distributed in any form or by any means, or stored in a database or retrieval system, without the prior written permission of
the publisher.

ISBN: 978-0-07-180596-4

MHID: 0-07-180596-6

The material in this eBook also appears in the print version of this title: ISBN: 978-0-07-148441-1,
MHID: 0-07-148441-8.

All trademarks are trademarks of their respective owners. Rather than put a trademark symbol after every occurrence of a trademarked name, we use names in
an editorial fashion only, and to the benefit of the trademark owner, with no intention of infringement of the trademark. Where such designations appear in this
book, they have been printed with initial caps.

McGraw-Hill eBooks are available at special quantity discounts to use as premiums and sales promotions, or for use in corporate training programs. To contact
a representative please e-mail us at bulksales@mcgraw-hill.com.

TERMS OF USE

This is a copyrighted work and The McGraw-Hill Companies, Inc. (“McGraw-Hill”) and its licensors reserve all rights in and to the work. Use of this work is
subject to these terms. Except as permitted under the Copyright Act of 1976 and the right to store and retrieve one copy of the work, you may not decompile,
disassemble, reverse engineer, reproduce, modify, create derivative works based upon, transmit, distribute, disseminate, sell, publish or sublicense the work or
any part of it without McGraw-Hill’s prior consent. You may use the work for your own noncommercial and personal use; any other use of the work is strictly
prohibited. Your right to use the work may be terminated if you fail to comply with these terms.

THE WORK IS PROVIDED “AS IS.” McGRAW-HILL AND ITS LICENSORS MAKE NO GUARANTEES OR WARRANTIES AS TO THE ACCURACY,
ADEQUACY OR COMPLETENESS OF OR RESULTS TO BE OBTAINED FROM USING THE WORK, INCLUDING ANY INFORMATION THAT
CAN BE ACCESSED THROUGH THE WORK VIA HYPERLINK OR OTHERWISE, AND EXPRESSLY DISCLAIM ANY WARRANTY, EXPRESS OR
IMPLIED, INCLUDING BUT NOT LIMITED TO IMPLIED WARRANTIES OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE.
McGraw-Hill and its licensors do not warrant or guarantee that the functions contained in the work will meet your requirements or that its operation will be
uninterrupted or error free. Neither McGraw-Hill nor its licensors shall be liable to you or anyone else for any inaccuracy, error or omission, regardless of cause,
in the work or for any damages resulting therefrom. McGraw-Hill has no responsibility for the content of any information accessed through the work. Under no
circumstances shall McGraw-Hill and/or its licensors be liable for any indirect, incidental, special, punitive, consequential or similar damages that result from the
use of or inability to use the work, even if any of them has been advised of the possibility of such damages. This limitation of liability shall apply to any claim
or cause whatsoever whether such claim or cause arises in contract, tort or otherwise.
Dedication

To our parents, Shulamit and Joseph Yoshor, and Julia and Isaac D. Mizrahi, who encouraged and
sustained us; and to our patients who inspire and teach us.

v
This page intentionally left blank
Contents

Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xiii

Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xv

SECTION I: TECHNIQUES

Chapter 1. Surface Anatomy as a Guide to Cerebral Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3


Gareth Adams, Jared Fridley, and Daniel Yoshor

Chapter 2. Structural Imaging for Identification of Functional Brain Regions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .13


Jean C. Tamraz and Youssef G. Comair

Chapter 3. Functional MRI for Cerebral Localization: Principles and Methodology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .31
Michael S. Beauchamp

Chapter 4. Functional MRI: Application to Clinical Practice in Surgical Planning and


Intraoperative Guidance. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
Michael Schulder and Robin Wellington

Chapter 5. Neuropsychological Testing: Understanding Brain–behavior Relationships . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .55


Mario F. Dulay, Corwin Boake, Daniel Yoshor, and Harvey S. Levin

Chapter 6. The Wada Test: Intracarotid Injection of Sodium Amobarbital to Evaluate Language
and Memory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
Brian D. Bell, Bruce P. Hermann, and Paul Rutecki

Chapter 7. Extraoperative Brain Mapping Using Chronically Implanted Subdural Electrodes. . . . . . . . . . . . . . . . . . . . . . . .93
David E. Friedman and James J. Riviello, Jr.

Chapter 8. Brain Mapping in the Operating Room . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .103


Sepehr Sani, Edward F. Chang, and Nicholas M. Barbaro

Chapter 9. Anesthesia for Brain Mapping Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .109


Nicholas P. Carling, Chris D. Glover, Daryn H. Moller, and Ira J. Rampil

Chapter 10. Clinical Applications of Magnetoencephalography in Neurology and Neurosurgery . . . . . . . . . . . . . . . . . . .119


Panagiotis G. Simos, Eduardo M. Castillo, and Andrew C. Papanicolaou

Chapter 11. Optical Spectroscopic Imaging of the Human Brain—Clinical Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . .131
Hongtao Ma, Minah Suh, Mingrui Zhao, Challon Perry, Andrew Geneslaw, and Theodore H. Schwartz

vii
viii CONTENTS

Chapter 12. Electrocorticographic Spectral Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .151


Mackenzie C. Cervenka and Nathan E. Crone

Chapter 13. Pediatric Brain Mapping: Special Considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .167


Robert J. Bollo, Chad Carlson, Orrin Devinsky, and Howard L. Weiner

SECTION II: SYSTEMS

Chapter 14. Mapping of the Sensorimotor Cortex. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .189


Roukoz Chamoun, Krishna Satyan, and Youssef G. Comair

Chapter 15. Mapping of Human Language . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .203


Nitin Tandon

Chapter 16. Mapping of the Human Visual System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .219


Muhammad M. Abd-El-Barr, Mario F. Dulay, Paul Richard, William H. Bosking, and Daniel Yoshor

Chapter 17. Mapping of Hearing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .241


Albert J. Fenoy and Matthew A. Howard

Chapter 18. Mapping of Memory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .269


Jeffrey G. Ojemann and Richard G. Ellenbogen

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 277
Contributors

Muhammad M. Abd-El-Barr, MD, PhD Nicholas P. Carling, MD


Department of Neurosurgery Department of Pediatrics (Anesthesiology)
University of Florida Texas Children’s Hospital
Gainesville, Florida Baylor College of Medicine
Houston, Texas
Gareth Adams, MD, PhD
Department of Neurosurgery Chad Carlson, MD
Baylor College of Medicine Comprehensive Epilepsy Center
Houston, Texas Department of Neurology
New York University School of Medicine
Nicholas M. Barbaro, MD New York, New York
Department of Neurological Surgery
Indiana University School of Medicine Eduardo M. Castillo, PhD
Indianapolis, Indiana Department of Pediatrics, Center for
Clinical Neurosciences
Michael S. Beauchamp, PhD Departments of Neurosurgery and Neurology
Department of Neurobiology & Anatomy University of Texas—Health Science Center at Houston
University of Texas Health Science Center Houston, Texas
Houston, Texas
Mackenzie C. Cervenka, MD
Brian D. Bell, PhD Department of Neurology
Department of Neurology The Johns Hopkins University School of Medicine
University of Wisconsin School of Medicine and Baltimore, Maryland
Public Health
Department of Neurology Roukoz Chamoun, MD
W.S. Middleton Memorial Veterans Hospital Department of Neurosurgery
Madison, Wisconsin Baylor College of Medicine
Houston, Texas
Corwin Boake, PhD
Department of Physical Medicine & Rehabilitation Edward F. Chang, MD, PhD
University of Texas Medical School Department of Neurological Surgery
Houston, Texas University of California
San Francisco, California
Robert J. Bollo, MD
Department of Neurosurgery Youssef G. Comair, MD
Baylor College of Medicine Department of Neurosurgery
Neurosurgery Service American University of Beirut
Texas Children’s Hospital Beirut, Lebanon
Houston, Texas
Nathan E. Crone, MD
William H. Bosking, PhD Department of Neurology
Max Planck Florida Institute The Johns Hopkins University School of Medicine
Jupiter, Florida Baltimore, Maryland

ix
x CONTRIBUTORS

Orrin Devinsky, MD Hongtao Ma, PhD


Comprehensive Epilepsy Center Department of Neurosurgery
Department of Neurology Weill Medical College of Cornell University
New York University School of Medicine New York, New York
New York, New York
Eli M. Mizrahi, MD
Mario F. Dulay, PhD Departments of Neurology and Pediatrics
Department of Neurosurgery Baylor College of Medicine
The Methodist Hospital Neurological Institute Houston, Texas
Houston, Texas
Daryn H. Moller, MD
Richard G. Ellenbogen, MD
Department of Anesthesiology
Department of Neurological Surgery
University at Stony Brook
University of Washington School of Medicine
Stony Brook, New York
Seattle, Washington

Albert J. Fenoy, MD Jeffrey G. Ojemann, MD


Department of Neurosurgery Department of Neurological Surgery
University of Texas Health Science Center University of Washington School of Medicine
Houston, Texas Seattle, Washington

Jared Fridley, MD Andrew C. Papanicolaou, PhD


Department of Neurosurgery Department of Pediatrics, Center for
Baylor College of Medicine Clinical Neurosciences
Houston, Texas Departments of Neurosurgery and Neurology
University of Texas—Health Science Center at Houston
David E. Friedman, MD Houston, Texas
Department of Neurosciences
Winthrop-University Hospital Challon Perry, MD
Mineola, New York Department of Neurosurgery
Weill Medical College of Cornell University
Andrew Geneslaw New York, New York
Department of Neurosurgery
Weill Medical College of Cornell University Ira J. Rampil, MD
New York, New York Department of Anesthesiology
University at Stony Brook
Chris D. Glover, MD
Stony Brook, New York
Department of Pediatrics (Anesthesiology)
Texas Children’s Hospital
Paul Richard, MD
Baylor College of Medicine
Department of Neurological Surgery
Houston, Texas
University of Pittsburgh Medical Center
Bruce P. Hermann, PhD Pittsburgh, Pennsylvania
Department of Neurology
University of Wisconsin School of Medicine and James J. Riviello, Jr., MD
Public Health Division of Pediatric Neurology
Madison, Wisconsin Department of Neurology
NYU Comprehensive Epilepsy Center
Matthew A. Howard, MD New York University
Department of Neurosurgery New York, New York
University of Iowa Hospitals and Clinics
Iowa City, Iowa Paul Rutecki, MD
Department of Neurology
Harvey S. Levin, PhD University of Wisconsin School of Medicine and
Departments of Physical Medicine & Rehabilitation, Public Health
Pediatrics, Neurosurgery, and Neurology Department of Neurology
Baylor College of Medicine W.S. Middleton Memorial Veterans Hospital
Houston, Texas Madison, Wisconsin
CONTRIBUTORS xi

Sepehr Sani, MD Jean C. Tamraz, MD, PhD


Department of Neurosurgery Department of Neuroscience & Neuroradiology
Rush University Medical Center Saint-Joseph University
Chicago, Illinois Beirut, Lebanon

Nitin Tandon, MD
Krishna Satyan, MD Department of Neurosurgery
Department of Neurosurgery University of Texas Medical School
Baylor College of Medicine Houston, Texas
Houston, Texas
Howard L. Weiner, MD
Department of Neurosurgery
Michael Schulder, MD Division of Pediatric Neurosurgery
Department of Neurosurgery Comprehensive Epilepsy Center
North Shore-LIJ Health System Department of Neurology
Manhasset, New York New York University School of Medicine
New York, New York

Theodore H. Schwartz, MD Robin Wellington, PhD


Department of Neurosurgery Department of Psychology
Weill Medical College of Cornell University St. John’s University
New York, New York Flushing, New York

Daniel Yoshor, MD
Panagiotis G. Simos, PhD Department of Neurosurgery
Department of Psychology Baylor College of Medicine
University of Crete Neuroscience Center
Rethymno, Greece St. Luke’s Episcopal Hospital
Houston, Texas

Minah Suh, PhD Mingrui Zhao, MD, PhD


Department of Neurosurgery Department of Neurosurgery
Weill Medical College of Cornell University Weill Medical College of Cornell University
New York, New York New York, New York
This page intentionally left blank
Preface

The localization of cerebral function is a critical task and not yet fully integrated into clinical practice, and
for both neurosurgeons and neurologists. In Clinical some techniques have their greatest utility in clinical re-
Brain Mapping we have addressed these localization ef- search. It is meant as a reference for neurosurgeons, neu-
forts from the perspectives of our different but, at times, rologists, neuroradiologists, neuropsychologists, clinical
overlapping backgrounds and clinical interests. One of neuroscientists and others actively involved in the care
us is a neurosurgeon (Daniel Yoshor) and the other a of those with or who are at risk for neurological impair-
neurologist (Eli M. Mizrahi). We began our discussions ment through intervention.
about clinical brain mapping early in our careers as we We have organized the volume into two sections:
cared for adults and children with medically intractable Techniques and Systems. The Techniques section con-
epilepsy who were being evaluated for epilepsy surgery. sists of chapters considering specific methods of cere-
We have worked together for several years at the Baylor bral location: operative anatomy, structural neuroimag-
Comprehensive Epilepsy Center considering the issues ing, functional MRI, magnetoencephalography, optical
of cerebral localization and weighing relative risks and imaging, neuropsychological testing, Wada testing, spe-
benefits of resective surgery for potential seizure con- cial intraoperative mapping techniques, extraoperative
trol, as well as in resective surgery for brain tumors. In brain mapping with implanted electrodes, electrocortico-
the course of our clinical practice, we realized the need graphic spectral analysis, special brain mapping tech-
for a concise and practical, but comprehensive, guide to niques for pediatric patients and anesthetic techniques
clinical brain mapping. In addition to our efforts with for intraoperative brain mapping. In the Systems section
patients, through our interactions with colleagues and there are discussions of somatomotor and somotosen-
trainees, we realized that such a volume would be of sory function, language, vision, hearing, and memory.
value to them both for reference and training. This was Each is written by experts in their respective fields.
beginning of the current volume. This book is intended to serve two purposes. It has
Although initially considered within the context of been developed as a practical guide to brain mapping
epilepsy surgery, Clinical Brain Mapping addresses a in the clinical setting and it is also designed to present
wide range of clinical concerns. It addresses the tech- the scientific basis of the cortical systems that we wish
niques and functional bases for all clinical situations that to localize and preserve in the care of our patients.
may require cerebral localization for diagnosis and man-
agement. Most of the techniques described are now part Daniel Yoshor, MD
of clinical care, others are just now emerging technology Eli M. Mizrahi, MD

xiii
This page intentionally left blank
Acknowledgments

Brain mapping is typically a collaborative effort in both Maunsell, PhD, and Michael S. Beauchamp, PhD, in de-
the clinical and research settings. The development of veloping a research program that strives to use rigor-
Clinical Brain Mapping has also been collaborative. ous scientific methodology in studying human cortical
We are grateful to those who have been instrumen- function.
tal in its production, including the 47 contributors to Dr. Mizrahi is grateful to his long-time colleagues
this volume who have given generously of their time and collaborators in the Peter Kellaway Section of Neu-
and expertise to write timely, insightful, and instructive rophysiology, Department of Neurology, Baylor College
manuscripts. of Medicine, particularly James D. Frost, Jr., MD, and
We have been fortunate to work in an enriched Richard A. Hrachovy, MD. They continue to provide
environment that fosters expert patient care and excel- valuable insights into the neurophysiological aspects of
lence in research. The clinicians, scientists, trainees and cortical mapping.
students at Baylor College of Medicine form an invig- As with any collaborative effort, there are many
orating intellectual community, which has fostered our people who have contributed directly and indirectly to
interests and growth as researchers and clinicians. Simi- Clinical Brain Mapping. We are most grateful to our
larly, St. Luke’s Episcopal Hospital has encouraged and co-workers, clinical and research colleagues, technolo-
supported our work and has proved to be a unique in- gists, trainees, and administrative staff for their diligence
stitution which promotes outstanding clinical care and and hard work on our behalf. In particular, Lisa Rhodes,
clinical research. R EEG/EP T., CLTM, has provided outstanding techni-
We are also grateful to Robert G. Grossman, MD, cal support for brain mapping studies in our patients
and the late Peter Kellaway, PhD. They initially taught for many years. Kathleen Pierson and May-Lin Basso
and mentored us, and then became professional col- provided critical and expert administrative assistance. Fi-
leagues and personal friends. nally, we express our sincere thanks to the editorial staff
Dr. Yoshor acknowledges the influence of Nicholas at McGraw-Hill Medical Publishing, Anne Sydor, PhD,
M. Barbaro, MD, Mitchel S. Berger, MD, and Raymond Executive Editor, and Regina Y. Brown, Senior Project
L. Sawaya, MD, in developing his interest in applying Development Editor and to Tilak Raj, Project Manager,
brain mapping to clinical practice, and of John H.R. Aptara Corporation, Inc.

xv
This page intentionally left blank
SECTION I

TECHNIQUES

1
This page intentionally left blank
Chapter 1
Surface Anatomy as a Guide to
Cerebral Function
Gareth Adams1 , Jared Fridley 1 , and Daniel Yoshor 1,2
1
Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
2
Neuroscience Center, St. Luke’s Episcopal Hospital, Houston, Texas

䉴 INTRODUCTION AND the localizationist theory, which held that brain func-
HISTORICAL PERSPECTIVE tions are localized to specific areas of the brain. Further
understanding of the localization of human brain func-
Our existing knowledge of a number of consistent re- tion was based on correlating the neurological deficits
lationships between specific anatomic landmarks and in patients with specific cortical lesions defined on post-
local cortical function allows the use of anatomy to pre- mortem examinations.2 For example, by performing au-
dict function with considerable accuracy, even without topsies on patients with aphasia, Paul Broca was able
direct physiological confirmation in an individual sub- to localize the functional areas responsible for the pro-
ject. Defining these landmarks with noninvasive struc- duction of speech to the pars triangularis and pars oper-
tural magnetic resonance imaging (MRI) is routinely cularis of the dominant frontal lobe. Carl Wernicke was
used to infer regional function, as described in Chap- able to localize language comprehension to the poste-
ter 2, “Structural Imaging for Identification of Functional rior, superior temporal gyrus. Correlation of lesions in
Brain Regions.” Direct inspection of anatomic clues, in the occipital lobes from shrapnel and penetrating trauma
particular examination of the exposed cortical anatomy with the visual field defects sustained by soldiers in
of the brain during craniotomy, can provide highly World War II combat provided further localization of
useful clues to functional localization. Because intersub- visual function in humans.3 Similarly, studies of sub-
ject variation in cortical anatomy and functional local- jects with cortical lesions and sensory and motor deficits
ization is not insignificant, and because local pathology demonstrated that motor and sensory functions are local-
such as a brain tumor maybe obscure anatomic clues, ized around the central sulcus.3 Collectively over a pe-
accurate identification of functional regions often re- riod of decades, a crude understanding of the anatomic
quires physiological mapping through other techniques location of functional regions emerged from these
presented in this book. But anatomic landmarks re- studies.
main invaluable, both as a primary method and as Two other methods have greatly further extended
an adjunct to the physiological techniques described our understanding of the functional organization of
in this book, for plotting regional brain function. This human cerebral cortex. Pioneering studies employing
chapter reviews anatomic methods for estimating re- direct cortical electrical stimulation in human neurosur-
gional functional by simple visual inspection. It is bro- gical patients demonstrated consistent relationships be-
ken down into sections detailing anatomic techniques tween cortical anatomy and cortical eloquence among
for surface localization of underlying cortical anatomy, many different subjects. For example, Wilder Penfield
and clues for localization of speech, motor and sensory demonstrated through human cortical mapping during
function, vision and hearing based on cortical surface planning for cortical excisions that motor and sensory
anatomy. function is localized around the central sulcus, and was
Historically, the understanding of the presence of able to map a motor and sensory homunculus to the
localized brain function has been based on experimen- central area.2,4 More recently, the advent of structural
tally created lesions in animals. During a prominent pub- and functional MRI (fMRI) has had an explosive im-
lic lecture and scientific debate in 1881, Sir David Ferrier pact on our understanding of the consistent relation-
convincingly showed that creating a lesion in a monkey’s ship between anatomy and regional function.5,6 Studies
left posterior frontal cortex resulted in a right-sided hemi- that combine both electrical stimulation and fMRI map-
plegia, and that bilateral lesions in the superior tempo- ping in individual subjects has further validated these
ral lobes resulted in deafness.1 This evidence buttressed relationships.7−9

3
4 SECTION I TECHNIQUES

50%
UPPER ROLANDIC POINT

NE
25%

LI
E PAR
LOB
75%

C
I ET

DI
L L I N LO
E

AN
T A EXT. PAR. OCC.
IAN BE

OL
N Y LV FIS
S

R
VIA
YL I N N
FR

LO PITAL
O T

S
P
ORB. PORAL L

BE
LINE
PREAUR
TEMP.

CI
ANG. M
TE

OC
0%
NASION PREAUR
POINT
100%
INION

Figure 1–1. Taylor–Haughton lines.Method for approximating the central sulcus and
sylvian fissure using the Taylor–Haughton lines. (From Taylor and Haughton’s Some
Recent Researches on the Topography and Convolutions of the Brain.)

䉴 APPROXIMATE LOCALIZATION from the nasion to the inion. The central sulcus can be
OF CORTICAL STRUCTURAL approximated by multiple methods. One method to ap-
proximate the central sulcus is to connect a point 2 cm
ANATOMY USING
posterior to the halfway point of the Taylor–Haughton
EXTERNAL CRANIAL line across the top of the cranium with a point 5 cm
LANDMARKS above the external auditory meatus. A second method is
to connect the point on the Taylor–Haughton line across
The location of important cortical anatomic features, the top of the cranium where it is intersected by the
such as the sylvian fissure and central sulcus, can be posterior ear line with the point on the approximated
approximated from the external anatomy of the skull.10 sylvian fissure intersected by the condylar line.11 Other
Taylor–Haughton lines (Fig. 1–1) can be simply con- techniques of localizing the sylvian fissure and central
structed from external landmarks by drawing four lines sulcus based on external cranial landmarks have been
on the cranium. The baseline, or Frankfurt plane, is de- described, and like Taylor–Haughton lines, are also
fined as a line passing the inferior margin of the or- quite accurate.12
bit through the superior margin of the external audi-
tory meatus. A second line is drawn from the nasion
to the inion across the top of the cranium and divided
䉴 ANATOMICAL LOCALIZATION OF
into quarters. Two more lines are drawn perpendicular
to the baseline. The posterior ear line is perpendicular MOTOR AND SENSORY
to the baseline and passes through the mastoid process. FUNCTION IN THE EXPOSED
The condylar line is perpendicular to the baseline and BRAIN
passes through the mandibular condyle.11
The location of the sylvian fissure can be approx- Motor and sensory functions are located in the Rolandic
imated by drawing a line from the lateral canthus to cortex surrounding the central, or Rolandic, fissure.
the three-quarter point along the Taylor–Haughton line Motor function is predominantly located in the anterior
CHAPTER 1 SURFACE ANATOMY AS A GUIDE TO CEREBRAL FUNCTION 5

wall of the central sulcus and in the precentral gyrus, the termination of the posterior ascending ramus of the
whereas sensory function is predominantly located in sylvian fissure is the supramarginal gyrus. The superior
the posterior wall of the central sulcus and in the post- temporal gyrus runs parallel to the sylvian fissure, first
central gyrus. The first step in identifying the Rolandic posteriorly then superiorly. It is capped by the angular
cortex is to identify the central sulcus. The position of gyrus, another horseshoe-shaped gyrus making up the
the central sulcus can be approximated on the surface of posterior portion of the inferior parietal lobule. The char-
the cranium using the techniques detailed in the previ- acteristic roles of these areas in language-related func-
ous section, and with the adjuvant use of image guidance tion are described in a separate section later.
at surgery. However, definitively identifying the central Primary motor and sensory function (Fig. 1–3), as
sulcus at surgery can be difficult even in the absence of demonstrated by Penfield,4 are organized along the pre-
anatomic abnormalities such as tumors or dysplasia, par- central and postcentral gyri in a somatotopic map, which
ticularly since much of the sulcal and gyral anatomy may he represented with a homunculus superimposed onto
be obscured by the draining veins and the pial vessels. the cortex. This homunculus is positioned with its feet
The central sulcus separates the frontal and pari- within the interhemispheric fissure and its head ex-
etal lobes. Broca described the central sulcus containing tending toward the sylvian fissure, and represents a
three curves and a superior and inferior genu. Superiorly somewhat crude, albeit useful, oversimplification of the
the central sulcus extends from the interhemispheric organization of motor cortex.
fissure and often extends onto the mesial aspect of The cortical representation of motor hand function
the hemisphere. Inferiorly it is usually separated from is typically located in the superior portion of the precen-
the sylvian fissure by the subcentral gyrus. It is rarely tral sulcus along the middle genu of the central sulcus.
interrupted.13−15 Localizing the central sulcus is possible The curve of the middle genu of the central sulcus be-
through its relationship to the sylvian fissure and to the comes more pronounced in the depths of the central
other surrounding sulci and gyri of the frontal, temporal, sulcus, forming a knob or omega shape. This knob was
and parietal lobes (Fig. 1–2). identified by Broca as the pli de passage moyen. Stud-
Naidich et al. published a systematic method of ies using fMRI have demonstrated that this area is the
identifying the anatomic relationships of the low-middle cortical functional location of hand motor function, in
convexity in 1995.16 The first step is to identify the syl- the precentral gyrus and on the anterior surface of the
vian fissure, which separates the frontal and tempo- central sulcus, and hand sensory function, in the pos-
ral lobes. It is composed of five rami, with the long terior surface of the central sulcus and the postcentral
obliquely oriented section visible on the cortical surface gyrus.14,17−19 This precentral knob is usually not visible
designated as the posterior horizontal ramus. The ante- initially during surgery as it is obscured by the arachnoid
rior horizontal ramus and anterior ascending ramus ex- and bridging veins and is deep within the central sulcus.
tend superiorly from the anterior section of the posterior The same area can be located intraoperatively by rely-
horizontal ramus forming a characteristic V or Y pattern. ing on other landmarks of the frontal lobe, as it is on the
These rami divide the inferior frontal gyrus from anterior central sulcus opposite the intersection of the superior
to posterior, into the pars orbitalis, pars triangularis, and frontal sulcus with the precentral sulcus.
pars opercularis. The frontal convexity is divided into Tongue sensory function is located within the in-
superior, middle, and inferior gyri by the superior and ferior widening of the postcentral gyrus immediately
inferior frontal sulci. These sulci extend posteriorly and above the sylvian fissure. Face sensory function is lo-
fuse with the precentral sulcus. Anterior and parallel to cated in the narrow portion of the postcentral gyrus su-
the precentral sulcus is the precentral gyrus. The middle perior to the tongue functional region.20
frontal gyrus often run into and fuses with the precentral While these anatomic landmarks do provide some
gyrus, forming a characteristic sideways capital T shape. localization of motor and sensory cortical function, it can
The postcentral gyrus lies posterior to the central be very difficult intraoperatively to identify the associ-
sulcus. It is typically narrower than the precentral sul- ated gyri and sulci, especially with a limited exposure.
cus. At its inferior border, it is bounded posteriorly by These landmarks can provide initial localization allow-
the posterior subcentral sulcus, giving the inferior end of ing the targeting of further studies to verify the location
the postcentral gyrus a characteristic widened appear- of the motor and sensory cortex, by phase reversal of so-
ance. The postcentral sulcus is located parallel to the matosensory evoked potentials waveforms or by direct
central sulcus immediately posterior to the postcentral cortical electrical stimulation.21
gyrus. It can be a single long sulcus or may be divided
into multiple segments. The parietal convexity is sepa-
rated into the superior and inferior parietal lobules by 䉴 LOCALIZATION OF LANGUAGE-
the intraparietal sulcus. The posterior ascending ramus RELATED FUNCTION
of the sylvian fissure hooks superiorly into the inferior
parietal lobule. The horseshoe-shaped gyrus in the ante- Language function is classically separated into two main
rior inferior parietal lobule superior to and surrounding cortical areas. Wernicke’s area is involved in language
6 SECTION I TECHNIQUES

A B

C D

E F

Figure 1–2. Identification of the central sulcus. A stepwise method for identifying the
central sulcus and the surrounding sulci and gyri. In panel A, the sylvian or lateral
fissure is identified with a (1). In panel B, the anterior horizontal ramus (2) and the
anterior ascending ramus (3) are identified in their typical Y shape. These rami define
the pars orbitalis (PObr), pars triangularis (PTr), and pars opercularis (POp). In panel C,
the precentral sulcus (4) is identified. In panel D, the central sulcus (5) and precentral
gyrus (PreC) are identified. In panel E, the postcentral sulcus (6) and the postcentral
gyrus (PostC) are identified. In panel F, the superior temporal sulcus (7), superior
temporal gyrus (STG), and middle temporal gyrus (MTG) are identified. (continued)
CHAPTER 1 SURFACE ANATOMY AS A GUIDE TO CEREBRAL FUNCTION 7

fused across the temporal lobe outside of the classical


Wernicke’s area.24,25 However, it is still useful to be able
to identify the classical locations of these areas to serve
as a starting point for cortical mapping (Fig. 1–4).
Broca’s area is classically described as being located
in the pars triangularis and pars opercularis of the infe-
rior frontal gyrus. The inferior frontal gyrus is bounded
by the sylvian fissure inferiorly and the inferior frontal
sulcus superiorly. The anterior horizontal ramus and an-
terior ascending sylvian ramus extend superiorly from
the sylvian fissure into the inferior frontal gyrus in a Y or
V shape.16 These two rami divide the inferior frontal sul-
cus into three parts, the pars orbitalis, pars triangularis,
G and pars opercularis, from anterior to posterior. The in-
ferior frontal gyrus is bounded posteriorly by the central
sulcus. Quinones-Hinojosa and colleagues used intraop-
Figure 1–2. (Continued) In panel G, the intraparietal
sulcus (8), the supramarginal gyrus (SMG), and the
erative mapping correlated with MRI to locate Broca’s
angular gyrus (AG) are identified. area in relation to the sulci defining the inferior frontal
gyrus.7 They proposed a method for localizing Broca’s
comprehension, including both spoken and read lan- area based on the intersection of lines drawn from de-
guage and is located in the posterior, superior tempo- fined points in the inferior frontal gyrus. The first line is
ral gyrus. Broca’s area is involved in the production drawn from the opercular tip posteriorly at a 45◦ angle
of speech and is located in the inferior frontal gyrus, between the sylvian fissure and the anterior ascending
classically localized to the pars triangularis and pars op- sylvian ramus. The second line is drawn superiorly, per-
ercularis. These two areas are connected by the arcu- pendicular from the sylvian fissure at the level of the pre-
ate fasciculus. In reality, there is significant variation in central sulcus. The third line is drawn anteriorly, parallel
the location of speech function between subjects.22−24 to the sylvian fissure at the level of the inferior tip of the
Surgical resections involving potential speech areas are central sulcus. The intersection of these three lines pro-
usually performed with the patient awake, which allows vides an estimate of the location of Broca’s area. While
cortical mapping of speech function through intraopera- this technique does provide an estimated location for
tive stimulation. Recent studies have shown that speech Broca’s area, it is only an estimate and accurate localiza-
function has a much wider distribution across the frontal tion of speech function is best determined with intraop-
lobe outside of the classical Broca’s area and is more dif- erative or extraoperative cortical stimulation mapping.

A B

Figure 1–3. Identification of motor and sensory cortex. A. In the left panel, primary
motor cortex is located in the precentral gyrus, with hand function (H) localized
perpendicular to the end of the superior frontal gyrus. B. In the right panel, primary
sensory cortex is located in the postcentral gyrus. Tongue sensory (T) is located in
the widened area of the postcentral gyrus close to the sylvian fissure. Face sensory
(F) is located in the narrow strip superior to tongue sensory, and hand sensory (H)
superior to face sensory.
8 SECTION I TECHNIQUES

Figure 1–4. Localization of speech. Broca’s area (B) is classically located in the pars
triangularis and pars opercularis of the inferior frontal gyrus. Wernicke’s area (W) is
located in the posterior portion of the superior temporal gyrus and the supramarginal
gyrus. However, direct stimulation during awake craniotomy has demonstrated
speech function over a much wider area than the classical speech areas, as indicated
by the red outlines compared to the blue outlines of the classical speech areas.

Wernicke’s area is classically located in the poste-


fissure. The fovea is located posteriorly near the occipi-
rior, superior temporal gyrus and in the supramarginal
tal pole. Peripheral vision is located anteriorly. There is
gyrus of the inferior parietal lobule adjacent to the syl-
significant magnification of the retinotopic map near the
vian fissure. These areas can be identified by locating
fovea, with a much larger cortical area corresponding
the sylvian fissure. The superior temporal gyrus runs
to the area around the fovea. Other visual areas extend
between the sylvian fissure and the superior temporal
superiorly and inferiorly from the calcarine fissure, cor-
sulcus, which runs parallel to the sylvian fissure. The
responding to areas V2, V3, and V4.26−29 An area homol-
posterior portions of both the sylvian fissure and the
ogous to the middle temporal (MT) region is located at
superior temporal sulcus hook superiorly and terminate
the junction of the temporal, parietal, and occipital lobes
in the inferior lobule of the parietal lobe. The supra-
in humans (Fig. 1–5). This area is involved in processing
marginal gyrus is the anterior portion of the parietal
of movement.30−32
lobe, which forms a horseshoe shape over the poste-
The calcarine sulcus is located on the mesial sur-
rior end of the sylvian fissure. The angular gyrus forms
face of the occipital lobe. It extends posteriorly from the
a similar horseshoe shape over the posterior end of
splenium of the corpus callosum to the occipital pole.
the superior temporal gyrus.13,16 Intraoperative mapping
It is divided into an anterior and posterior portion by
during awake craniotomy has demonstrated that lan-
the parietal-occipital sulcus. The posterior portion of the
guage function is highly variable between subjects and
calcarine sulcus splits into a Y shape as it approaches
can be widely dispersed over the temporal lobe and pari-
the occipital pole, with the superior portion of the Y
etal lobe outside the classical Wernicke’s area.24
sometimes extending onto the lateral surface of the oc-
cipital lobe. The calcarine sulcus ranges from 2.5 to 3 cm
deep.14,15 The MT region is located near the junction of
䉴 LOCALIZATION OF VISUAL
the temporal, parietal, and occipital lobes.30−32
FUNCTION
Primary visual cortex (V1) is located in the occipital lobe
on the mesial surface both within the calcarine sulcus 䉴 LOCALIZATION OF AUDITORY
and on the surrounding cortex. The visual cortex is or- FUNCTION
ganized in a retinotopic map with the fovea located pos-
teriorly near the occipital pole. The vertical meridian is Penfield and Rasmussen localized hearing function to
located at the calcarine fissure and the horizontal merid- the superior temporal gyrus by direct electrical stim-
ian is deep within the calcarine fissure. Functional MRI ulation of the human cortex. Further studies using
mapping of the visual cortex has demonstrated that the positron emission tomography, fMRI, and direct cortical
V1 is located mostly within the folds of the calcarine recordings have demonstrated that the auditory cortex is
CHAPTER 1 SURFACE ANATOMY AS A GUIDE TO CEREBRAL FUNCTION 9

Figure 1–5. Localization of vision. The primary visual cortex (V1) is located within the
calcarine sulcus, extending slightly out onto the medial surface of the occipital lobe
(left panel). The fovea (F) is represented at the occipital pole. Additional retinotopic
visual areas spread out from the calcarine sulcus and extend onto the lateral surface
of the occipital lobe. Middle temporal (MT), which is involved in motion tracking, is
located near the parieto-occipital-temporal junction.

located bilaterally on the superior temporal gyrus both Heschl’s gyrus is located completely within the syl-
on the exposed cortical surface and in the depths of vian fissure. It is bounded posteriorly by the posterior
the sylvian fissure on the transverse temporal gyrus of transverse supratemporal sulcus, which originates at the
Heschl (Fig. 1–6). The primal auditory field is thought sylvian fissure and extends from the lateral surface of
to be primarily located in the posteromedial portion the temporal lobe with an anterior oblique orientation.
of Heschl’s gyrus. Brugge et al. have demonstrated the Anteriorly, Heschl’s gyrus is bounded by the anterior
presence of three auditory cortical fields, with two on transverse temporal sulcus. The gyrus extends either
Heschl’s gyrus and one on the posterolateral surface of obliquely or perpendicularly to the sylvian fissure.13
the superior temporal gyrus.33−35

䉴 SUMMARY

While there is significant variation in the cortical surface


anatomy of the human brain there are some constants
that can be used to roughly identify functional locations
of motor function, sensory function, speech, and vision
on the cortex. Anatomic and functional imaging prior
to surgery can provide initial localization of function.
Combining these imaging techniques with knowledge
of the relationship between cranial anatomy and the un-
derlying cortex allows the planning of a targeted cran-
iotomy. Once the craniotomy has been performed the
combination of image guidance and the anatomical land-
marks described in the preceding sections can be used
for initial targeting of further localization of function us-
ing techniques such as awake craniotomy with direct
stimulation for speech mapping or phase inversion for
Figure 1–6. Localization of hearing. Primary auditory locating the motor and sensory cortex. Because there
cortex is located in Heschl’s gyrus (H) buried on the
temporal lobe surface buried within the sylvian
is significant variation in the location of cortical func-
fissure. Auditory cortex also extends onto the lateral tion between subjects, anatomical clues should only be
temporal lobe on the posterior superior temporal used as a starting point for other mapping techniques to
gyrus, as indicated by the blue outline. definitively identify the cortical functional locations.
10 SECTION I TECHNIQUES

䉴 PEARLS AND PITFALLS area. Neurosurgery 2002;50(6):1296-1309, discussion 1309-


1310.
t There is significant variation between subjects 3. Fishman RS. Gordon Holmes, the cortical retina, and the
both in the sulcal and gyral anatomy and in the wounds of war. The seventh Charles B. Snyder Lecture.
Doc Ophthalmol 1997;93(1-2):9-28.
location of function on the cortex.
t 4. Penfield W. Epilepsy and the Functional Anatomy of the
During a craniotomy, even locating the central Human Brain, 1st edition. Boston: Little Brown, 1954,
sulcus and the surrounding Rolandic cortex can pp. xv-896.
be difficult due to limited exposure, overlying 5. Van Essen DC, Dierker DL. Surface-based and probabilistic
arachnoid membrane, and surface vessels. atlases of primate cerebral cortex. Neuron 2007;56(2):209-
t The central sulcus is best identified by following a 225.
stepwise pattern of identification of surrounding 6. Gaillard WD. Functional MR imaging of language, mem-
sulci that have relatively little variance between ory, and sensorimotor cortex. Neuroimaging Clin N Am
subjects. The sylvian fissure is the most reliable 2004;14(3):471-485.
starting landmark. 7. Quinones-Hinojosa A, Ojemann SG, Sanai N, et al. Preop-
t Broca’s area is classically located in the pars trian- erative correlation of intraoperative cortical mapping with
magnetic resonance imaging landmarks to predict localiza-
gularis and pars opercularis, which can be iden-
tion of the Broca area. J Neurosurg 2003;99(2):311-318.
tified by finding the classic V or Y shape of 8. Krings T, Schreckenberger M, Rohde V, et al. Func-
the anterior ascending and anterior horizontal ra- tional MRI and 18F FDG-positron emission tomography
mus of the sylvian fissure. However, intraopera- for presurgical planning: comparison with electrical cor-
tive mapping has shown that speech function is tical stimulation. Acta Neurochir (Wien) 2002;144(9):889-
distributed over a much larger region of the 899; discussion 899.
frontal lobe outside the classic Broca’s area. 9. Bartos R, Jech R, Vymazal J, et al. Validity of primary
t Wernicke’s area is classically located in the su- motor area localization with fMRI versus electric cortical
perior temporal gyrus at the posterior end of stimulation: a comparative study. Acta Neurochir (Wien)
the sylvian fissure, sometimes extending into the 2009;151(9):1071-1080.
supramarginal gyrus of the parietal lobe. Lan- 10. Ribas GC, Yasuda A, Ribas EC, et al. Surgical anatomy of mi-
croneurosurgical sulcal key points. Neurosurgery 2006;59
guage mapping has shown that language function
(4 Suppl 2):ONS177-ONS210; discussion ONS210-ONS211.
is distributed over a larger region of the tempo- 11. Greenberg MS, Arredondo N. Handbook of Neurosurgery,
ral lobe, particularly posterior and inferior to the 6th edition. New York: Thieme Medical Publishers, 2006,
classic Wernicke’s area. pp. xii-1013.
t Primary visual cortex is reliably located on the 12. Reis CV, Sankar T, Crusius M, et al. Comparative study
mesial aspect of the occipital lobe mostly within of cranial topographic procedures: Broca’s legacy toward
the folds of the calcarine sulcus with the fovea practical brain surgery. Neurosurgery 2008;62(2):294-310;
represented at the occipital pole. Other areas of discussion 310.
the visual cortex extend out onto the surface of 13. Tamraz JC, Comair YG. Atlas of regional anatomy of the
the occipital lobe. Area MT, involved with tracking brain using MRI. Berlin: Springer, 2000.
of motion, is located on the lateral cortical surface 14. Yousry TA, Schmid UD, Alkadhi H, et al. Localization of
the motor hand area to a knob on the precentral gyrus. A
near the junction of the occipital, parietal, and
new landmark. Brain 1997;120(Pt 1):141-157.
temporal lobes. 15. Rhoton AL Jr. The cerebrum. Neurosurgery 2002;51(4
t Auditory cortex is located bilaterally in Heschl’s Suppl):S1-S51.
gyrus buried within the sylvian fissure and in the 16. Naidich TP, Valavanis AG, Kubik S. Anatomic relation-
posterior portion of the superior temporal gyrus. ships along the low-middle convexity: Part I–Normal spec-
t Anatomical landmarks should only be considered imens and magnetic resonance imaging. Neurosurgery
as a starting point for further localization using 1995;36(3):517-532.
other techniques, particularly when working close 17. Boling W, Olivier A, Bittar RG, Reutens D. Localization of
to eloquent areas involved in motor, sensory, and hand motor activation in Broca’s pli de passage moyen. J
speech function. Neurosurg 1999;91(6):903-910.
18. Boling W, Parsons M, Kraszpulski M, Cantrell C, Puce A.
Whole-hand sensorimotor area: cortical stimulation local-
ization and correlation with functional magnetic resonance
REFERENCES imaging. J Neurosurg 2008;108(3):491-500.
19. Boling WW, Olivier A. Localization of hand sensory func-
1. Tyler KL, Malessa R. The Goltz-Ferrier debates and the tion to the pli de passage moyen of Broca. J Neurosurg
triumph of cerebral localizationalist theory. Neurology 2004;101(2):278-283.
2000;55(7):1015-1024. 20. Boling W, Reutens DC, Olivier A. Functional topography
2. Boling W, Olivier A, Fabinyi G. Historical contributions of the low postcentral area. J Neurosurg 2002;97(2):388-
to the modern understanding of function in the central 395.
CHAPTER 1 SURFACE ANATOMY AS A GUIDE TO CEREBRAL FUNCTION 11

21. Kombos T, Suess O, Funk T, Kern BC, Brock M. Intra- tive fields in human visual cortex mapped with surface
operative mapping of the motor cortex during surgery electrodes. Cereb Cortex 2007;17(10):2293-2302.
in and around the motor cortex. Acta Neurochir (Wien) 29. DeYoe EA, Carman GJ, Bandettini P, et al. Mapping striate
2000;142(3):263-268. and extrastriate visual areas in human cerebral cortex. Proc
22. Lubrano V, Roux FE, Demonet JF. Writing-specific sites Natl Acad Sci U S A 1996;93(6):2382-2386.
in frontal areas: a cortical stimulation study. J Neurosurg 30. Tootell RB, Taylor JB. Anatomical evidence for MT and
2004;101(5):787-798. additional cortical visual areas in humans. Cereb Cortex
23. Simos PG, Papanicolaou AC, Breier JI, et al. Localiza- 1995;5(1):39-55.
tion of language-specific cortex by using magnetic source 31. Tootell RB, Reppas JB, Kwong KK, et al. Functional anal-
imaging and electrical stimulation mapping. J Neurosurg ysis of human MT and related visual cortical areas using
1999;91(5):787-796. magnetic resonance imaging. J Neurosci 1995;15(4):3215-
24. Sanai N, Mirzadeh Z, Berger MS. Functional outcome af- 3230.
ter language mapping for glioma resection. N Engl J Med 32. Watson JD, Myers R, Frackowiak RS, et al. Area V5 of
2008;358(1):18-27. the human brain: evidence from a combined study us-
25. Ojemann SG, Berger MS, Lettich E, Ojemann GA. Localiza- ing positron emission tomography and magnetic resonance
tion of language function in children: results of electrical imaging. Cereb Cortex 1993;3(2):79-94.
stimulation mapping. J Neurosurg 2003;98(3):465-470. 33. Brugge JF, Volkov IO, Oya H, et al. Functional localization
26. Sereno MI, Dale AM, Reppas JB, et al. Borders of of auditory cortical fields of human: click-train stimulation.
multiple visual areas in humans revealed by functional Hear Res 2008;238(1-2):12-24.
magnetic resonance imaging. Science 1995;268(5212):889- 34. Fenoy AJ, Severson MA, Volkov IO, Brugge JF, Howard MA
893. 3rd. Hearing suppression induced by electrical stimulation
27. Tootell RB, Hadjikhani NK, Vanduffel W, et al. Functional of human auditory cortex. Brain Res 2006;1118(1):75-83.
analysis of primary visual cortex (V1) in humans. Proc Natl 35. Howard MA, Volkov IO, Mirsky R, et al. Auditory cortex
Acad Sci U S A 1998;95(3):811-817. on the human posterior superior temporal gyrus. J Comp
28. Yoshor D, Bosking WH, Ghose GM, Maunsell JH. Recep- Neurol 2000;416(1):79-92.
This page intentionally left blank
Chapter 2
Structural Imaging for Identification of
Functional Brain Regions
Jean C. Tamraz1 and Youssef G. Comair 2
1
Department of Neuroscience and Neuroradiology, Saint-Joseph University, Beirut, Lebanon
2
Department of Neurosurgery, American University of Beirut, Beirut, Lebanon

䉴 HISTORICAL LANDMARKS 30th week of gestation (Table 2–1). Secondary (Table


2–2) and tertiary sulci (Table 2–3) occur later in devel-
Leuret and his pupil Gratiolet1 first attempted to classify opment and are responsible for giving the adult brain
brain fissures before Meynert2 expanded on this find- its characteristic highly involuted and gyriform appear-
ing and gave a detailed account of the regional varia- ance. For a better understanding of the sulcal and gyral
tions existing in the cortical mantle and their structural anatomy, we will focus primarily on the primary and
and functional relationships. A large body of literature secondary sulci. Tertiary sulci are subject to marked in-
from the second part of the 19th and the early 20th cen- dividual variations and are, therefore, difficult to iden-
turies reported on sulcal patterns in the human brain. tify on MRI; only those that are relatively constant across
Encephalometry, pioneered by Ariens Kappers in 1847,3 subjects will be annotated.
was one of the most extensively used methods to study
the brain. Major contributions to our understanding of
cortical architecture and surface morphology were made 䉴 THE LATERAL SURFACE OF THE
by von Economo and Koskinas,4 who in 1925 developed CEREBRAL HEMISPHERE
a highly detailed nomenclature for cortical surface pat-
terns, accompanied by a description of the cytoarchitec- Brain sulcation is most efficiently investigated using 3D-
tural peculiarities of each region. MRI surface renderings. However, anatomic correlations
The evolution of brain imaging followed a sim- may also be achieved indirectly using cross-sectional
ilar path. High-field, 3.0-T MR now provides a high- anatomic atlases based on definite referentials6,7,8,9,10,11
resolution sectional atlas of each imaged brain region or by using two-dimensional (2D) MR slices12 typically
and allows us to obtain a comprehensive rendering of encountered in clinical practice, particularly those ex-
an entire brain in three dimensions (3D). Knowledge of tending from the lateral aspect of the hemisphere to
the gyral and sulcal anatomy and its variations remains reach the insular level, or those showing a parasagittal
essential in understanding the relationship between mor- view.
phology and function.

䉴 THE LATERAL SULCUS AND THE


䉴 BRAIN MORPHOLOGY AND SYLVIAN OPERCULA
FISSURAL PATTERNS
The lateral or sylvian fissure is the major landmark on
The classification of cerebral sulci as primary, secondary, the lateral surface of the brain. It develops in the 14th
or tertiary is widely used by neuroanatomists. Primary week of gestation and is the most important and con-
fissures can be described using comparative and ontoge- stant of the cerebral sulci. It may be divided into three
netic approaches. On the basis of comparative anatomy, segments. The first, or hidden stem segment, extends
sulci found in all gyrencephalic primates may be defined from the lateral border of the anterior perforated sub-
as primary. Embryologically, these fissures appear early stance and courses over the limen insula in a posteriorly
in telencephalic development. On reviewing previous concave path before ending at the falciform sulcus, sep-
works and the work of Larroche and Feess-Higgins,5 we arating the lateral orbital gyrus from the temporal pole.
classified primary sulci as those appearing before the The second, or the horizontal segment, is the longest and

13
14 SECTION I TECHNIQUES

䉴 TABLE 2–1. CLASSIFICATION OF BRAIN 䉴 TABLE 2–3. CLASSIFICATION OF FAIRLY


PRIMARY SULCI75 CONSTANT TERTIARY BRAIN SULCI75

Weeks of Gestation Sulcal Maturation Lobe Sulcus

13–15 Early sylvian fissure Frontal lobe Intermediate frontal


16–17 Cingulate sulcus Diagonal
Callosal sulcus Radiate
Parieto-occipital sulcus Anterior subcentral
19–20 Calcarine Parietal lobe Transverse parietal
22–23 Circular sulcus Intermediate sulci
25–26 Central sulcus Primary intermediate
Superior temporal sulcus (left) Temporal lobe Sulcus acousticus
Superior part of precentral sulcus Occipital lobe Various individual variations
Olfactory sulcus
28–30 Intraparietal sulcus
Inferior frontal sulcus
Branching of lateral sulcus terminal segment usually bifurcates at its terminus, form-
Paracentral sulcus ing long terminal ascending and short terminal descend-
Collateral sulcus ing branches. The latter is the shallower posterior trans-
Superior frontal sulcus verse supratemporal sulcus found on the right in about
two-thirds of cases. The cortical regions adjacent to the
lateral sulcus are the frontal, parietal, and temporal op-
ercula covering the insular lobe.
the deepest, coursing on the lateral surface of the hemi-
sphere. The third segment is limited anteriorly by the
transverse supratemporal sulcus, separating Heschl’s gyri
from the planum temporale and cutting into the superior
temporal gyrus. This last segment is complex and asym-
metrical, and correlates with hemispheric dominance. In
right-handed individuals, it ascends at an acute angle on
the nondominant right side, whereas on the left side it
assumes a superiorly directed oblique course (Fig. 2–1).
Several branches are distinguished on the second
segment; two sulci of similar length (2–3 cm) are noted
cutting into the inferior frontal gyrus: the horizontal ra-
mus and the vertical ramus run divergent courses and
define a triangular space whose apex faces the sylvian
fissure. These rami begin as branches from the sylvian
fissure, either separately in about two-thirds of subjects
or from a common trunk in the other one-third. The

䉴 TABLE 2–2. CLASSIFICATION OF BRAIN


SECONDARY SULCI75 Figure 2–1. Three-dimensional MR sagittal cut
through the perisylvian region. 1, lateral fissure; 1a,
Lobe Sulcus ascending ramus of lateral fissure; 1b, horizontal
ramus of lateral fissure; 1c, common trunk of
Frontal lobe Precentral ascending and horizontal rami; 1d, terminal ascending
Frontomarginal ramus; 1e, terminal descending ramus; 1f, temporal
Orbitofrontal planum; 1g, anterior transverse supratemporal sulcus;
Rostral sulci 1h, posterior transverse temporal sulcus; 1i;
Parietal lobe Subparietal intermediate transverse temporal sulcus; 2, central
Occipital lobe Paracalcarine sulci sulcus; 3, inferior precentral sulcus; 4, inferior
Lateral occipital postcentral sulcus; 5, inferior frontal gyrus; 6,
Transverse occipital precentral gyrus; 7, postcentral gyrus; 8, frontal
Lunate operculum; 9, parietal operculum; 10, superior
Temporal lobe Rhinal temporal gyrus; 11, supramarginal gyrus; 12, inferior
Transverse temporal frontal gyrus, pars triangularis; 13, inferior frontal
Inferior temporal gyrus, pars orbitalis; 14, inferior frontal gyrus, pars
Insular lobe Sulcus centralis insulae opercularis; 15, transverse temporal gyri; 16, superior
temporal sulcus; 17, middle temporal gyrus.
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 15

The horizontal ramus is a deep sulcus originating


from the lateral sulcus and cutting through the inferior
frontal gyrus to reach the circular sulcus of the insula
at its anterior border. Superiorly, it does not reach the
inferior frontal sulcus. It is absent on the right in 8% and
on the left in 16% of cases, according to Ono et al.13
This ramus forms the anterior border of the pars trian-
gularis (part of the classic Broca’s area), which is limited
posteriorly by the vertical ramus. In the absence of the
horizontal ramus, a diagonal sulcus may mimic the usual
triangular shape of the pars triangularis, but will not ex-
tend to the insular level.
The vertical ramus is defined by its extension to the
circular sulcus of the insula.14 It arises from the lateral
fissure as a separate sulcus or from a common trunk
with the horizontal ramus in one-third of the cases. It is
almost constant, absent in 3% of cases,14 and does not
reach superiorly the inferior frontal sulcus.
Figure 2–2. Three-dimensional MR view of the lateral
aspect of the brain. 1, central sulcus; 2, superior
frontal sulcus; 3, superior precentral sulcus; 4, inferior
䉴 THE INFERIOR PRECENTRAL frontal sulcus; 5, inferior precentral sulcus; 6,
intermediate frontal sulcus; 7, lateral sulcus; 8,
SULCUS postcentral sulcus or ascending segment of
intraparietal sulcus; 9, superior postcentral sulcus; 10,
The inferior precentral sulcus is an important landmark, intraparietal sulcus, horizontal segment; 11, lateral
representing an extension of the inferior frontal sulcus occipital or descending ramus of intraparietal sulcus;
in about 30% of subjects. 12, Central gyrus; 13, annectent gyrus or “pli de
passage” between middle frontal gyrus and
precentral gyrus; 14, postcentral gyrus; 15, superior
frontal gyrus; 16, middle frontal gyrus; 17, inferior
䉴 THE CENTRAL SULCUS frontal gyrus; 18, superior parietal gyrus; 19, inferior
parietal lobule; 20, common stem of vertical and
The central sulcus is a deep sulcus that extends verti- horizontal rami; 21, diagonal sulcus; 22, posterior
cally across the convexity and separates the frontal lobe subcentral sulcus; 23, transverse temporal sulcus;
24, superior temporal sulcus; 25, inferior temporal
from the parietal lobe. The central sulcus runs anteri-
sulcus; 26, superior temporal gyrus; 27, middle
orly oblique from superior to inferior. It is typically com- temporal gyrus.
posed of three curves defined by a superior genu (knee)
and an inferior genu. These genua are convex anteriorly
with an intervening concave bend. The cortex located hemisphere, until it terminates inferiorly just short of the
between these genua represents the portion of the pre- lateral fissure, with a hook-like end at its inferior margin
central gyrus that innervates the arm. Smaller spurs cut that contributes to the frontoparietal operculum. Anas-
into the adjacent gyri as well as two submerged and an- tomoses with the subcentral, precentral, and postcentral
nectant gyri are often noted, one at the superior and sulci occur in about 50% of cases. Extension into the
one at the inferior end of the central sulcus. The straight lateral fissure is found in less than 20% of cases, with
length of the adult central sulcus is 9 cm ± 0.6 cm, and an anastomosis with the anterior or posterior subcentral
when the central sulcus is measured with its curves, it sulci. Superiorly, the rolandic sulcus reaches the supe-
measures 10 cm ± 0.7 cm.15 rior border of the hemisphere and may extend over the
The central sulcus is deepest at the level of the mesial aspect of the hemisphere as a small sulcus giving
hand–arm representation, which lies roughly at the mid- the appearance of a comma,16 found in about 70% of
portion of the sulcus. At the level of the face represen- cases (Fig. 2–2).
tation, corresponding to its first 3 cm, it is slightly less
deep, averaging 15 mm. At the level of the trunk repre-
sentation, the recurrence of the annectant gyrus reduces 䉴 THE INFERIOR FRONTAL SULCUS
its depth to 12 mm. In the interhemispheric portion,
which is the site of the leg representation, the sulcal The inferior frontal sulcus arises anteriorly at the level
depth approaches 13 mm. The central sulcus is rarely of the lateral orbital gyrus, and courses roughly parallel
interrupted along most of its course along the lateral to the lateral sulcus. It is a deep sulcus almost reaching
16 SECTION I TECHNIQUES

frontal sulci. The superior end of the inferior precentral


sulcus is located anterior to the inferior end of the supe-
rior precentral sulcus (Fig. 2–1). The inferior end of the
inferior precentral sulcus may connect with the lateral
sulcus either directly or through the anterior subcentral
or the diagonal sulcus. The superior precentral sulcus is
usually smaller than the inferior precentral sulcus, and
has a complex relationship with the rolandic motor cor-
tex. It rarely reaches the superior hemispheric border,
and instead is replaced by one or two sulci: a horizontal
sulcus running parallel to the interhemispheric fissure,
that is, the shallow marginal precentral sulcus; and the
medial precentral sulcus, a vertical sulcus situated an-
terior to the precentral sulcus and perpendicular to the
interhemispheric border. Therefore, there is no clear sul-
cal demarcation between the supplementary motor area
Figure 2–3. Three-dimensional MR view of the (SMA) and the primary motor cortex (Fig. 2–3).
superior aspect of the brain showing the sulcation
and gyration of the upper central region. 1, central
sulcus; 2, superior precentral sulcus; 3, superior
postcentral sulcus; 4, medial precentral sulcus; 5, 䉴 THE INTRAPARIETAL SULCUS
superior frontal sulcus; 6, marginal ramus of callosal
sulcus; 7, Precentral gyrus; 8, superior extent of The intraparietal sulcus is divided into three parts, the
precentral gyrus; 9, postcentral gyrus; 10, superior ascending postcentral, horizontal, and descending or oc-
extent of postcentral gyrus; 11, superior frontal gyrus; cipital segments. The ascending segment is a vertical seg-
12, superior parietal gyrus; 13, interhemispheric
ment that corresponds to the inferior portion of the post-
fissure.
central sulcus and may extend, especially on the right
hemisphere, to the lateral sulcus. The horizontal or true
the insular plane and ending at the inferior precentral intraparietal segment has variable relationships with the
sulcus. More developed and constant than the superior inferior and superior postcentral sulci, but is continuous
frontal sulcus, it is interrupted in about 30% of cases. with both the inferior and superior postcentral sulci in
almost 40% of cases. The inferior postcentral segment is
continuous with the superior postcentral sulcus in about
䉴 THE SUPERIOR FRONTAL 60% of the cases. The postcentral sulcus shows wide
SULCUS variations and is frequently deeper than the central sul-
cus. Its horizontal segment is one of the deeper sulci
The superior frontal sulcus arises from the orbital in the human brain (2 cm in depth). The descending
margin of the hemisphere and courses parallel to the segment almost always terminates in the occipital lobe
interhemispheric fissure, extends along about two-thirds and may even reach its pole. This segment shows a
of the frontal lobe and gradually separates from the T-shaped ending in about two-thirds of cases,13 de-
interhemispheric fissure. It is frequently doubled or scribed as the transverse occipital sulcus. The superior
interrupted along its course,13 and ends posteriorly at end of the postcentral sulcus terminates most frequently
the precentral sulcus in a T-shaped branching in half of on the lateral aspect of the hemisphere without exten-
cases (Fig. 2–3). Anteriorly, it may anastomose with the sion to the medial aspect, in a Y-shaped configuration.
frontomarginal sulcus. At this Y-shaped end, it is joined by the marginal ra-
mus of the cingulate sulcus as it extends to the lateral
aspect of the hemisphere indenting its superior border
䉴 THE PRECENTRAL SULCUS (Fig. 2–3). This forms a useful landmark for identifying
the rolandic cortex on the interhemispheric surface.
In two-thirds of cases, the precentral sulcus is divided
into superior and inferior precentral sulci, which are sep-
arated by a connection between the precentral and the
middle frontal gyri, with the latter extending into the 䉴 THE SUPERIOR TEMPORAL
central motor cortex. It is composed of three segments SULCUS
in about 15% of cases. The precentral sulcus courses an-
terior and parallel to the central sulcus and is formed by The superior temporal sulcus, one of the oldest of the
the posterior bifurcations of the inferior and the superior primate brain, is also called the parallel sulcus because
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 17

it follows closely the course of the lateral fissure (sylvian


fissure). It is a deep sulcus (2.5–3 cm) almost reaching
the level of the inferior border of the insula. Infrequently,
it divides into anterior and posterior segments, usually
below the inferior end of the central sulcus. At the level
of the central sulcus, an inconstant sulcus acousticus may
be present, which originates from the parallel sulcus and
courses toward the lateral fissure, limiting the anterior
extent of the gyrus of Heschl. The posterior part of the
parallel sulcus is the angular sulcus, which penetrates
into the inferior parietal lobule.

䉴 THE FRONTOMARGINAL SULCUS

The frontomarginal sulcus is fairly constant and deep,


found at the frontal pole parallel to the orbital margin. Figure 2–4. Three-dimensional MR view of the brain
It is connected posteriorly with the middle frontal sul- showing the sulcal and gyral pattern of the inferior
cus more frequently than with the superior frontal. It frontal region and the anterior speech cortical area.
separates the transverse frontopolar gyri from the fron- 1, lateral sulcus; 2, horizontal ramus of lateral sulcus;
3, ascending ramus of lateral sulcus; 4, radiate
tomarginal gyrus inferiorly.
sulcus; 5, inferior precentral sulcus; 6, diagonal
sulcus; 7, central sulcus; 8, anterior subcentral sulcus;
9, posterior subcentral sulcus; 10, precentral gyrus;
䉴 GYRI OF THE LATERAL SURFACE 11, annectent gyrus; 12, postcentral gyrus; 13, pars
opercularis of inferior frontal gyrus; 14, pars
OF THE CEREBRAL HEMISPHERE triangularis of inferior frontal gyrus; 15, pars orbitalis
of inferior frontal gyrus; 16, superior temporal gyrus;
The convolutions on the lateral aspect of the cerebral 17, middle frontal gyrus; 18, superior frontal gyrus;
hemisphere determined by these primary fissures are the 19, frontomarginal gyrus; 20, inferior parietal lobule.
inferior, middle, and superior frontal gyri, the pre- and
postcentral gyri, and the inferior and superior parietal
convolutions, in the suprasylvian region, and the supe- runs into the basal orbital aspect of the hemisphere. The
rior, middle and inferior temporal gyri, in the infrasylvian opercular component merges with the lower extension
region. The gyri of the parietal and the temporal lobes of the precentral gyrus, together constituting the frontal
merge posteriorly with the variable occipital gyri, which operculum. The inferior frontal gyrus is more devel-
in turn are generally delimited by a superior, lateral, and oped in the dominant hemisphere, particularly the pars
inferior occipital sulci (Fig. 2–2). triangularis and pars opercularis, which together form
Broca’s area, the cortical region that is most essential
for expressive speech. The pars triangularis is traversed
䉴 THE FRONTAL LOBE in more than one-third of cases by the radiate sulcus
(Fig. 2–4).
The frontal lobe, the largest of the hemisphere, is com- The middle frontal gyrus is located between the in-
prised of four gyri. The gyri on the lateral aspect of ferior and the superior frontal sulci and is separated from
the frontal lobe are the inferior, the middle, and the su- the precentral gyrus posteriorly by the precentral sulci.
perior frontal gyri, which follow a course that roughly It is connected to the precentral gyrus by a deep an-
parallels the superior border of the hemisphere. Me- nectent gyrus. It is traversed by an inconstant interme-
dially, the frontal lobe consists of a hook-like gyrus diate frontal sulcus, which usually ends as a part of the
bounded inferiorly by the cingulate sulcus. Posteriorly, frontomarginal sulcus.
the precentral gyrus parallels the central sulcus, which The superior frontal gyrus is situated between the
forms the boundary between the frontal and parietal superior frontal sulcus and the dorsal margin of the
lobes. hemisphere. It continues on the medial aspect of
The inferior frontal gyrus is situated between the the hemisphere as the medial frontal gyrus and is con-
inferior frontal sulcus and the lateral sulcus and includes nected posteriorly to the central gyrus. The precentral
both horizontal and vertical rami. These rami divide the gyrus is located between the central sulcus and the in-
gyrus into three parts: the pars orbitalis, the pars trian- ferior and superior frontal sulci. It is limited inferiorly
gularis, and the pars opercularis. The orbital component by the lateral sulcus and extends superiorly to reach the
18 SECTION I TECHNIQUES

superior border of the hemisphere, where it is continu-


ous with the paracentral lobule on the medial aspect of
the hemisphere (Fig. 2–2).

䉴 THE PARIETAL LOBE

The parietal lobe is located superior to the lateral fis-


sure and behind the central sulcus, extending posteri-
orly to an arbitrary line connecting the lateral extent of
the parieto-occipital sulcus to the preoccipital notch. It
extends to the medial aspect of the hemisphere as the
medial postcentral gyrus anteriorly and as the precuneus
gyrus posteriorly. Its largest portion on the lateral surface
of the hemisphere is divided into three gyri by the intra-
parietal sulcus: the inferior parietal, the superior parietal,
and the postcentral gyri.
The postcentral gyrus is found posterior to the cen- Figure 2–5. Three-dimensional MR view of the lateral
tral sulcus, with its lower end connected to the inferior aspect of the brain showing the sulcal and gyral
anatomy of the inferior temporo-parieto-occipital
precentral gyrus. The inferior parietal lobule is situated
region and the posterior speech cortical area. 1,
between the lateral fissure inferiorly, the horizontal seg- interhemispheric fissure; 2, lateral sulcus; 3, terminal
ment of the intraparietal sulcus superiorly, and the as- ascending ramus of lateral sulcus; 4, parallel sulcus;
cending postcentral segment of the intraparietal sulcus 5, terminal ascending branch of parallel sulcus or
anteriorly. It is composed from front to back as the angular sulcus; 6, intraparietal sulcus, horizontal
supramarginal gyrus arching over the terminal ascend- segment; 7, sulcus intermedius primus; 8, sulcus
ing ramus of the lateral fissure, the angular gyrus arching intermedius secundus; 9, superior occipital sulcus; 10,
transverse occipital sulcus; 11, lateral occipital sulcus;
over the extremity of the upturned branch of the paral-
12, inferior temporal sulcus; 13, Postcentral gyrus; 14,
lel sulcus, and the posterior parietal gyrus, which may inferior parietal lobule; 15, superior parietal gyrus; 16,
cap the posterior end of the inferior temporal sulcus. supramarginal gyrus; 17, angular gyrus; 18, posterior
The supramarginal and the angular arched convolutions parietal gyrus; 19, superior temporal gyrus; 20, middle
are separated by a short sulcus, the primary intermediate temporal gyrus; 21, inferior temporal gyrus; 22,
sulcus.17 The angular gyrus may be separated from the inferior occipital gyrus; 23, superior occipital gyrus;
posterior parietal by the secondary intermediate sulcus18 24, middle occipital gyrus, 25 occipital pole.
(Fig. 2–5).
The superior parietal lobule is located dorsal to the
inferior parietal lobule, limited inferiorly by the intra- in number: the superior, middle, and inferior temporal
parietal sulcus, anteriorly by the superior postcentral gyri (Fig. 2–2).
sulcus, and extends posteriorly to the lateral extremity The superior temporal gyrus is located between the
of the parieto-occipital sulcus, beyond which it passes lateral fissure above and the parallel superior temporal
into the occipital lobe as the arcus parieto-occipitalis sulcus below. Its anterior extent contributes to the for-
or the superior parieto-occipital “pli de passage” of mation of the temporal pole, and its posterior extremity
Gratiolet.19 merges with the supramarginal gyrus. The dorsal surface
of this gyrus called the operculoinsular surface is divided
into an opercular and an insular segment. The former is
䉴 THE TEMPORAL LOBE located in relation to the frontal and parietal opercula
and the latter is related to the insula. One or two trans-
Somewhat pyramidal in shape, the temporal lobe has verse temporal gyri of Heschl,20 cross the dorsal aspect
lateral, basal, and dorsal aspects and an anterior apex of the superior temporal gyrus, obliquely forward, at the
or pole. The lateral aspect is bounded superiorly by the depth of the lateral fissure. More frequently doubled on
lateral fissure, which separates it from the frontoparietal the right side, these gyri are separated at least partly by
lobes. Caudally, it is continuous with the inferior parietal an intermediate transverse temporal sulcus. These gyri
lobule superiorly, and with the occipital lobe, inferiorly. are posteriorly separated from the planum temporale,
Ventrally, the temporal lobe extends to the collateral sul- by the transverse supratemporal sulcus of Holl21 origi-
cus at the basal aspect of the hemisphere, which sepa- nating from the lateral fissure. The frontal boundary of
rates it from the limbic lobe. The lateral convolutions of the Heschl gyri is marked by the anterior limiting sulcus
the temporal lobe, oriented anteroposteriorly, are three of Holl.
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 19

The middle temporal gyrus is separated from the lentiform nucleus, separated from it laterally to mesially
superior temporal gyrus by the superior temporal sulcus by the extreme capsule, the claustrum, and the external
and bounded inferiorly by the inferior temporal sulcus, capsule. The sulci of the insula bear a relatively constant
which is regularly interrupted. This gyrus is continuous relationship with the overlying cortical sulci. The central
posteriorly with the angular gyrus superiorly, and with sulcus appears to be continuous with the central sulcus
the occipital lobe inferiorly. of the insula, interrupted at the level of the hidden cen-
The inferior temporal gyrus is bounded superiorly tral operculum.
by the inferior temporal sulcus and extends inferiorly
over the basolateral border of the cerebral hemisphere,
to the inferior surface limited by the occipitotemporal 䉴 TOPOGRAPHICAL AND
sulcus. It is largely discontinuous extending like the oc-
FUNCTIONAL ANATOMY OF THE
cipitotemporal gyrus close to the preoccipital notch. At
this level, it is continuous posteriorly and inferiorly with SENSORIMOTOR CORTEX
the inferior occipital gyrus.
The primary motor and sensory cortices include the
precentral and postcentral gyri. The extent of the sen-
䉴 THE OCCIPITAL LOBE sorimotor complex was defined mostly on the basis of
stimulation studies performed under local anesthesia by
The occipital lobe occupies the posterior aspect of the several workers in the field. Penfield22 defines the pre-
hemisphere and is the smallest of all hemispheric lobes. central gyrus as a “sensorimotor functional unit.” This
It is formed by the presence of two longitudinal parieto- definition was later extended to the concept of a central
occipital “plis de passage” of Gratiolet: the first occupies lobe by Rasmussen23 (Fig. 2–2).
the superior aspect of the hemisphere and joins the su-
perior parietal gyrus to the superior occipital gyrus, lim-
ited laterally by the occipital segment of the intraparietal THE PRECENTRAL GYRUS
sulcus; the second joins the angular gyrus to the mid-
dle occipital gyrus. This gyrus is the largest of the lateral Anatomically, the precentral gyrus can be divided into
aspect of the occipital lobe and may be subdivided into four segments, defined by its three bends and the para-
superior and inferior portions by the occipital lateral sul- central lobule.
cus, which may anastomose anteriorly with the parallel The inferior segment is convex anteriorly, and it
sulcus. Two other temporal occipital “plis de passage,” is close to the lateral fissure, which marks the inferior
which are separated by an inconstant inferior occipital boundary of the precentral gyrus. It commonly commu-
sulcus that may correspond to a side branch of the in- nicates with the postcentral gyrus, forming the central
ferior temporal sulcus, occupy the inferior lateral aspect operculum. Medially, it reaches the insula and frequently
of the occipital lobe. The anterior extent of the inferior communicates with the pars opercularis of the frontal
occipital gyrus is ill-defined and continuous anteriorly lobe.
with the inferior temporal gyrus (Fig. 2–5). The middle segment is convex posteriorly. The
junction between the inferior and middle segments is
characterized by a tapering of the gyrus, which corre-
䉴 THE INSULA OF REIL sponds to the transition area between face and thumb
representation. This segment has no clearly defined lim-
The insula of Reil is the smallest of the cerebral lobes its anteriorly, as it extends into the premotor area, due
found in the depth of the lateral fissure. It is triangular to the interruption of the precentral sulcus in this region.
in shape with an apex directed anteriorly and inferiorly, Posteriorly, it is sharply bound by the central sulcus and
called the monticulus, and overhangs the falciform sul- medially by the corona radiata.
cus and the preinsular region. The latter is connected The superior segment is convex anteriorly. In its
to the anterior perforated substance through the limen initial segment, it is sharply distinct from the premo-
insulae. The insula is separated from the frontoparietal tor cortex. Toward the interhemispheric fissure, these
and the temporal opercula by the circular sulcus. boundaries become difficult to define as this gyrus
The insula proper is constituted of convergent gyri merges with the SMA. It is sharply bound posteriorly
presenting a fan-like arrangement, usually separated into by the central sulcus, superiorly by the interhemispheric
three short and one or two long convolutions by the cen- fissure, and medially by the corona radiata. The direction
tral sulcus insulae, the deepest and longest of all insular of the superior segment assumes a less oblique course
furrows reaching the circular sulcus. It originates from than that of the middle segment.
the superior limiting sulcus and is directed obliquely to- The paracentral segment occupies the posterior ex-
ward the falciform sulcus. The insular lobe covers the tent of the paracentral lobule with no sharp anterior
20 SECTION I TECHNIQUES

boundaries. Posteriorly, it appears to be demarcated for 䉴 IMAGING APPROACHES FOR


a short distance by the central sulcus. Most stimulation THE LOCALIZATION OF THE
studies do not, however, corroborate a functional cor-
CENTRAL SULCUS
relate to this anatomy, as primary motor and sensory
responses are not elicited in the inferior portion of the
Different approaches have been used for the localization
paracentral lobule.
of the central sulcus. Historically, indirect approaches
have relied on skull landmarks and, more recently, on
brain reference coordinates. Two widely used indirect
THE POSTCENTRAL GYRUS methods are the Talairach method based on the AC–PC
(anterior commissure–posterior commissure) reference
The postcentral gyrus can also be divided into four seg- plane8,25,26,27,28 and the Olivier method based on the cal-
ments. Its configuration closely resembles the precentral losal reference plane.29,30,31 Direct anatomic approaches
gyrus. Inferiorly, in the opercular region, it is wider and rely on identification of the central sulcus with mod-
thicker than its motor counterpart. The middle and su- ern imaging modalities. Various authors have described
perior segments are thinner and more sharply defined landmarks for localization of the central sulcus, which
by the postcentral sulcus. Superiorly, since the postcen- are visible on axial and sagittal MR scans,12,32 ). Iden-
tral sulcus terminates caudal to the marginal ramus of tification of the superior frontal sulcus and the distinct
the cingulate sulcus, the primary sensory area of the leg “hand know” on axial MR image usually permits straight-
extends beyond the paracentral lobule. forward identification of the central sulcus. On sagittal
images, the marginal ramus of the cingulated sulcus can
usually be followed superiorly to identify the postcentral
䉴 THE PREMOTOR CORTEX sulcus, which lies one sulcus posterior the central sulcus.
Using oblique 2D cuts obtained parallel to the “forniceal
The premotor cortex is a transitional area located be- reference plane,” the pericentral anatomy can be eas-
tween the frontal pole and the primary motor cortex. Its ily and precisely displayed.62 All these 2D methods lack
boundaries in humans are not well defined and gyral the ability to visualize the full extent of the central sul-
patterns in this region appear to vary considerably be- cus. For this reason, 3D MR has been used as a superior
tween subjects. The anterior boundary is arbitrarily de- alternative. Another approach relies on direct identifica-
fined as a line joining the anterior extent of the SMA with tion of the sensorimotor cortex using functional imaging
the frontal eye field. Posteriorly, the premotor cortex is techniques such as positron emission tomography, mag-
delimited by the precentral sulcus. netoencephalography, or functional MRI. While these
methods add additional complexity to image acquisition,
they allow identification of sensorimotor cortex in sub-
䉴 THE MOTOR AND SENSORY jects with aberrant anatomy, such as patients with corti-
REPRESENTATION IN THE cal dysplasia or with tumors that efface nearby gyri and
sulci.
CENTRAL CORTEX
Although there is clear evidence for a functional overlap
in the representation of specific body areas,24 the pri-
mary motor and sensory cortices that straddle the central 䉴 GYRAL ANATOMY AND
sulcus follow a generally orderly pattern of somatotopic IMAGING OF THE ANTERIOR
organization that is well represented in the classic ho- SPEECH AREA
munculus of Penfield and Rasmussen.22 These authors
hypothesized that the motor and sensory units were ar- The anterior speech region was defined by
ranged in horizontal strips extending from precentral to Broca33,34,35,36 as including the posterior third of the
postcentral sulci and across the central sulcus. Thus, the left inferior frontal gyrus. Rasmussen defines the speech
sensorimotor cortex can be divided from inferior to su- area as including the pars triangularis and pars opercu-
perior into four functional units:1 the face unit, extending laris of the dominant frontal lobe. However, extensive
from the lateral sulcus (sylvian fissure) superiorly to ap- cortical stimulation studies by Penfield and Roberts,37
proximately 3 cm;2 the hand–arm unit, starting with the Penfield and Rasmussen,22 Ojemann et al.38 and Sanai
thumb representation corresponding to the inferior genu et al.90 have shown marked individual variations in the
and ending at the shoulder area;3 the trunk unit, border- anterior speech area. In fact, many cortical stimulation
ing on the interhemispheric fissure;4 the leg–foot unit, and functional imaging studies have shown anterior
located at the mesial aspect of the hemisphere within speech representation outside of the anatomically
the paracentral lobule (Fig. 2–2). defined Broca’s area (Fig. 2–4).
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 21

Broca’s anterior speech area includes the cytoar- parietal lobule were pointed out by Naidich et al.12 who
chitecturally defined Brodmann’s area 45. This speech reported the presence of accessory supernumerary gyri
area is anatomically limited anteriorly by the horizontal in the inferior parietal lobule: a presupramarginal gyrus,
ramus of the lateral sulcus and posteriorly by the in- found in 16% of cases on the left and 4% on the right
ferior segment of the precentral sulcus. Inferiorly it is side, and a preangular gyrus found in 28% on the left
limited by the posterior ramus of the lateral sulcus and and 16% on the right side.
superiorly by the inferior frontal sulcus. Two gyri thus The posterior speech area includes: Heschl’s
constitute this area, the pars triangularis and the pars gyrus,20 the temporal planum, the parietal operculum,
opercularis of the inferior frontal lobe. The pars trian- the parietal and temporal speech related gyri.
gularis is limited anteriorly by the horizontal ramus and Heschl’s gyrus is a hidden arch-like gyrus located
posteriorly by the vertical ramus of the lateral sulcus. entirely within the lateral sulcus and assuming a poste-
It is characteristically U-shaped in the dominant hemi- rior oblique orientation within the supratemporal plane.
sphere and Y-shaped in the nondominant hemisphere. This relationship is seen on axial MR cuts performed in
It is traversed superiorly by the incisura capitis branch the sylvian plane orientation, as obtained using the “CH–
of the radiate sulcus. The pars triangularis extends deep PC (chiasmatico-commissural) reference plane.”40,41 Its
into the third frontal convolution, reaching the level of anatomical landmarks are: the chiasmal point (CH) an-
the insula. The pars opercularis is located in between the teriorly and the PC posteriorly, readily shown on a mid-
vertical ramus and the inferior precentral sulcus. It is lim- sagittal MR scout view.
ited inferiorly by the sylvian fissure, reaching the inferior Heschl’s gyrus is limited anteriorly and posteriorly
frontal sulcus superiorly. It communicates with the pars by transverse supratemporal sulci. According to Bailey
triangularis superiorly and anteriorly. Posteriorly and in- and von Bonin, the posterior transverse supratemporal
feriorly it can communicate with the precentral gyrus. It sulcus is the most constant sulcus and is easily visualized
may be divided into two parts by the shallow diagonal on the lateral surface of the temporal lobe originating
sulcus.14 More frequently found on the left (72%) than from the sylvian fissure with a distinct anterior oblique
over the right (64%) side, the diagonal sulcus appears orientation, located in close proximity to the postcen-
to almost always be connected to the sylvian fissure on tral sulcus. It separates Heschl’s gyrus from the planum
the right and only infrequently on the left, according to temporale. The anterior transverse supratemporal sulcus
Ono et al.13 constitutes the anterior border of Heschl’s gyrus, reach-
ing the lateral aspect of the temporal lobe at the level of
the central sulcus. The intermediate sulcus is inconstant
and does exist when two Heschl’s gyri, mainly on the
䉴 GYRAL ANATOMY AND right side, are noted.42 As Duvernoy notes, the sulcus
IMAGING OF THE POSTERIOR acusticus is the furrow that originates from the parallel
SPEECH AREA sulcus and heads toward Heschl area. Heschl’s gyri are
typically larger and more obliquely oriented on the left
Because of its marked variability,37 the anatomic local- side and shorter on the right side (Fig. 2–1). Although
ization and extent of the posterior speech area is diffi- it is assumed that the entire Heschl’s gyrus corresponds
cult and can only be determined by cortical stimulation. to the primary auditory cortex, stimulation studies have
Functional MRI has contributed some insight into the elicited responses from its posteromedial extent close
organization and localization of speech. to the level of the insula.43 Strainer et al.,44 using pure
Numerous stimulation studies and cortical excisions tone activation, showed different tonotopic organization
(Fig. 2–5) have shown that the receptive speech area is within Heschl’s gyri, depending on the frequency of the
most frequently infrasylvian and includes the posterior auditory stimulus: responses elicited for tones in the
extent of the first temporal convolution and the mid and lower frequencies (1000 Hz) predominated in the lat-
posterior second temporal gyrus. Speech appears not eral transverse temporal gyrus, whereas those of higher
to extend to the third temporal convolution (the infe- frequencies (4000 Hz) appear localized in the medial
rior temporal gyrus) and is, in some cases, exclusively transverse temporal gyrus.
suprasylvian. Thus, it is localized to the supramarginal The temporal planum is a triangular cortical sur-
and angular gyri. The inferior parietal lobule is divided face, apparent as early as the 29th week of gestation,51
into three contiguous convolutions: the supramarginal, studied initially by von Economo and Horn45 and subse-
the angular, and the posterior parietal gyri, which are quently by others.27,42,46,47,48,49,50 It is limited laterally by
separated by two intermediate sulci. The primary inter- the lateral sulcus and anteriorly by the posterior trans-
mediate sulcus is present in 24% of cases on the right verse supratemporal sulcus. Its posterior limit is not well
side and in 80% on the left. The secondary intermediate defined. Habib et al.52,53,54 and Steinmetz et al.,55,56,57
sulcus is present in 64% of cases on the right and 72% on considered the terminal descending branch of the lat-
the left.13 Variations of the gyral pattern of the inferior eral sulcus as the posterior limit. In the absence of
22 SECTION I TECHNIQUES

this branch, the temporal planum includes the entire


supratemporal extent of the third division of the sylvian
fissure (Fig. 2–1). Cytoarchitecturally, it corresponds to
the posterior portion of area 22 of Brodmann. In most in-
dividuals, the left temporal planum is wider than its right-
sided counterpart and is formed by several small gyri
that assume a superior oblique orientation. The right-
side planum has a smaller cortical surface and a flat
surface.54,58 Dominance of the left cerebral hemisphere
for speech was noted early in the 19th century by Marc
Dax in 1836,63 promoter of the concept of speech local-
ization in the left hemisphere followed by Paul Broca36
and Wernicke,64 according to Alajouanine.65
Imaging identification of the posterior speech area
has been carried out extensively by Salamon and
collaborators54,59,60,61 using the bicommissural AC–PC Figure 2–6. Two-dimensional MR parasagittal cut of
coordinates. These authors demonstrated that the peri- the brain, showing the main sulci and gyri of the
sylvian cortical speech area and the inferior parietal lob- mesial aspect of the hemisphere. 1,callosal sulcus; 2,
cingulate sulcus; 3, marginal ramus of cingulate
ule may be reliably explored using a limited number
sulcus; 4, central sulcus; 5, paracentral sulcus; 6,
of cuts (four slices, 5 mm thick), oriented parallel to parieto-occipital sulcus; 7, subparietal sulcus; 8,
the bicommissural plane, and performed at 45 mm and superior rostral sulcus; 9, inferior rostral sulcus; 10,
50 mm above the reference line, to display the posterior calcarine sulcus, posterior segment; 11, calcarine
part of the first and second temporal gyri, and at 60 mm sulcus, anterior segment; 12, transverse parietal
and 70 mm, to explore the supramarginal and angular sulcus; 13, cingulate gyrus; 14, medial frontal gyrus;
gyri.60 Using MRI, the temporal planum is best explored 15, paracentral lobule; 16, postcentral gyrus; 17,
precentral gyrus; 18, subcallosal gyrus; 19,
in the axial plane using CH–PC coordinates and in coro-
fronto-orbital gyri; 20, isthmus cinguli; 21, cuneus; 22,
nal plane perpendicular to the sylvian CH–PC reference precuneus; 23, lingual gyrus; 24, parahippocampal
as obtained using the PC–OB (posterior commissure– gyrus; 25, parietolimbic “pli de passage”; 26,
obex) reference plane. This coronal approach permits cuneolingual gyrus; 27, retrocalcarine sulcus; 28,
identification of the sylvian fissure followed on more frontopolar gyri; 29, gyrus descendens; 30, temporal
posterior sections on the left and located higher on the pole.
right. It allows a direct evaluation of the depth of the
planum and an easy depiction of Heschl’s gyri on both in the subcallosal region, before it sweeps around the
sides. The axial cuts performed parallel to the CH–PC ref- genu paralleling the corpus callosum, separating the me-
erence plane, which corresponds to the sylvian fissure dial frontal gyrus from the cingulate gyrus, ending as a
orientation plane,39,40,62 best evaluate the supratemporal marginal ramus in the parietal lobe and separating the
region displaying, from anterior to posterior, the gyral precuneus from the paracentral lobule. The marginal ra-
anatomy of the planum polare, the transverse temporal mus has a fairly constant relationship to the central sul-
gyri, and posteriorly the temporal planum. cus, ending about 10 mm posterior to it. Up to three
interruptions are frequently noted along its course lead-
ing to invaginations of the mesial frontal gyrus into the
䉴 THE MESIAL SURFACE OF THE cingulate gyrus, as the “plis de passage frontolimbiques”
CEREBRAL HEMISPHERE of Broca (Fig. 2–6).
The superior rostral sulcus courses anteroposteri-
The specific gyral patterns characteristic of the inter- orly around the rostrum of the corpus callosum and ends
hemispheric area are influenced by the development of closely behind the frontal pole. It is roughly parallel to
the callosal connections. Sulci and gyri of the mesial as- the anterior cingulate sulcus and is very frequently dou-
pect of the hemisphere are evident on a sagittal and bled by an inferior, shallower accessory rostral sulcus
2D-parasagittal cuts of the brain (Fig. 2–6). (Fig. 2–6).

䉴 THE CINGULATE SULCUS AND 䉴 THE PARIETO-OCCIPITAL


THE ROSTRAL SULCI SULCUS
Also called the callosomarginal sulcus, the cingulate sul- The parieto-occipital sulcus is a deep, constant sulcus
cus begins below the rostrum of the corpus callosum, of the primate brain, situated on the posterior mesial
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 23

aspect of the hemisphere, extending downward from lower half in the ventral part. Regarding the disposition
the dorsal margin forward to the caudal aspect of the of the macular fibers, Holmes considered it to be located
splenium where it joins the stem of the calcarine sulcus. on the tip of the posterior pole of the cerebral hemi-
It continues as the external incisure on the lateral aspect sphere, whereas according to Polyak, a wide distribution
of the hemisphere for a short distance (about 10–12 mm) of these fibers along the calcarine sulcus is observed.
cutting deeply into its edge. A line connecting the The striate cortex (area 17) is intimately related to
parieto-occipital incisure to the preoccipital notch draws the parastriate cortex (area 18), which lies in a portion of
the arbitrary boundary on the lateral surface separating the occipital lobe contiguous to the latter. The Gennari
the occipital lobe from the temporal and parietal lobes. band is not found in this area. The peristriate area (area
19) is much larger than area 18, lying on the lateral as-
pect of the cerebral hemisphere and extending beyond
the medial aspect of the hemisphere to surround the
䉴 THE CALCARINE SULCUS AND parastriate area from above and below. Most of the peri-
THE STRIATE VISUAL CORTEX striate area lies in the posterior part of the parietal lobe.
It extends inferomedially to the posterior portion of the
The calcarine sulcus arises behind and just below the temporal lobe (Fig. 2–5).
splenium of the corpus callosum and proceeds posteri- Considering imaging, many authors have proposed
orly toward the occipital pole. This deep sulcus is di- reference planes and have tried to describe the ideal an-
vided into two segments at the point of its junction with gulations to use with respect to the orbitomeatal line or
the parieto-occipital sulcus. The first, cephalad to this to an anthropologically based line.39,40,62,67,68,69,70,71,72,73,
junction, is the anterior calcarine sulcus. The second is The best compromise would be a reference plane suit-
the posterior calcarine sulcus, a caudal division that typ- able for exploration of both the optic pathways and the
ically ends posteriorly in a bifurcation on the medial as- brain. For this reason, the neuro–ocular plane, as the
pect of the hemisphere. One or two submerged gyri, the anatomophysiological and anthropological cephalic ref-
anterior and the posterior cuneolingual folds of Déjerine, erence plane, appears to be the most suitable for study-
may be found within the posterior calcarine segment, ing the visual pathway.68,69,71,72 It is, in our opinion,
and may be seen on parasagittal MR images. The up- also efficient enough for evaluation of the retrochias-
per and the lower lips of the posterior calcarine sulcus matic pathways in routine practice. In order to facilitate
and the lower lip only of the anterior calcarine sulcus, the neuroanatomical approach and optimize topometric
correspond to the striate visual cortex (area 17). studies of the brain and the retrochiasmal visual path-
The striate or primary visual cortex is limited pos- ways, the CH–PC line, defining a CH–PC reference plane
teriorly by the lunate sulcus, when present, and may is used.39,40,74,75
extend beyond the occipital pole of the hemisphere for The calcarine fissures and striate cortex are shown
a distance of 1–1.5 cm. This extension onto the medial on the midsagittal cut of the brain, and may also be de-
posterior aspect of the occipital pole shows important picted on coronal and axial cuts. Their close relationship
individual variations. The cortex of the visual sensory to the occipital horns of the lateral ventricle may aid in
area identified histologically by a white line (or stria- their recognition. However, there is no ideal cephalic
tion), the line of Gennari, which is a layer of myelinated orientation for studying the calcarine fissures, as they
terminals of optic radiations fibers. The parieto-occipital are variable in shape among individuals. Note that the
sulcus limits the striate cortex anteriorly. An average of CH–PC reference line intersects posteriorly the common
67% of the visual cortex is buried in the depth of the stem of the parieto-occipital and calcarine sulci.
calcarine fissure and its branches. The area of the striate
cortex is greater below than above the calcarine fissure,
extending about 2 cm more anteriorly. The striate cor- 䉴 GYRI OF THE MESIAL SURFACE
tex is situated between the cuneus, a wedge-shaped area OF THE CEREBRAL HEMISPHERE
located above the calcarine sulcus whose surface is gen-
erally indented by one or two small sulci, and the lingual Seven gyri constitute the mesial hemisphere.76 These
gyrus lying below, between the calcarine sulcus superi- are described as follows, from anterior to posterior
orly and the collateral sulcus inferiorly (Fig. 2–6). The (Fig. 2–6).
cuneus and lingual gyri are both part of the extrastriate
visual cortex.
Considering the functional and anatomic aspects of THE GYRUS RECTUS
the visual cortex,66 there is general agreement regarding
the following conception of cortical representation: the The gyrus rectus is limited inferiorly by the floor of the
upper half of each retina is retinotopically represented anterior cranial fossa, laterally by the olfactory sulcus,
in the dorsal part of the occipital striate cortex and the and superiorly by the superior rostral sulcus (Fig. 2–7).
24 SECTION I TECHNIQUES

THE MEDIAL FRONTAL GYRUS AND


THE SUPPLEMENTARY MOTOR AREA

The medial frontal gyrus is limited ventrally and ante-


riorly by the gyrus rectus. Superiorly, it constitutes the
superior border of the hemisphere and posteriorly it is
limited by the paracentral sulcus. The SMA as described
by Penfield and Welch77 is of variable extent and located
in the mesial aspect of the first frontal convolution, an-
terior to the representation of the lower extremity in the
primary motor cortex and superior to the cingulate sul-
cus. It may extend superiorly onto the lateral convexity
of the hemisphere. The anatomic boundaries of the SMA
have been a subject of considerable debate.78 The an-
terior extent of the SMA was defined by Talairach and
Bancaud79 as a line perpendicular to the AC–PC line,
passing by the anterior-most extent of the genu of the
corpus callosum.

THE PARACENTRAL LOBULE

The paracentral lobule is limited superiorly by the su-


Figure 2–7. Three-dimensional MR view of the sulcal perior border of the hemisphere and anteriorly by the
and gyral anatomy of the basal aspect of the paracentral sulcus. Posteriorly, the paracentral lobule is
hemisphere. 1, olfactory sulcus; 2, orbital sulci (“H”); bounded by the marginal ramus, which ends at the hemi-
2a, medial orbital sulcus; 2b, lateral orbital sulcus; 2c, spheric border and in more than two-thirds of cases
arcuate or transverse orbital sulcus; 3, extends to the lateral surface. The paracentral lobule
interhemispheric fissure; 4, gyrus rectus; 5, medial proper usually extends anterior to the precentral sulcus.
orbital gyrus; 6, lateral orbital gyrus; 7, anterior orbital
The relationship of the marginal ramus to the central
gyrus; 8, Posterior orbital gyrus; 9, superior temporal
gyrus and temporal pole; 10, middle temporal gyrus; area has not been adequately assessed. The marginal ra-
11, inferior temporal gyrus; 12, parallel sulcus; 13, mus has a relatively constant relationship with the cen-
parahippocampal gyrus; 14, uncus; 15, optic chiasm; tral sulcus and ends posterior to the central sulcus. The
16, pons; 17, medulla oblongata. anterior limit of the primary motor area over the mesial
hemisphere tends to be unclear. The postcentral sulcus
extends to the mesial surface in about one-third of ex-
amined hemispheres. Most commonly, it is posterior to
the marginal ramus (Fig. 2–3). Thus, the primary sensory
THE CINGULATE GYRUS leg–foot area appears to extend anatomically beyond the
limits of the paracentral lobule.
The cingulate gyrus is limited ventrally by the callosal
sulcus, ventrally and anteriorly by the anterior paraol-
factory sulcus, superiorly by the cingulate sulcus, su-
periorly and posteriorly by the subparietal sulcus, and THE PRECUNEUS
posteriorly and inferiorly by the anterior calcarine sul-
cus. It is continuous with the parahippocampal gyrus The precuneus is limited anteriorly by the marginal ra-
through the isthmus. The anterior cingulate is followed mus, superiorly by the superior border of the hemi-
in the subcallosal region, where it abuts the subcal- sphere, posteriorly by the parieto-occipital sulcus, and
losal gyrus, which is limited anteriorly by the ante- inferiorly by the subparietal sulcus. The parieto-occipital
rior subcallosal sulcus and bounded posteriorly by the sulcus is a deep constant sulcus that demarcates the pre-
posterior subcallosal sulcus, the latter limiting anteri- cuneus from the cuneus of the occipital lobe. The sub-
orly the paraterminal gyrus. Together with the subcal- parietal sulcus is a variable sulcus, which may show var-
losal, the isthmus hippocampi, and the parahippocampal ious branching or may appear as a posterior branching
gyri, the cingulate gyrus forms the limbic lobe of Broca of the cingulate sulcus. It is frequently traversed by one
(Fig. 2–6). or two parietolimbic “plis de passage” (Fig. 2–6).
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 25

THE CUNEUS tudinal sulci are the medial and the lateral orbital sulci,
which divide the orbital surface into medial and lateral
Triangular in shape, the cuneus is the only occipital orbital gyri and an intermediate orbital cortex. This is
gyrus that is consistently well delimited. It is bounded subdivided transversely into anterior and posterior mid-
anteriorly by the parieto-occipital sulcus, superiorly by dle orbital gyri by the arcuate orbital sulcus. The lateral
the superior border of the hemisphere, and inferiorly by orbital sulcus limits the orbitofrontal lobe from the lateral
the posterior calcarine sulcus. It is continuous with the aspect of the inferior frontal gyrus (Fig. 2–7).
surface of the lateral hemisphere. The calcarine sulcus
extends anteriorly and stops at the most posterior ex-
tent of the collateral sulcus. The cuneus is connected to 䉴 THE POSTERIOR BASAL
the posterior aspect of the cingulate gyrus by a deep
TEMPORAL SULCI AND GYRI
cuneolimbic “pli de passage” (Fig. 2–6).
The basal surface of the temporal lobe extends later-
ally from the inferior lateral border of the hemisphere
THE LINGUAL GYRUS to the mesial temporal border at the lateral wing of the
transverse fissure. It extends from the temporal pole to
The lingual gyrus constitutes the inferior mesial aspect the inferior occipital lobe without definite demarcation.
of the occipital lobe. It is bordered superiorly by the cal- From mesial to lateral, two longitudinal sulci, the collat-
carine sulcus and connected to the cuneus through the eral and the occipitotemporal divide the basal tempo-
retrocalcarine sulcus by one or two cuneolingual gyri.6 It ral lobe into three gyri, the parahippocampal gyrus, the
is continuous anteriorly with the parahippocampal gyrus fusiform gyrus, and the inferior temporal gyrus, at the
(Fig. 2–6). junction between the lateral and the inferior aspects of
the hemisphere.
The collateral sulcus, a primary sulcus also called
䉴 THE INFERIOR AND MESIAL the medial occipitotemporal sulcus, is a constant, elon-
SURFACE OF THE CEREBRAL gated S-shaped sulcus of the basal aspect of the temporal
HEMISPHERE lobe. It usually originates near the temporal pole and
separates the parahippocampal gyrus and the lingual
Sulci and gyri of the basal aspect of the frontotemporal gyrus from the more laterally situated fusiform gyrus.
lobes are best imaged and analyzed on coronal cuts per- The lateral occipitotemporal sulcus is a secondary
formed parallel to the PC–OB brain reference line.39,40,80 sulcus that runs lateral and roughly parallel to the
Coronal anatomic correlations and cross-sectional atlas collateral sulcus, near the inferolateral margin of the
are available.9,75 We will separate the basal surface into hemisphere, and end near the preoccipital notch. It is
an anterior basal orbitofrontal lobe and a posterior basal frequently interrupted by additional small sulci. It con-
temporal lobe (Fig. 2–7). stitutes the medial boundary of the occipitotemporal or
fusiform gyrus. This gyrus does not reach the temporal
pole anteriorly and its width increases from the pole to
䉴 THE ANTERIOR BASAL its posterior extremity before it merges with the inferior
ORBITOFRONTAL SULCI AND occipital lobe (Fig. 2–7).
GYRI
The orbital surface of the frontal lobe includes a primary 䉴 THE LIMBIC LOBE AND THE
sulcus, the olfactory sulcus, and a secondary composite MESIAL TEMPORAL REGION
sulcus, the orbital sulci, that varies considerably between
individuals. The limbic lobe, corresponding to the “grand lobe lim-
The olfactory sulcus originates at the anterior per- bique” of Paul Broca includes anatomical structures bor-
forated substance. It courses in the posterior–anterior dering the diencephalon.85 Ontogenetically, this arch
plane roughly parallel to the anterior interhemispheric convolution located at the inferomedial aspect of the
fissure, and terminates about 1.5 cm posterior to the cerebral hemisphere is limited continuously by major
frontal pole. It is closely related anatomically to the olfac- primary sulci represented mainly by the cingulate sulcus
tory tract. This sulcus separates the gyrus rectus medially dorsally and ventrally, the collateral sulcus posteriorly
from the orbital gyri laterally. and the rhinal sulcus anteriorly. These sulci laterally limit
The orbital sulci show numerous variations,81 with the uncus and the parahippocampal gyrus, which curves
two longitudinal sulci connected by a transverse furrows posteriorly through the narrow isthmus around the sple-
arranged in the shape of an “H” or “X” or “K.” The longi- nium of the corpus callosum covering and surrounding
26 SECTION I TECHNIQUES

it completely. It is separated from the corpus callosum Coronal slices performed parallel to the PC–OB refer-
by the callosal sulcus. Anteriorly, below the callosal ros- ence plane are very useful for volumetric analysis of the
trum, it merges with the paraterminal gyrus, which cor- amygdala–hippocampal complex.
responds to the prehippocampal rudiment, immediately Embryologically, the fornices together with the
in front of the lamina terminalis. Starting in the subcal- choroidal fissures develop along an oblique plane with
losal region, the limbic lobe structures encircle the upper a complex arciform course. This shape is focused on
brainstem and the corpus callosum as the subcallosal, by temporalization of the brain. Imaging along the “for-
cingulate, parahippocampal and hippocampal, and den- niceal plane” is therefore particularly useful.75 This plane
tate gyri (Fig. 2–8). is defined by a line joining the lateral aspect of the fim-
Imaging of the limbic structures has dramatically bria fornix in the hippocampus with the lateral aspect
improved with the advent of high-resolution 3D MRI. of the crus fornicis. These structures are visualized and

Figure 2–8. Two-dimensional reformation of the mesial temporal region using the
“forniceal plane” (A) to display the limbic belt medially (B) and the central lobe laterally
(C). (A) MR topogram showing the orientation of the sagittal oblique “forniceal
(fimbria-fornix)” reference plane oriented at about 45˚ to the midsagittal plane. (B) 1,
hippocampus; 2, fimbria; 3, fornix; 4, amygdala; 5, splenium of corpus callosum; 6,
collateral sulcus; 7, parahippocampal sulcus; 8, fusiform gyrus; 9, rostral sulcus; 10,
occipital lobe; 11, temporal pole; 12, interhemispheric fissure. (C) 1, central sulcus; 2,
superior precentral sulcus; 3, inferior precentral sulcus; 4, superior frontal sulcus; 5,
inferior frontal sulcus; 6, inferior postcentral sulcus; 7, superior postcentral sulcus; 8,
intraparietal sulcus; 9, lateral sulcus; 10, precentral gyrus; 11, postcentral gyrus; 12,
superior frontal gyrus; 13, middle frontal gyrus; 14, inferior frontal gyrus; 15, inferior
parietal lobule.
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 27

joined on the coronal PC–OB reference plane itself. Se- PC–OB reference line displaying the nuclear mass dor-
rial oblique cuts are then acquired or reformatted parallel sal to the hippocampal formation and rostral to the tip
to the forniceal plane to cover the entire mesial temporal of the inferior horn of the lateral ventricle. The supe-
lobe and extend to the convexity. This reference plane rior aspect of the amygdala is partly continuous with the
appears especially interesting for the study of hippocam- inferior margin of the claustrum, and is separated from
pal, amygdala, and temporal polar cortex formations. We the inferior aspect of the putamen and the pallidum by
have found the lateral slices particularly helpful for the fibers of the external capsule and the ventral striatum,
study of the central brain region as well. The frontal in close contact with the optic tract. At this level, the
lobe perisylvian convolutions are well depicted, facili- amygdala fuses with the tip of the tail of the caudate nu-
tating the identification of the sulcal and gyral anatomy cleus. The amygdala is a corticonuclear transition area
of this region (Fig. 2–8). located dorsomedially in the temporal lobe and form-
The anatomy of the mesial temporal region is partic- ing the ventral superior and medial walls of the inferior
ularly complex and is composed of the temporal pole, horn of the lateral ventricle. Numerous subdivisions of
the amygdala, the hippocampus, the parahippocampal the amygdala have been reported. According to Gloor,85
gyrus, and certain cortical areas related to these struc- the amygdala can be divided into three large subnu-
tures. clei: the basolateral, corticomedial, and central group of
The temporal polar cortex corresponds to area 38 nuclei.
of Brodmann. Its cytoarchitectural characteristics have Perhaps the earliest description of the hippocam-
been defined by Insausti.82 The temporal pole, extend- pus was given by Arantius (1587) and J.G. Duvernoy
ing from the tip of the temporal lobe laterally and ventro- provided the first illustrations in 1729, according to H.
laterally to the level of the superior or inferior temporal Duvernoy.86,87 Reviews of the complex terminology may
gyri, is characterized by the appearance of the superior be found in Tilney88 and Klingler.89 The hippocam-
or inferior temporal sulci. Ventromedially, the temporal pus consists of two cortical lamina each interlocked
polar cortex blends with the perirhinal area and dor- with the other: the Ammon’s horn or the hippocampus
sally extends to the level of the limen insulae. The tem- proper, and the gyrus. The hippocampal formation may
poral pole includes three surfaces: dorsal, lateral, and be subdivided morphologically into a precommissural,
mesial. a supracommissural, and a retrocommissural portion.
The entorhinal area occupies most of the anterior The retrocommissural portion represents the hippocam-
extent of the parahippocampal gyrus and extends dor- pal formation, the other parts being vestigial anatomi-
somedially to the periamygdaloid cortex (area 28 of cal structures. The precommissural portion occupies the
Brodmann). Caudomedially, it reaches the presubiculum caudal part of the area subcallosa. The supracommis-
and laterally extends into the medial bank of the collat- sural portion is represented by the indusium griseum or
eral sulcus. The entorhinal area extends rostrally from supracallosal gyrus. The hippocampus is located in the
the level of the limen insulae and medially to the pe- mesial temporal lobe and protrudes into the temporal
riamygdaloid cortex. It is separated from the amygdala horn of the lateral ventricle after it rolls in on itself along
by the sulcus semiannularis. Its extension over the un- the hippocampal sulcus during ontogenesis. Its rostral
cus includes the ambient gyrus and reaches laterally to extremity extends ventrally to the amygdala. The major
the level of the collateral sulcus. According to Insausti topographical structures of hippocampal formation are
et al.,83 the rhinal sulcus has a limited value in defining seen on coronal anatomic and MR cuts perpendicular to
the extent of the entorhinal area. At the level of the pos- the long axis of the inferior horn of the lateral ventricle as
teromedial uncus, its extent is limited by the hippocam- obtained according to the PC–OB reference plane62,75,80
pal fissure. The perirhinal area corresponds to area 35 and/or perpendicular to the CH–PC reference plane.39,40
of Brodmann.84 It follows the collateral sulcus along its The shape of the hippocampus and the localization of its
rostrocaudal extent, occupying its fundus and its medial constitutive structures are explained by the cortical fold-
bank. In addition, the perirhinal area comprises area 36. ing, which occurs during development. The external as-
This area is medial to area 20 of Brodmann and anteriorly pects of the subiculum and the dentate gyrus face each
continues with area 38. The perirhinal cortex constitutes other in the depths of the hippocampal sulcus. When
the lateral border of the vestigial rhinal sulcus and, more opening the hippocampal sulcus, the dentate gyrus is
caudally, the collateral sulcus. seen between the hippocampal sulcus, which is below,
The amygdala, or amygdaloid nuclear complex, re- and the fimbria, which are above. The dentate gyrus may
ceived its name from its shape, which resembles that be followed backward accompanied by the fimbria until
of an almond. It constitutes, along with the hippocam- it reaches the splenium of the corpus callosum. The cor-
pus, one of the two major telencephalic components tical zone medial to the dentate gyrus is the subiculum,
of the limbic system. The topographical relationships of which is contiguous with the parahippocampal gyrus
the amygdala are complex and are best appreciated and or entorhinal area. Ammon’s horn, which is the third
evaluated on MR coronal sections obtained using the longitudinal structure of the hippocampal formation, is
28 SECTION I TECHNIQUES

the hippocampus proper. The hippocampal formation is ability and in a manner that is practically useful in the
covered by a lamina of whiter matter fibers, the alveus, clinical setting. One major caveat, however, is that struc-
which converges on the medial aspect of the hippocam- tural imaging cannot account for functional variability
pus to form the fimbria and then the fornix. that frequently exists across different individuals. For this
The hippocampal formation is divided into two reason, functional imaging and other functional mapping
parts, the hippocampus proper or the Ammon’s horn, techniques must often be used to supplement anatomic
and the dentatus gyrus or the fascia dentata. The hip- data.
pocampus proper is a cylindrical structure, voluminous
anteriorly, extending as much as 4–5 cm from the tip of
the temporal horn to the splenium of the corpus callo- REFERENCES
sum where it becomes continuous with the fornix. Its
course assumes a strong inner concavity. It is commonly 1. Leuret F, Gratiolet P. Anatomie Comparée du Système
divided into three parts: head, body, and tail. The hip- Nerveux, Considérée Dans ses Rapports Avec l’intelligence,
pocampal head is characterized by several digitations vols 1 and 2. Masson, Paris: Baillière, 1839.
that are apparent on its ventricular surface, representing 2. Meynert T. Der Bau der Grosshirnrinde und seine
the foldings of the hippocampal formation. At the level örtlichen Verschiedenheiten, nebst einem pathologisch-
anatomischen Corollarium, 1867/1869; Vierteljahrsch Psy-
of the body, the hippocampal proper is limited later-
chiatr 1:77-93, 125-217, 381-403; 2:88-113.
ally by the collateral eminence, created in the temporal 3. Ariens Kappers CU. Anatomie Comparée Du Système
horn by the collateral sulcus. Its ventricular surface, the Nerveux. Paris: Masson and Cie, 1947.
alveus, constitutes the output of the hippocampus, and is 4. Economo von C, Koskinas GN. Die Cytoarchitektonik der
continuous with the fimbria fornix. The anterior segment Hirnrinde Des Erwachsenen Menschen. Textband und At-
of the tail resembles the body of the hippocampus. It is, las. Vienna: Springer, 1925.
however, distinct due to a progressive thinning of the 5. Larroche J-C, Feess-Higgins A. Development of the human
hippocampus proper and increase in size of the fimbria foetal brain. An anatomical atlas. Editions INSERM. Paris:
fornix. The posterior aspect of the tail has a markedly dif- Masson, 1987.
ferent geometry as two important structures, the dentate 6. Déjerine J. Anatomie Des Centres Nerveux, vols 1 and 2.
gyrus and the fimbria fornix, assume divergent destina- Paris: Rueff, 1895.
7. Delmas A, Pertuiset B. Topométrie Crânioencéphalique
tions. The dentate gyrus courses toward a supracallosal
chez L’homme. Paris: Masson, 1959.
destination to become the indusium griseum, and the 8. Talairach J, Szikla G, Tournoux P, et al. Atlas D’anatomie
fornix, together with the rest of the body, moves toward Stéréotaxique Du Télencéphale. Paris: Masson, 1967.
an infracallosal course. 9. Tamraz J. Atlas d’anatomie céphalique dans le plan
The uncus includes parts of the amygdala, neuro-oculaire (PNO). MD Thesis. Paris: Université René
hippocampus, and piriform cortex. It is posteriorly con- Descartes, Paris V, 1983.
tinuous with the parahippocampal gyrus and its postero- 10. Cabanis EA, Tamraz J, Iba-Zizen MT. Corrélations anato-
lateral extent is separated from this latter structure by miques normales dans 3 dimensions, selon l’orientation du
the uncal sulcus. The amygdala produces a characteris- plan neuro-oculaire In: Atlas d’IRM de L’encéphale et de la
tic protrusion on the surface of the uncus, the semilunar Moelle: Aspects Normaux. Paris: Masson, 1988, pp. 11-120.
gyrus. The semilunar gyrus is separated from the entorhi- 11. Duvernoy H, Cabanis EA, Iba-Zizen, et al. The Human
Brain Surface Three-Dimensional Sectional Anatomy and
nal cortex by the semiannular or amygdaloid sulcus. An-
MRI. Vienna: Springer, 1991.
teriorly, the amygdala or rhinal cortex blends with the 12. Naidich P, Valavanis G, Kubik S. Anatomic relation-
prepiriform cortex, the lateral olfactory striae and is sep- ships along the low-middle convexity. I. Normal spec-
arated from the perforated substance by the endorhinal imens and magnetic resonance imaging. Neurosurgery
sulcus. Posteromedially, it is bordered by the optic tract. 1995;36(3):517-531.
The posterior aspect of the uncus is occupied by parts 13. Ono N, Kubik S, Abernathy DG. Atlas of the Cerebral Sulci.
of the head of the hippocampal formation. Stuttgart: Thieme, 1990.
14. Eberstaller O. Das stirnhirn. Ein Beitrag zur Anatomie des
Oberfläche des Großhirns. Vienna: Urban and Schwarzen-
䉴 CONCLUSION berg, 1890.
15. Bailey P, von Bonin G. The Isocortex of Man. Urbana: Uni-
versity of Illinois Press, 1951.
Contemporary MR imaging is remarkably adept at iden-
16. Broca P. Nomenclature cérébrale. Rev Anthropol 1878;2:3.
tifying specific cortical and subcortical brain structures, 17. Jensen J. Die Furchen und windungen der menschlichen
including regions that are well correlated with specific grosshirnhemispharen. Allg Z Psychiatr 1870;27:473-515.
clinically important functions, such as the primary mo- 18. Eberstaller O. Zur Oberflächen-anatomie der grosshirn-
tor are in the precentral gyrus. Careful imaging meth- hemisphären. Wien Med Bl 1884;7:644-646.
ods described in this chapter allow clinicians to identify 19. Gratiolet P. Mémoire Sur les Plis Cérébraux de L’homme et
many functionally important areas with very high reli- Des Primates. Paris: Bertrand, 1854.
CHAPTER 2 STRUCTURAL IMAGING FOR IDENTIFICATION OF FUNCTIONAL BRAIN REGIONS 29

20. Heschl RL. Uber Die Vordere Quere Schläfenwindung Des Magnétique: le plan chiasmatocommissural. CR Acad Sci
Menschlichen Grosshirns. Vienna: Braumuller, 1878. Paris, 311, 1990;III:115-121.
21. Holl M. Die Insel des Menschen- und Affenhirns in 40. Tamraz J, Saban R, Reperant J, et al. A new cephalic
ihrer Beziehung zum Schläfenlappen. Sitz Ber D k Akad reference plane for use with magnetic resonance imag-
Wiessensch Wien, Math Naturw Kl 1908; 117:365-410. ing: the chiasmato-commissural plane. Surg Radiol Anat
22. Penfield W, Rasmussen T. The cerebral cortex of man. 1991;13:197-201.
A Clinical Study of Localization of Function. New York: 41. Tamraz J, Brain Anatomy. Structural neuroimaging, part IV.
Macmillan, 1950. In Epilepsy Surgery, 2nd edition, edited by HO Luders and
23. Rassmussen T. The neurosurgical treatment of focal Y Comair. Lippincott-Williams & Wilkins, 2000, p. 1088.
epilepsy. In Epilepsy, recent views on its therapy, diag- 42. Pfeifer RA. Pathologie der Hörstrahlung und der corticalen
nosis and treatment (Moderns problems of pharmacopsy- Hörsphäre. In Handbuch der Neurologie, vol 6, edited by
chiatry, vol 4), edited by E Niedermeyer. Basel: Karger, O Bumke, O Foerster. Berlin: Springer, 1936, p. 533.
1969. 43. Liegeois-Chauvel C, Musolino A, et al. Localization of the
24. Penfield W, Jasper H. Epilepsy and the Functional Anatomy primary auditory area in man. Brain 1991;114:139-153.
of the Human Brain. Boston: Little Brown, 1954. 44. Strainer JC, Ulmer JL, Zerrin Yetkin F, et al. Functional MR
25. Talairach J, De Ajuriaguerra J, David M. Etudes of the primary auditory cortex: an analysis of pure tone
stéréotaxiques des structures encéphaliques chez l’homme. activation and tone discrimination. AJNR Am J Neuroradiol
Presse Med 1952;28:605-609. 1997;18(4):621-623.
26. Szikla G, Talairach J. Coordinates of the Rolandic sulcus 45. Economo C von, Horn L. Ueber windungsrelief, masse
and topography of cortical and subcortical motor responses und rindenarchitektonik der supratemporalfläche, ihre in-
to low frequency stimulation in a proportional stereotactic dividuellen und ihre seitenunterschiede. ZGes Neurol Psy-
system. Confin Neurol (Basel) 1965;26:474-475. chiatrie 1930;130:678-757.
27. Szikla G, Bouvier G, Hori T. In vivo localization of brain 46. Geschwind N, Levitsky W. Human brain: left-right asym-
sulci by arteriography: a stereotactic anatomoradiological metries in temporal speech region. Science 1968;161:
study. Brain Res 1975;95:497-502. 186-187.
28. Talairach J, Tournoux P. Co-Planar Stereotaxic Atlas of the 47. Galaburda AM, Le May M, Kemper TL, et al. Right-left asym-
Human Brain. 3-Dimensional Proportional System: an Ap- metries in the brain. Science 1978a;199:852-856.
proach to Cerebral Imaging. Stuttgart: Thieme, 1988. 48. Galaburda M, Sanides F, Geschwind N. Human brain –
29. Olivier A, Peters TM, Clark JA, et al. Intégration de cytoarchitectonic left-right asymmetries in the temporal
l’angiographie numérique, de la résonance magnétique, de speech region. Arch Neurol 1978b;35:3-7.
la tomodensitométrie et de la tomoencephalographie. Neu- 49. Galaburda AM, Corsiglia J, Rosen GD, et al. Planum tempo-
rophysiol Clin 1987;17:25-43. rale asymmetry: reappraisal since Geschwind and Levitsky.
30. Villemure JG, Marchand E, Peters, et al. Magnetic resonance Neuropsychologia 1987;25(6):853-868.
imaging stereotaxy: recognition and utilization of the com- 50. Galaburda AM. The planum temporale. Arch Neurol
missures. Appl Neurophysiol 1987;50:57-62. 1993;50:457.
31. Devaux B, Meder JF, Missir O, et al. La ligne rolandique: 51. Larroche JC. Maturation morphologique du système
une ligne de base simple pour le repérage de la région nerveux central: ses rapports avec le développement
centrale. J Neuroradiol 1996;23:6-18. pondéral de foetus et son âge gestationnel. In Regional
32. Naidich T, Brightbill TC. Systems for localizing frontopari- Development of the Brain in Early Life, edited by M
etal gyri and sulci on axial CT and MRI. Int J Neuroradiol Minkowski. Philadelphia: Davis, 1967, pp. 247-256.
1996;4:313-338. 52. Habib M, Renucci RL, Corbaz JM, et al. Computed tomog-
33. Broca P. Perte de la parole, ramollissement chronique et raphy localization of visual and auditory primary cortex.
destruction partielle du lobe antérieur gauche du cerveau. Otoneuroophtalmol 1983a;55:39-46.
Société d’Anthropologie. Bull SocAnthropol 1861;11:235- 53. Habib M, Renucci RL, Corbaz JM, et al. Reperage en to-
238. modensitométrie des aires corticales visuelle et auditive
34. Broca P. Remarques sur le siège de la faculté du language primaires. Rev Otoneuroophtalmol 1983b;55:39-46.
articulé, suivies d’une observation d’aphémie (perte de la 54. Habib M, Renucci RL, Vanier M, et al. CT assessment of
parole). Bull Soc Anat 1861;36:330-357. right-left asymmetries in the human cerebral cortex. J Com-
35. Broca P. Localisation des fonctions cérébrales – siège du put Assist Tomogr 1984;8(5):922-927.
langage articulé. Bull Soc Anthropol Paris 1863;4:200-208. 55. Steinmetz H, Rademacher J, Huang Y, et al. Cerebral asym-
36. Broca P. Du siège de la faculté du langage articulé metry: MR planimetry of the human planum temporale. J
dans l’hémisphère gauche du cerveau. Bull Soc Anthropol Comput Assist Tomogr 1989;13:996-1005.
1865;VI:377-393. 56. Steinmetz H, Volkmann J, Janke L, et al. Anatomical left-
37. Penfield W, Roberts L. Speech and Brain–Mechanisms. right asymmetry of language-related temporal cortex is dif-
Princeton, NJ: Princeton University Press, 1959. ferent in left and right handers. Ann Neurol 1990a;29:315-
38. Ojemann G, Ojemann J, Lettich, et al. Cortical language lo- 319.
calization in left, dominant hemisphere: an electrical stim- 57. Steinmetz H, Rademacher J, Janke L, et al. Total surface
ulation mapping investigation in 117 patients. J Neurosurg of temporoparietal intrasylvian cortex: diverging left-right
1989;71:316-326. asymmetries. Brain Lang 1990b;39:357-372.
39. Tamraz J, Saban R, Cabanis EA, et al. Définition d’un 58. Szikla G, Bouvier G, Hori T, et al. Angiography of the Hu-
plan de référence céphalique en Imagerie par Résonance man Brain Cortex. Berlin: Springer, 1977.
30 SECTION I TECHNIQUES

59. Gelbert F, Bergvall U, Salamon G, et al. CT identification cephalic reference determined by the optic pathways.
of cortical speech areas in the human brain. J Computer Biométrie Hum 1982;17:21-48.
Assist Tomogr 1986;10(1):39-46. 74. Tamraz J. Neuroradiologic investigation of the visual sys-
60. Salamon G, Gelbert F, Alicherif A, et al. Le repérage in tem using magnetic resonance imaging. J Clin Neurophys-
vivo des aires corticales du langage. Rev Neurol (Paris) iol 1994;11:500-518.
1987;143:8-9, 580-587. 75. Tamraz JC, Comair YG. Atlas of Regional Anatomy of the
61. Rumeau C, Gouaze A, Salamon, et al. Identification of cor- Brain using MRI – with Functional Correlations. Berlin:
tical sulci and gyri using magnetic resonance imaging: a Springer, 2000, p. 330. Reprint 2006.
preliminary study. In Brain Anatomy and Magnetic Reso- 76. Comair Y, Hong SC, Bleasel A. Invasive investigation and
nance Imaging, edited by A Gouaze and G Salamon. Berlin: surgery of the supplementary motor area. In Advances
Springer, 1988, pp. 11-32. in Neurology, vol 70, edited by HO Lüders. Philadelphia:
62. Tamraz JC, Comair YG. Cephalic reference lines suitable Lippincott-Raven, 1996.
for neuroimaging. In Atlas of Regional Anatomy of the 77. Penfield W, Welch K. The supplementary motor area of
Brain Using MRI – with Functional Correlations, edited the cerebral cortex. A clinical and experimental study. Arch
by JC Tamraz and YG Comair. Berlin: Springer, 2000, Neurol Psychiatr 1951;66:289-317.
pp. 11-50. 78. Luders HO. The supplementary sensorymotor area: an
63. Dax M. Lesions de la moitié gauche de l’encéphale coinci- overview. In Supplementary Sensorimotor Area (Advances
dant avec l’oubli des signes de la pensee. Gazette Hebdo in Neurology, vol 70), edited by HO Luders. Philadelphia:
Med Chir 1836;33:259. Lippincott-Raven, 1996, pp. 1-16.
64. Wernicke C. Das Urwindungsstystem des menschlichen 79. Talairach J, Bancaud J. The supplementary motor area
gehirns. Arch Psychiatr Nervenkr 1876;6:298-326. in man. Anatomic functional findings by stereotactic en-
65. Alajouanine T. L’Aphasie et le Langage Pathologique. Paris: cephalography in epilepsy. Int J Neurol 1966;5:330-347.
Baillière, 1968. 80. Goncalves Pereira PM, Oliveira E, Secca MF. Assessment of
66. Stensaas SS, Eddington DK, Dobelle WH. The topography the preferred scout sagittal orientation for temporal lobe
and variability of the primary visual cortex in man. J Neu- imaging with magnetic resonance. MAGMA 2004;18(1):19-
rosurg 1974;40:747-755. 25.
67. Hilal SK, Trokel SL. Computerized tomography of the or- 81. Kanai T. Über die Furchen und Windungen der Or-
bit using thin sections. Semin Roentgenol 1977;12:137- bitalfläche des Stirnhirns bei Japanern. Okajimas Fol Anat
147. Jpn 1938;18:229-306.
68. Cabanis EA, Salvolini U, Rodallec A, et al. Computed 82. Insausti R, Juottonen K, Soininen H, et al. MR volumet-
tomography of the optic nerve: part II Size and shape ric analysis of the human entorhinal, perirhinal and tem-
modifications in papilledema. J Comput Assist Tomogr poropolar cortices. AJNR Am J Neuroradiol 1998;19:659-
1978;2:150-155. 671.
69. Salvolini U, Cabanis EA, Rodallec A. Computed tomogra- 83. Insausti R, Amaral DG, Cowan WM. The entorhinal cor-
phy of the optic nerve. I. Normal results. J Comput Assist tex of the monkey. II. Cortical afferents. J Comp Neurol
Tomogr 1978;2:141-149. 1987;264:396-408.
70. Unsold R, Newton TH, Hoyt. CT examination technique of 84. Brodmann K. Vergleichende Lokalisationslehre der
the optic nerve. J Comput Ass – Zuckerkandl E (1906) Zur Großhirnrinde. Leipzig: Barth, 1909.
Anatomie der Fissura calcarina. Arb Neurol Inst Univ Wien 85. Gloor P. The Temporal Lobe and the Limbic System. Lon-
1980;13:25-61. don: Oxford University Press, 1997.
71. Cabanis EA, Iba-Zizen MT, Coin JL. Les voies visuelles 86. Duvernoy H. The Human Hippocampus, 2nd edition.
un “nouveau” plan d’orientation de la tête (Plan Neuro- Berlin: Springer, 1988.
Oculaire). Bull Soc Ophtalmol Fr 1981a;81:433-439. 87. Duvernoy H. The Human Hippocampus, 3rd edition.
72. Cabanis EA, Iba-Zizen MT, Pineau H, et al. CT scan- Berlin: Springer, 2005, p. 232.
ning in the “neuro-ocular plane”: the optic pathways as a 88. Tilney F. The hippocampus and its relations to the corpus
“new” cephalic plane. Neuroophthalmology 1981b;1:237- callosum. J Nerv Ment Dis 1939;89:433-513.
251. 89. Klingler J. Die makroskopische anatomie der ammonsfor-
73. Cabanis EA, Iba-Zizen MT, Pineau H, et al. Le plan neuro- mation. Denkschriften der schweizerischen naturforschen-
oculaire (PNO) en tomodensitométrie (TDM ou scanner den Gesellschaft. Fretz, Zürich, 1948;78:82.
RX), détermination d’un “nouveau” plan horizontal de 90. Sanai N, Mirzadeh Z, Berger MS. Functional outcome af-
référence céphalique orienté selon les voies visuelles/The ter language mapping for glioma resection. N Engl J Med
neuro-ocular plane (NOP) with CT: a new horizontal 2008;358(1):18-27.
Chapter 3
Functional MRI for Cerebral
Localization: Principles and
Methodology
Michael S. Beauchamp
Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, Texas

䉴 INTRODUCTION used to measure changes in blood oxygen in the human


brain linked to brain activity.6−8 This new method was
The widespread availability of magnetic resonance imag- termed functional MRI (fMRI) because it measured brain
ing (MRI) scanners has allowed clinicians a clearer pic- function, and to distinguish it from the more traditional
ture of brain structure and function. In addition to the anatomical or structural MRI.
static anatomy imaged with traditional MRI, functional
MRI (fMRI) allows for the measurement of brain function. 䉴 A THUMBNAIL SKETCH OF fMRI
The most widely used fMRI technique is known as blood
oxygen level dependent (BOLD) fMRI. BOLD fMRI has As neuronal activity in the brain fluctuates, the local
become a powerful tool for studying brain function, and blood oxygen content varies. A particular type of MR
has led to important advances in our understanding of image, a T2*-weighted image (effective T2), is sensitive
the basic neurobiology of the brain. However, there are to the blood oxygen content. What distinguishes fMRI
significant obstacles to translating these advances into from MRI is that instead of collecting a single image (to
clinically useful information. In this chapter, we give a inspect brain anatomy), a whole series of brain images
brief overview of the techniques used to create BOLD is collected to find brain areas with fluctuating blood
images and the data analysis necessary to identify ac- oxygen content. When this type of image is acquired,
tive brain areas. Finally, we examine the difficulties in active brain regions become brighter. Figure 3–1 shows
drawing clinical inferences from fMRI studies. a thumbnail sketch of a simple fMRI experiment. When
the patient views a blank screen, there is little activity
in visual cortex in the occipital lobe. When the patient
䉴 A BRIEF HISTORY OF fMRI views a picture, neurons in visual cortex become active
and the blood oxygenation level increases, resulting in
Paul Lauterbur and Peter Mansfield received the 2003 an increase in MR signal intensity. When the picture dis-
Nobel Prize for developing techniques to transform NMR appears, neurons in visual cortex cease firing and the
(nuclear magnetic resonance), which gives information MR signal returns to baseline intensity.
about a uniform sample of material, into a technique that In order to perform an fMRI experiment, special-
provides a two-dimensional (2D) image of an object.1,2 ized equipment over and above that required for typical
This method is now known as MRI, a considerably more clinical MRI is required. Most importantly, sensory stim-
patient-friendly term than the original name, which was uli must be delivered to the patient, and these stimuli
“NMR Zeugmatography” (zeugma, Greek for yoke, refer- must by synchronized with the MR image acquisition
ring to joining a magnetic field gradient to a local region (Fig. 3–2).
to form an image). Because of its medical importance, A thriving cottage industry exists to provide this
it is likely that a future Nobel Prize will honor those equipment; often the scanner manufacturers will com-
involved in the development of fMRI. A probable hon- bine and resell the equipment from a combination
oree is Seiji Ogawa, who discovered that hemoglobin of vendors as part of an “fMRI package.” However,
could serve as a natural contrast agent to measure blood third party vendors often offer capabilities unavailable
oxygenation.3−5 Following Ogawa’s discovery, a num- from the scanner manufacturer. Headphones allow the
ber of groups realized that this BOLD MRI could be delivery of auditory stimuli (such as single words) or

31
32 SECTION I TECHNIQUES

Figure 3–1. Top: visual display viewed by a subject. The subject views a blank screen,
followed by an image of a face, then a blank screen. Bottom: T2∗ MR images from the
subject’s brain collected while viewing the display. An axial slice showing visual cortex
in the occipital lobe (green arrow). When neuronal activity in visual cortex increases,
there is a concomitant brightening of the voxels in visual cortex. When the stimulus is
turned off, the brightness returns to baseline.

instructions (tap the fingers in your right hand). The nication, but is not sufficient for an auditory fMRI experi-
stock headphones included with MR scanners are pneu- ment in which the brain responses to carefully calibrated
matic (as in old-fashioned airline headsets): sound is stimuli are measured. Therefore, at least three vendors
generated in a speaker and transmitted through plastic produce high-fidelity MR compatible headphones. Visual
tubes to the headset. This is adequate for patient commu- stimuli may be delivered using goggles (containing small

A B

D E C

Figure 3–2. Apparatus for delivering sensory stimuli and recording behavioral
responses during a functional MRI experiment. (A) Picture of the MR scanner showing
bed and head coil. (B) Close-up view with patient holding button response device,
wearing earphones, and viewing visual stimulus in mirror attached to head coil. (C)
View behind the scanner, showing screen with visual display viewed by subject via
mirror (white arrow) and video-based infrared eye tracker (ASL Inc., green arrow). (D)
Close-up view of button response device (Current Designs, Inc.). (E) Close-up view of
somatosensory stimulation device consisting of a piezoelectric bender attached to
subject’s hand with an elastic bandage.
CHAPTER 3 FUNCTIONAL MRI FOR CEREBRAL LOCALIZATION 33

LCD panels) or via a mirror viewed by the subject. This map (Fig. 3–4A). To acquire this single anatomical vol-
allows the patient to see visual stimuli projected onto ume takes about 5 minutes. Images for BOLD activity
a screen behind or in front of the scanner. Piezoelec- are acquired much more quickly, usually limited by the
tric vibrotactile devices allow delivery of somatosensory speed of the magnetic field gradients used for image ac-
stimuli. Because subjects may close their eyes or fall quisition. The typical goal in an fMRI study is to collect
asleep, a useful tool for fMRI is an eye tracker, which data from the entire brain. If a single slice of approx-
allows experimenters to view a video image of the sub- imately 3 mm thickness is acquired in 50 ms, we can
ject’s eyes and monitor their behavioral state. It is also collect 40 such slices within a 2-second repetition time
important to monitor responses to be sure subjects are or TR (the TR is the time required to collect the entire
able to perform the task. For instance, a subject might volume of interest). This allows us to collect data from
press one button when seeing a male face and another a 12-cm-thick slab of tissue, or about the inferior to su-
button when seeing a female face. This helps the sub- perior extent of the average human brain (Fig. 3–4B).
ject maintain alertness and makes sure that the subject BOLD fMRI studies generate huge amounts of data.
understands the instructions and is performing the task. A 5-minute anatomical MR scan series generates a single
brain volume. The single volume is viewed as a series
of slices on a workstation or a light box, and can be
䉴 MRI AND MR SCANNERS quickly read by a radiologist who examines each slice
for abnormalities. In the same 5-minute period, a typi-
MRI scanners can be viewed as complex computers that cal fMRI scan series would collect brain volumes every 2
can use different programs (known as pulse sequences) seconds, producing 150 brain volumes (Fig. 3–4C). Even
to acquire many different kinds of images that depend if these brain volumes were each manually examined, it
on different physiological properties. Because no ioniz- would be difficult to detect the slight changes in im-
ing radiation is used during MRI scanning, a typical MR age intensity that signal the presence of brain activity.
examination includes many different pulse sequences, Therefore, we can consider BOLD fMRI data analysis as
with the primary limit being the time that the subject a problem of compression, or reduction, in which the
can remain in the scanner. hundreds of brain volumes acquired are reduced to one
By far, the most common method for performing or a few brain volumes that can be easily examined by
fMRI of the brain is BOLD fMRI. To perform BOLD fMRI, the scientist or clinician.
a pulse sequence must be sensitive to the amount of
blood oxygenation in the tissue, and it must acquire im-
ages quickly. A typical spoiled-GRASS pulse sequence 䉴 ANALYZING fMRI DATA
used for anatomical imaging of the brain takes minutes
to acquire a single brain image. As generally applied, fMRI studies acquire data from thousands of brain loca-
BOLD fMRI uses a pulse sequence with two key fea- tions, leading to the analysis problem of deciding which
tures: first, it generates images with T2* contrast, a con- brain locations are involved in the experimental task. To
trast that is sensitive to the amount of oxygenated blood address this problem, techniques developed to analyze
in the tissue, and second, it generates images of the other types of time varying data have been adapted to
entire brain in a second or two (rather than minutes) fMRI.
using echo-planar imaging. These images are referred Figure 3–5 shows some sample raw data from vox-
to as T2*, echo-planar imaging (EPI), or functional im- els in the ventricle (where there is no neural response to
ages (as opposed to purely anatomical images). Figure the visual stimulus) and from voxels in the visual cortex
3–3 shows the basic physiological mechanism of BOLD (where there is a great deal of response to the visual
as it is currently understood: increased neuronal activity stimulus).
causes spatially localized increase in blood flow without The challenge faced by fMRI analysis is to accu-
as large an increase in oxygen consumption, resulting in rately distinguish these two populations, differentiating
a net increase in blood oxygenation and an increased active voxels from inactive voxels. An arbitrary statisti-
MR signal. cal threshold is used, which inevitably classifies some
truly active voxels as inactive, or vice versa. There is
no universally agreed upon threshold for use in creat-
䉴 COLLECTING MRI DATA ing activation maps, or even universally agreed upon
way of calculating significance; that is, a map that is la-
Because the rapidly acquired T2* images used to mea- beled as “active voxels, p < 0.05” in one study may use
sure BOLD activity have poor contrast and are of low a completely different statistical test than that used in a
resolution, fMRI experiments nearly always acquire a different study that uses the same nominal threshold.
high-resolution T1-weighted anatomical brain volume to Before fMRI, the most popular method for brain
serve as an anatomical underlay for the functional brain imaging was positron emission tomography (PET). In a
34 SECTION I TECHNIQUES

Figure 3–3. Mechanism of blood oxygen level dependent functional MRI. (A) The
cerebral vasculature in a small region of auditory cortex. A precapillary arteriole (red)
gives rise to a collateral capillary (green) which divided into two terminal capillaries
(orange and cyan) before joining with other capillary loops and entering a
postcapillary venule (blue). (Adapted from Harrison RV, Harel N, Panesar J, Mount RJ.
Blood capillary distribution correlates with hemodynamic-based functional imaging in
cerebral cortex. Cereb Cortex 2002;12(3):225–233.) (B) Enlargement showing
perivascular control structures near capillary branching points that may control blood
flood. (Adapted from Harrison RV, Harel N, Panesar J, Mount RJ. Blood capillary
distribution correlates with hemodynamic-based functional imaging in cerebral cortex.
Cereb Cortex 2002;12(3):225–233.) (C) At rest, there is relatively little blood flow into
an area of cortex. Black lines show a capillary, hexagonal shapes represent red blood
cells, red color represents oxyhemoglobin, and blue color represents
deoxyhemoglobin. Most of the hemoglobin is deoxygenated, weakening the echo
from nearby protons in water molecules (black sinusoidal curve). (D) During
activation, more blood flow enters the capillary. Only some of the extra oxygen is
extracted, resulting in less deoxygenated hemoglobin and greater signal from protons
in nearby water molecules.

typical PET experiment, only a single brain image would tween task epochs (e.g., subject looking at a picture) and
be collected for each condition for each subject. Then, rest epochs (e.g., subject looking at a blank screen). The
a subtraction was performed to show areas with differ- flaws in this approach are immediately apparent when
ential blood flow between conditions.9 examining Fig. 3–5C. A t-test assumes that values collect
In the early days of fMRI, people would treat the in task and baseline periods are similar to each other.
fMRI data like PET data, simply performing a t-test be- However, the MR time series in active voxels in visual
CHAPTER 3 FUNCTIONAL MRI FOR CEREBRAL LOCALIZATION 35

C B

Figure 3–4. Data collected in a typical functional MRI experiment. (A) High-resolution
anatomical image (T1-weighted) showing fine detail of anatomical structures. Green
lines show location of T2∗ -weighted images pictured in (B). (B) T2∗ -weighted
echo-planar images collected in a single 2-second repetition time (TR). 33 slices are
pictured in a 3 × 11 montage, from inferior (top left) to superior (bottom right). Each
slice is 3-mm thick, with 2.75 mm × 2.75 mm in plane resolution. (C) A T2∗ -weighted
brain volume is collected every 2 seconds, leading to 150 volumes in a typical
5-minute scan series.

cortex does not resemble a square wave, but more analyzing fMRI data was to use multiple regression.11 In
closely resembles a sine wave. Treating all points on this most common analysis method, also referred to as
the top half of the sine wave (for task epochs) and the multivariate generalized linear hypothesis or general
bottom half of the sine wave (for baseline epochs) as linear model, regressors are created that correspond to
equivalent is inaccurate. Therefore, the next stage in the temporal sequence of different events experienced
the development of fMRI analysis techniques was to use by the subject during scanning.12−14 Brain regions in
cross-correlation,10 which allows knowledge about the which the MR time series is time-locked to these events
expected shape of the MR time series to be incorporated are classified as “active.” In this section, we discuss
into the analysis. The next (and consensus) method for the methods and procedures for deciding whether any
36 SECTION I TECHNIQUES

A
B

Figure 3–5. (A) Axial slice through the brain. Grids show voxel locations (enlarged) of
voxels shown in (B) and (C). (B) A plot of the intensity in nine voxels in the red grid in
(A), located in a ventricle. Each black trace shows the intensity of that voxel in each of
150 images, collected every 2 seconds over a 5-minute scan series. No regular
variation in the intensity is apparent. (C) A plot of the intensity of the nine voxels in the
green grid shown in (A) located in visual cortex. The signal becomes brighter and
dimmer as the subject views visual stimuli.
CHAPTER 3 FUNCTIONAL MRI FOR CEREBRAL LOCALIZATION 37

brain area is active in a given stimulus or behavioral thermal noise. Effectively, this noise in the MR time se-
task. ries limits the ability to detect active brain regions. If the
To understand these statistical analyses, we use as fluctuations in the MR signal introduced by the stimulus
an example a simple visual stimulation experiment. Fig- are much smaller than the noise in the MR signal, active
ure 3–5 shows a single axial slice through a brain vol- regions cannot be detected.
ume, along with the time series. In a single 5-minute scan Visual examination of the MR time series is a crit-
series, we collected 150 such images. We may plot the ical step in performing quality control in fMRI experi-
intensity of each voxel in a simple x–y plot, with time ments. Hardware problems, such as hardware-generated
along the horizontal axis and image intensity along the “spikes” in the MR signal, can be easily detected. How-
vertical axis. Plotting the time series of a few neighboring ever, visual inspection of the time series in each voxel
voxels gives even more information. In voxels in visual to determine whether it is active or not is impractical.
cortex, we see a periodic variation in the image intensity, Therefore, quantitative methods are used to generate a
with seven peaks over the course of the 5-minute scan. measure of the degree of activity in each voxel.
These peaks correspond to the times at which the sub- We next show how the generalized linear hypoth-
ject was looking at visual stimuli (in this case, a movie esis can be used to analyze fMRI time series (Fig. 3–6).
entitled “Winged Migration”). The troughs correspond to Each voxel is fit separately, termed univariate analysis.
times at which the subject was viewing a blank fixation The model requires the analyst to generate a series of
screen. predictions about what might be happening in the time
A number of important points can be made by look- series in each voxel. These are known as “predictors” or
ing at these time series. First, notice that the time series “regressors” and each must have the same length as the
in neighboring voxels is not identical. Even though they time series in each voxel.
are only separated by 3 mm, the voxel in the top left The regressors are placed into two groups: one is
has a relatively small signal change and the voxel in the the “baseline” group, which can be considered as the
middle has a very large signal change (on the order of “null hypothesis”: what should the time series look like
10%). This demonstrates that the BOLD fMRI signal is in a voxel in which there is no neural activity related
not “all or nothing.” The amount of BOLD signal change to the stimulus. In Fig. 3–6A, a single baseline regressor
in a voxel is thought to be proportional to the summed is shown, consisting of a flat line. The null hypothesis
neuronal activity of all of the neurons in that voxel (on in this case is that the brightness of the voxel should
the order of a million neurons if the voxel contains en- not change over time, because there is change in voxel
tirely gray matter). The voxel in the upper left may have intensity due to blood oxygenation changes. In a real
fewer neurons, or it may have neurons that do not re- fMRI experiment, a number of other baseline regressors
spond as strongly to the visual stimulus as the voxel would also be used. For instance, there are often slow
in the center of the display. Next, note that (even for fluctuations in intensity due to respiration. This can be
the center voxel) the height of each peak varies. Be- modeled as slow changes in voxel intensity. Or, the sub-
cause each peak represents presentation of a different ject’s head movement can be estimated. This means that
segment of the movie, the different peaks are a measure an inactive voxel might be predicted to have some in-
of how strongly the neurons in the voxel responded to tensity fluctuations related to head motion.
that particular segment of the movie. Note that the third The second set of regressors represent the “exper-
peak is the highest in the center voxel, and in the other imental hypothesis.” These are the predictions about
voxels as well, suggesting that all of the neurons in this what should happen in voxels in which there is neu-
neighborhood preferred the third movie segment. ral activity related to the stimulus or task. In Fig. 3–6B,
Next, notice that the troughs of the signal are not a single experimental regressor is shown, representing
identical, even though the subject viewed the exact same a sine wave of the same frequency of the visual stim-
visual stimulus (a blank screen) in each trough. This sug- ulus (on and off 8 times over the course of 5-minute
gests that the variation in the signal intensity is not de- run). Voxels containing neurons responding to the vi-
termined solely by the neuronal activity in the voxel. sual stimulus would be predicted to get brighter and
This can be seen more clearly by examining the time darker as the visual stimulus turns on and off. In a real
series from voxels in the ventricle and adjacent white fMRI experiment, we might have multiple experimental
matter. No periodic variation of the signal intensity re- regressors. If two different types of visual stimuli were
lated to visual stimulation is observed. However, the sig- shown (such as faces and houses), we would have two
nal does have peaks and dips. These variations in signal separate regressors, each representing the amplitude of
intensity have a number of sources, including respira- the response to each type of stimulus.
tion, which introduces changes in tissue oxygenation; The next step is to fit all of the baseline regressors
the cardiac cycle, which can introduce changes in tissue to the voxel time series. For a sample ventricle voxel
oxygenation; gross movement of the brain because of time series, this is shown in Figs. 3–6C and D. The
pulsations or subject head movements; and scanner or flat baseline regressor is fit to the time series, and the
38 SECTION I TECHNIQUES

0 sec 300 sec 0 sec 300 sec


A B

Baseline error - Full error


Full error

Baseline error = 134 Full error = 123

C D

Baseline error - Full error


Full error

Baseline error = 377 Full error = 0.4

E F

Figure 3–6. (A) Baseline (null-hypothesis) regressor consisting of a flat line,


equivalent to constant intensity in the voxel over time. The y-axis is arbitrary, since the
waveform fits separately to each voxel. (B) Experimental regressor consisting of a
sinusoidal waveform. Black lines under x-axis show the time when each visual
stimulus is on screen. The voxel intensity is expected to increase when the stimulus is
on screen and decrease when it is not. (C) The black line shows the actual MR time
series from a voxel in a ventricle over the course of a 5-minute MR scan series. The
red line shows the best-fit baseline regressor from (A). The baseline error is the
difference between the black and red lines. (D) The red line shows the best-fit
combination of the baseline regressor in (A) and the experimental regressor in (B) (full
statistical model). The full error is the difference between the black and red lines. (E)
The black line shows the actual MR time series from a voxel in visual cortex over the
course of a 5-minute MR scan series. The red line shows the best-fit baseline
regressor from (A). The baseline error is the difference between the black and red
lines (large error). (F) The red line shows the best-fit combination of the baseline
regressor in (A) and the experimental regressor in (B) (full statistical model). There is
a good match between the lines, resulting in low error.

error (difference between best fit and actual data) is cal- is correct, the key question is how much extra variance
culated. Next, the combination of baseline regressors is accounted for by the experimental regressors. To do
and experimental regressors are fit to the voxel time this, we calculate an F -ratio, the ratio of the explained
series and error is again calculated. This error will al- variance to unexplained variance. In this case, the
ways be less, because fitting additional curves always explained variance is the extra variance accounted for
fits better. To determine if the experimental hypothesis by the experimental regressors (full error – baseline
CHAPTER 3 FUNCTIONAL MRI FOR CEREBRAL LOCALIZATION 39

error) divided by the total error. In Figs. 3–6C and D, Another popular preprocessing step is spatial
this extra variance is very small, leading to a small F - smoothing. If each voxel contains noise from physio-
ratio (0.14). To calculate statistical significance, we can logical or scanner sources, averaging data across sev-
examine a table that requires two values, the numerator eral voxels can reduce the noise. The averaging is usu-
and denominator degrees of freedom. The numerator ally performed only within images acquired at the same
degrees of freedom specifies how many experimental time (spatial smoothing) as opposed to images acquired
regressors are used (as more regressors are used, more across time (temporal smoothing).
variance is accounted for by chance) and the denomina- Both motion correction and spatial smoothing re-
tor degrees of freedom is approximately the number of duce the error in the full model fit, increasing the re-
points in the MR time series (as this number increases, sulting F -statistic and meaning that more voxels pass
power increases as well, meaning that it is important to the significance threshold. There is no single statistical
always collect as much fMRI data as possible). threshold that is correct for all patients and experiments.
The same process for an active voxel is shown in It is important to examine fMRI data using a range of
Figs. 3–6E and F. The baseline error is large because the statistical thresholds, or completely unthresholded, as in
stimulus-evoked fluctuations in the MR signal are not the Fig. 3–7, in order to get a clear understanding of the
well fit by the baseline straight-line function. In contrast, data.
the full model error is low, because the sinusoidal ex- fMRI activations can be considered as a virtual
perimental regressor fits the MR intensity very well. This “mountain range.” At a very high threshold, only the
results in a very large F -statistic. very tops of the highest mountain peaks will be visible.
This process of model fitting is repeated for every As the threshold is lowered, the peaks will enlarge
voxel in the brain. as the flanks of the tall mountains emerge above the
threshold, and the tops of smaller activation mountains
will also be visible.
䉴 CREATING ACTIVATION MAPS This effect is shown in Fig. 3–7. At very high thresh-
olds, only voxels in early visual cortex (V1/V2) are ac-
To construct an activation map, the range of F -values is tive; as the threshold is lower, voxels in other regions of
mapped to a color scale. In the Fig. 3–7, low F -values extrastriate cortex become active. In order to determine
are assigned a green color, and higher F -values are as- whether activations are “real,” or represent false posi-
signed yellow to red colors. Then, each voxel is colored tives, it is important to examine the time series from the
in according the F -value calculated for it. Figure 3–7B voxels in question.
shows the result of this process. Most of the brain is In this example, we focussed only on a simple two-
green (meaning low F -rations) except for voxels in oc- condition experiment, in which the subject alternately
cipital lobe, which have yellow to red colors. In order views a moving visual stimulus and a blank fixation
to make the binary judgement of which voxels are “ac- screen. However, the exact same analysis procedures
tive” or not, a statistical threshold must be chosen. In this can be used with much more complex designs. For
example, an F -statistic of ten corresponds to a chance instance, we could alternate between viewing a visual
probability of p < 10–7 . If there are 105 voxels in the stimulus, hearing an auditory stimulus, and viewing a
brain volume, this means the odds of a single voxel be- blank screen (and hearing nothing). In this experiment,
ing active by chance is p < 0.01 using the conservative the full model would contain two different predictor
Bonferroni correction for multiple comparisons. If we functions, one for the visual stimulus and one for the
display only voxels that have an F -value greater than auditory stimulus. Activation maps could then be cre-
15, we see mainly voxels in the occipital lobe. ated for both functions, either functions, or the contrast
In order to be more sensitive to weak activations, between auditory and visual functions. This process can
other processing steps are often performed before the be repeated indefinitely, for instance, ten different types
F -ratio is calculated for every voxel. An important pre- of visual stimuli. A predictor function is created for each
processing step is motion correction. Gross head motion predictor functions, and the full model fit to each voxel’s
can occur as the head settles into the foam padding of time series. Activation maps can be created to show vox-
the MR head coil, or the body gradually relaxes, over els responding to any stimulus (full-F ) or any combina-
the course of the scan session. By comparing each vol- tion of individual stimuli.
ume to the initial volume (or the mean of all volumes),
these slow motions can be corrected fairly well. Quick
motions, such as those that occur during a sneeze are
difficult to correct. Therefore, it is important to explain 䉴 VISUALIZATION OF ACTIVATION
to the patient the necessity of holding the head still and ON THE CORTICAL SURFACE
attempt to restrict head motion with padding or commer-
cial products (such as foam beads) while still maintaining Because the T2* EPI images used to create fMRI acti-
patient comfort. vation maps are relatively low resolution, they are not
40 SECTION I TECHNIQUES

A C
B

D E

F G

Figure 3–7. Visualization of active brain areas. (A) Original T2∗ brain image. (B) Brain
image with each voxel colored by the significance of the activation (calculated as in
Fig. 3–6). Green is low significance and red is high significance (F-values shown by
color scale). (C) Combination of (A) and (B): active voxels above a certain threshold
(F > 10) are colored; all other values are from original brain image. (D) Map created
with a more liberal threshold (F > 5). (E) Map created with a stricter threshold
(F > 15). (F) Thresholded voxels (F > 10) interpolated and overlaid on a high-resolution
T1 image from the same subject. (G) Enlargement of visual cortex region in (F).
Colored voxels are made transparent to allow visualization of anatomical structure
and functional activation. Notice correspondence between gray matter in T1 image
and significant (colored) voxels.
CHAPTER 3 FUNCTIONAL MRI FOR CEREBRAL LOCALIZATION 41

ideal for clinical decision-making. The typical strategy is The surface model can also be rotated to get dif-
to collect a high-resolution T1 anatomy in the same pa- ferent views of how it would look like in various clini-
tient during the same scanning session. Then, the active cal situations, for instance when exposed during a cra-
voxels calculated from the EPI images are overlaid on niotomy.
this high-resolution T1 anatomy (see Fig. 3–8). Because of the many complex steps required to
There are two problems inherent in this approach. analyze and visualize fMRI data, analysis is typically
First, the subject may move between collection of the conducted with specialized software packages. Many
T1 anatomy and the functional EPI images. By aligning of these packages, including AFNI15 and SPM,9 are
(motion correcting) the EPIs relative to the T1, this can freely available and include sample datasets, providing
be compensated for. Second, EPI images are distorted an easily accessible way to become familiar with fMRI
relative to the true brain anatomy pictured in T1 images data.
because of magnetic susceptibility and other artifacts.
This means that even without any patient motion, there
can be shifts of many millimeters between the EPI and
the T1. 䉴 DRAWING CLINICAL
Therefore, even if activations maps are viewed over- INFERENCES FROM fMRI
laid on a T1, it is critical to also visualize them overlaid STUDIES
on the original EPI to ascertain the true location of acti-
vation. In order to draw clinical inferences from fMRI studies, we
Additional processing steps are often performed to must make a number of inferences from our fMRI activa-
make the T1 anatomy more visualization friendly. These tion maps. The first inference is that active voxels reflect
include skull stripping (removing the skull) and normal- activity in neurons located inside the voxel. How justified
izing the brain so that it is in a standard space. This is this assumption? There is good evidence from experi-
means that regardless of the patient’s position in the ments in animal models that changes in blood oxygena-
scanner, the brain will have a consistent alignment with a tion are tightly coupled to local neuronal activity.16,17
template. If the template has labels attached, for instance This coupling is made possible because of the dense
the location of Brodmann areas or anatomical structures, network of capillaries in cortex (see Fig. 3–3).18 Individ-
these labels can be applied to the individual subject brain ual capillaries appear to open and close with metabolic
to make identification of different anatomical structures demand, producing a spatially localized fMRI BOLD sig-
easier. nal. This is seen in fMRI studies, which image cortical
Another useful tool for visualizing MRI data are ocular dominance columns (on the order of ∼1 mm
three-dimensional (3D) models of the cortical surface. in size) in human primary visual cortex. These maps
As shown in the Fig. 3–8, computerized tools are avail- are reproducible within the same scanning session and
able to automatically segment the MR image into con- across different scanning sessions on different days.19 In
tours tracing the boundary between gray and white mat- diseases in which the vasculature is impaired, such as
ter and the boundary between gray matter and CSF stroke or tumor, the spatial localization of the BOLD sig-
(pial surface of the brain). After creation of these con- nal may also be impaired. In general, it is important to
tours, a surface tessellation is created consisting of a obtain converging evidence for neural organization ob-
mesh of thousands of triangles (middle panel and en- served with fMRI. An example of converging evidence
largement). This tessellation is physically equivalent to is shown in Fig. 3–9.
the cortical surface, which also consists of a single With high-resolution fMRI, it was shown that re-
sheet of tissue, folded to fit within the confines of the gions of human superior temporal sulcus contains segre-
skull. gated, patchy regions that respond to auditory stimula-
When viewed in 2D slices, it can be difficult to iden- tion, such as speech, and visual stimulation, such as face
tify specific sulci or gyri. When viewed as a whole, it is movements, or both. However, this conclusion is tem-
easy to identify a specific sulcus, such as the superior pered by the knowledge that fMRI is an indirect measure
temporal sulcus, at any location along its anterior to pos- of neural activity. Therefore, critical additional evidence
terior extent. Most of the cortex is buried in the sulcal is provided by two additional methods, anatomical in-
folds. With a cortical surface model, the links between formation from tracer injections in macaque monkeys;20
nodes can be relaxed, allowing the surface to inflate like and direct recordings of neural responses using pene-
a balloon. This reveals tissue previously hidden in the trating microelectrodes in monkeys.21 Both of these very
sulci. fMRI activations can be visualized on the cortical different techniques showed similar evidence of patchy
surface (with the same caveat as overlaying them on the organization, suggesting that the organization observed
T1). This has the enormous advantage of allowing view- with fMRI is real, and not an artifact of differential vascu-
ing of all active regions in the brain in a single glance, larization, experimental design, stimulus set, or analysis
instead of sorting through stacks of images. method.
42 SECTION I TECHNIQUES

A B C

F
D E

G H

I J

Figure 3–8. Structural MRI analysis stream for functional MRI. (A) Midsagittal section
of T1-weighted anatomical scan. (B) Skull-stripped and intensity normalized brain. (C)
Automated tracing of cortical gray matter and CSF boundary (pink line) and cortical
gray matter and white matter boundary (yellow line). Enlargement of blue area shown
in (A). (D) Cortical surface model, rendered as surface. (E) Cortical surface model
rendered to show individual mesh elements. (F) Enlargement showing individual
triangular elements (face sets) of cortical surface mesh. (G) Functional activation
(thresholdF> 15) mapped to cortical surface as colored regions. (H) Lateral view of
cortical surface with activation. (I) Inflated cortical surface model (medial view). (J)
Inflated cortical surface model (lateral view).
CHAPTER 3 FUNCTIONAL MRI FOR CEREBRAL LOCALIZATION 43

B
A C

Figure 3–9. Converging evidence on neural organization from functional MRI (fMRI)
and other methods. (A) Anatomical data set showing the superior temporal sulcus
(STS) of the macaque monkey. (Adapted from Seltzer B, Cola MG, Gutierrez C,
Massee M, Weldon C, Cusick CG. Overlapping and nonoverlapping cortical
projections to cortex of the superior temporal sulcus in the rhesus monkey: double
anterograde tracer studies. J Comp Neurol 1996;370(2):173–190.) Tracer injection of
anterograde tracer into macaque monkey auditory cortex and visual cortex. Blue
regions of STS receive input from auditory cortex. Orange regions receive input form
visual cortex. Note the patchy organization, with some regions receiving auditory
input, some regions receiving visual input. (B) fMRI data demonstrating patchy
organization of human STS, with interleaved regions responding to auditory
stimulation (blue), visual stimulation (orange), or both (green). (Adapted from
Beauchamp MS, Argall BD, Bodurka J, Duyn JH, Martin A. Unraveling multisensory
integration: patchy organization within human STS multisensory cortex. Nat Neurosci
2004;7(11):1190–1192.) (C) Single-unit recording from macaque STS showing uneven
patchy distribution of neurons responding to auditory stimulation (blue), visual
stimulation (orange), or both (green). (Adapted from Dahl CD, Logothetis NK, Kayser
C. Spatial organization of multisensory responses in temporal association cortex. J
Neurosci 2009;29(38):11924–11932.)

A second inference is that the region of activity brain regions. Statistical criteria or a priori criterion are
is necessary for the cognitive function being studied. applied to the fMRI data in order to classify a subset
This problem is particularly acute in studies of language, of these active regions as being specifically involved in
memory, and other complex cognitive functions. In a particular elements of the cognitive task. For instance,
typical cognitive study (such as picture naming) in which if the subjects’ task is to press a button in response
subjects are presented with stimuli, make cognitive de- to an auditory tone, activations on the superior tem-
cisions about them, and then produce a motor response, poral gyrus would be classified as due to the auditory
activations would be expected in unisensory regions re- tone, whereas activations along the central sulcus would
sponding to the sensory stimulus, multisensory regions be attributed to the motor response and associated so-
that integrate across modalities, cognitive regions that matosensory feedback.22 However, even in this simple
are important for decision-making, and response selec- task, other brain activations are not so clear cut: sec-
tion and motor regions that produce the behavioral out- ondary somatosensory cortex in the parietal operculum,
put. Even without an explicit behavioral task, (such as which might be active in response to somatosensory
viewing, but not naming, pictures) subjects may perform feedback from the button press, is adjacent to auditory
language and memory operations when presented with association areas, which might also be active depending
a stimulus (and without a task there is no measure of on the nature of the auditory tone, with no clear bound-
subjects’ alertness or attention, or the amount of pro- ary between them. A further complicating factor is brain
cessing performed on each stimulus). With or without reorganization or anatomical distortion due to injury or
a task, fMRI experiments typically find activity in many tumor.23
44 SECTION I TECHNIQUES

Finally, BOLD fMRI depends on patient compliance. ping with magnetic resonance imaging. Proc Natl Acad Sci
For instance, normal volunteers will reliably tap their fin- USA 1992;89(13):5951-5955.
gers and keep their head still when instructed to, result- 9. Friston KJ, Frith CD, Liddle PF, Frackowiak RS. Compar-
ing in excellent maps of motor cortex. Patients who do ing functional (PET) images: the assessment of significant
not understand instructions, or are unwilling to comply, change. J Cereb Blood Flow Metab 1991;11(4):690-699.
10. Bandettini PA, Jesmanowicz A, Wong EC, Hyde JS. Process-
will produce poor quality or uninterpretable activation
ing strategies for time-course data sets in functional MRI of
maps. This is one reason that it is important to collect be- the human brain. Magn Reson Med 1993;30(2):161-173.
havioral data from subjects while they are in the scanner. 11. Worsley KJ, Friston KJ. Analysis of fMRI time-series
For instance, if there is little activation in motor cortex revisited–again. Neuroimage 1995;2(3):173-181.
during a finger-tapping task, is it because motor cortex 12. Bowerman BL, O’Connell RT. Linear Statistical Models: An
is impaired? Or, because the subject was asleep in the Applied Approach. Boston: PWS-Kent Pub. Co., 1990, pp.
scanner and was not performing the task? If an objective xvi-1024.
measure, such as the number of finger taps per minute, 13. Neter J, Wasserman W, Kutner MH. Applied Linear Statis-
is available, this can answer the question. tical Models: Regression, Analysis of Variance, and Experi-
In summary, fMRI provides the most powerful tech- mental Designs. Homewood, IL: Irwin, 1990, pp. xvi-1181.
nique currently available to measure human brain func- 14. Rencher AC. Methods of Multivariate Analysis. New York:
Wiley, 1995.
tion. Rapid advances continue more than 20 years after
15. Cox RW. AFNI: software for analysis and visualization
the basic principles were first demonstrated, suggesting of functional magnetic resonance neuroimages. Comput
a bright future for the technique. Biomed Res 1996;29:162-173.
16. Logothetis NK. The neural basis of the blood-oxygen-level-
dependent functional magnetic resonance imaging signal.
REFERENCES Philos Trans R Soc Lond B Biol Sci 2002;357(1424):1003-
1037.
1. Lauterbur PC. Image formation by induced local interac- 17. Lee JH, Durand R, Gradinaru V, et al. Global and local fMRI
tions: examples employing nuclear magnetic resonance. signals driven by neurons defined optogenetically by type
Nature 1973;242:190-191. and wiring. Nature 2010;465(7299):788-792.
2. Lauterbur PC. Progress in n.m.r. zeugmatography imaging. 18. Harrison RV, Harel N, Panesar J, Mount RJ. Blood capillary
Philos Trans R Soc Lond B Biol Sci 1980;289(1037):483- distribution correlates with hemodynamic-based functional
487. imaging in cerebral cortex. Cereb Cortex 2002;12(3):225-
3. Ogawa S, Lee TM. Magnetic resonance imaging of blood 233.
vessels at high fields: in vivo and in vitro measurements 19. Cheng K, Waggoner RA, Tanaka K. Human ocular domi-
and image simulation. Magn Reson Med 1990;16(1):9-18. nance columns as revealed by high-field functional mag-
4. Ogawa S, Lee TM, Kay AR, Tank DW. Brain magnetic res- netic resonance imaging. Neuron 2001;32(2):359-374.
onance imaging with contrast dependent on blood oxy- 20. Seltzer B, Cola MG, Gutierrez C, Massee M, Weldon C, Cu-
genation. Proc Natl Acad Sci USA 1990;87(24):9868-9872. sick CG. Overlapping and nonoverlapping cortical projec-
5. Ogawa S, Lee TM, Nayak AS, Glynn P. Oxygenation- tions to cortex of the superior temporal sulcus in the rhesus
sensitive contrast in magnetic resonance image of ro- monkey: double anterograde tracer studies. J Comp Neurol
dent brain at high magnetic fields. Magn Reson Med 1996;370(2):173-190.
1990;14(1):68-78. 21. Dahl CD, Logothetis NK, Kayser C. Spatial organization of
6. Bandettini PA, Wong EC, Hinks RS, Tikofsky RS, Hyde JS. multisensory responses in temporal association cortex. J
Time course EPI of human brain function during task acti- Neurosci 2009;29(38):11924-11932.
vation. Magn Reson Med 1992;25(2):390-397. 22. Beauchamp MS, Lee KE, Argall BD, Martin A. Integration of
7. Kwong KK, Belliveau JW, Chesler DA, et al. Dynamic auditory and visual information about objects in superior
magnetic resonance imaging of human brain activity dur- temporal sulcus. Neuron 2004;41(5):809-823.
ing primary sensory stimulation. Proc Natl Acad Sci USA 23. Ulmer JL, Hacein-Bey L, Mathews VP, et al. Lesion-induced
1992;89(12):5675-5679. pseudo-dominance at functional magnetic resonance imag-
8. Ogawa S, Tank DW, Menon R, et al. Intrinsic signal changes ing: implications for preoperative assessments. Neuro-
accompanying sensory stimulation: functional brain map- surgery 2004;55(3):569-579; discussion 580-581.
Chapter 4
Functional MRI: Application to
Clinical Practice in Surgical Planning
and Intraoperative Guidance
Michael Schulder 1 and Robin Wellington 2
1
Department of Neurosurgery, North Shore-Long Island Jewish Health System, Manhasset, New York
2
Department of Psychology, St. John’s University, Flushing, New York

䉴 INTRODUCTION This step includes motion correction, statistical analysis,


and registration of the analyzed data to anatomical im-
Since functional magnetic resonance imaging (fMRI) was ages. The aim is to identify voxels with activation levels
described in 1991 by Belliveau et al.,1 this noninvasive that are (statistically) significant in activity versus rest
method of mapping the brain has generated a veritable states. The ultimate appearance of the maps will de-
industry of research and clinical applications. A PubMed pend to a large part on statistical thresholding. Clinicians
search for fMRI yields many thousands of references. The who plan on using the data must appreciate this, and
majority of this work has been done by cognitive neu- should be prepared to take an active role in the analysis.
roscientists and psychologists using fMRI to learn more For instance, they may accept a significance level greater
about how memories, emotions, and behaviors are pro- than 0.05 if the result is a useful visual presentation of
cessed and generated by the brain.2−5 It has also become a particular functional volume; conversely, the clinician
a valuable method of assessing patients with movement may insist on a lower threshold in order to eliminate
disorders, especially Parkinson’s disease.6,7 irrelevant areas of activation.
Neurosurgeons’ interest in fMRI has been mainly in fMRI can be viewed for surgical planning as a stand-
its use for surgical planning and intraoperative guidance, alone image, on hard copy or on a picture archiving
which will be the subject of this chapter. and communication system. This is adequate for surgical
planning, but ideally for intraoperative use, fMRI should
be registered for neuronavigation (see later).10
䉴 TECHNIQUE OF fMRI

Clinicians seeking to use fMRI must remember that 䉴 PARADIGMS IN fMRI


the activation maps generated by this modality are not
“images” per se, but rather statistical maps. These maps When requesting an fMRI study, the clinician should
are generated by statistical analyses that compare “rest” understand the tasks, or paradigms, that patients per-
and “activity” states. Practically speaking, patient coop- form for data acquisition. A “robust” paradigm is one
eration is needed to acquire the data for these analyses. that reliably supplies an accurate activation map that is
The subjects need to perform the appropriate task and easy to interpret. The tasks are relatively simple and easy
to lie still for what can be a relatively prolonged time for the patient to accomplish. Thus, mapping of the pri-
in the closed MRI bore.8 It may be possible to give sen- mary motor cortex via finger tapping, or of the visual
sory and visual stimuli to anesthetized patients but this cortex by presentation of patterns, usually provide large
would not work for other areas of functionally important areas of activation at a high significance threshold.11,12
cortex. Light touch sensation can be used to identify primary
Since described by Ogawa, blood oxygen level de- sensory cortex, although this area’s location can be in-
pendent (BOLD) fMRI has been most commonly ac- ferred from that of the motor cortex immediately across
quired because of its completely noninvasive nature.9 the central sulcus.13,14 It is somewhat intuitive that these
Most often, fMRI data are processed after acquisition in functions would and do yield the most robust fMRI
a laboratory separate from the site of data acquisition. results. Moving beyond these, the complexity of data

45
46 SECTION I TECHNIQUES

A B

Figure 4–1. Diffusion-weighted images of language functional magnetic resonance


imaging in a 42-year-old woman with a right parietal high-grade glioma. (A) Silent
word generation to map expressive language area in left hemisphere. (B) Text
comprehension to identify receptive language showing activation in both
hemispheres.

acquisition and the reliability of the data become increas- sections of lesions that on anatomical images appear to
ingly questionable. be in or near eloquent areas may be changed to less
A specific and sensitive means of noninvasively invasive treatments. Conversely, surgery thought to be
mapping language areas is highly desirable. Paradigms prohibitively dangerous can be planned with reasonable
for assessment of expressive and receptive language assurance if functional areas are seen with fMRI to be at
continue to be modified.15,16 fMRI is a reliable means a safe distance from the lesion. Stereotactic approaches
of identifying the hemisphere dominant for language, at likewise can be planned to avoid eloquent areas (Fig.
least in right-handed patients who clearly have left-sided 4–2). While fMRI has played an important role in defin-
language activation (Fig. 4–1).17,18 Still, any question at ing targets for surgical treatment of movement disorders
all regarding dominance in patients whose impending or psychiatric disorders,22,23 these data do not have the
surgery may put language function at risk should be spatial precision required for targeting based on the ac-
evaluated with a Wada test (see Chapter 6) or have their tivation itself. As noted above, if language areas are at
surgery performed with an awake language mapping.19 risk in surgery, then awake mapping should be done to
The use of fMRI to identify areas of memory, cogni- avoid new deficits.
tive function, emotion, and so forth remains primarily in If fMRI is to be used for surgical decision-making,
the domain of neuroscientific investigation.20,21 If preop- the surgeon should have at least a basic understanding
erative patients will tolerate a prolonged fMRI acquisi- of acquisition and analysis. The surgeon should be in-
tion session, it may be worthwhile to attempt imaging of volved with image interpretation and should confirm that
these functions, but surgical planning decisions cannot the visible activation maps are based on valid statistical
be made on their basis at this stage of fMRI understand- analysis.
ing (see Chapter 18).

SURGICAL NAVIGATION
䉴 NEUROSURGICAL APPLICATIONS
OF fMRI To fully take advantage of fMRI as an adjunct to intracra-
nial surgery, the data should be available in the operating
SURGICAL PLANNING room in an interactive fashion. Studies have confirmed
the possibility of registering fMRI on a surgical naviga-
fMRI data are most often used in neurosurgical planning tion platform, and by so doing confirming the stereotac-
as a means of assessing the likely risks of surgery. Re- tic accuracy of the data. Schulder et al. registered motor
CHAPTER 4 FUNCTIONAL MRI 47

Figure 4–2. Motor functional magnetic resonance imaging (fMRI) in a 46-year-old


woman with a right frontal ganglioglioma. Activation was posterior to the tumor,
allowing for complete resection. Surgery was done with intraoperative MRI guidance,
and based on the fMRI, an anterior to posterior approach was used to reach the
tumor.

fMRI from a finger tapping paradigm and compared the can be registered to intraoperative MRI (iMRI) for naviga-
predicted center of activation with the location of hand tional use (Fig. 4–3). Anatomical images can be updated
motion by bipolar cortical stimulation or phase reversal with iMRI, but, of course, this in itself will not compen-
of somatosensory evoked potentials.10 sate for brain shift affecting fMRI-based navigation.
Other studies have compared the accuracy of fMRI
to positron emission tomography24,25 and magnetic
source imaging26 and shown a high concordance be- BRAIN TUMORS
tween the various methods. These reports have estab-
lished that fMRI, if properly registered to a stereotactic The goal of brain tumor surgery is to maximize resec-
anatomical image, can be relied upon to indicate the tion while preserving function. fMRI can play a vital
location of eloquent cortex. Like all navigation based role toward this end. This is most obvious for patients
on preoperative datasets, however, the surgeon must with gliomas. These intraaxial tumors may infiltrate ar-
remain alert to the reality of brain shift. Loss of cere- eas of eloquent cortex even while function is preserved,
brospinal fluid after dural opening, and lesion removal it- especially in patients with low-grade tumors.29 fMRI
self, may quickly render navigation inaccurate.27,28 fMRI may steer the surgeon away from resection as a result
48 SECTION I TECHNIQUES

Figure 4–3. A 64-year-old woman with recurrent parasagittal meningiomas. (A)


Image fusion of preoperative motor functional magnetic resonance imaging (fMRI)
and anatomical contrast-enhanced anatomical image acquired in the operating room
with intaroperative MRI. (B) Surgical navigation with registered fMRI. (continued)

(Fig. 4–4). At the same time, by confirming that the tu- ARTERIOVENOUS MALFORMATIONS
mor is separate from critical function, fMRI can provide
the basis for a planned gross resection (Fig. 4–2). Hall fMRI in patients with arteriovenous malformations
and colleagues’ use of fMRI allowed for complete resec- (AVMs) poses a special challenge. The BOLD method is
tion, with minimal morbidity, in two-thirds of patients based on minute differences in blood flow between task
with tumors in or near the motor cortex.30 and rest states. Changes in regional perfusion because of
fMRI need not be restricted to use in patients with the AVM can, therefore, interfere with fMRI acquisition.31
gliomas. Metastatic tumors displace brain but may often
be adjacent or deep to eloquent cortex, making fMRI
a useful adjunct for surgical planning and navigation. EPILEPSY
Likewise, meningiomas are extra-axial tumors, and as
long as the surgeon respects the border between tumor Resection of epileptic foci in the temporal lobe and else-
capsule and brain, no deficit will occur. However, in where may involve removal of functionally viable brain.
practice, even histologically benign meningiomas may Surgery in the dominant temporal lobe in particular
invade the adjacent pia, and knowing the relationship has been studied in patients who underwent resections
between tumor and eloquent brain will aid in surgical for medically refractory seizures.32 fMRI may be useful
planning (Fig. 4–5). in certain patients as a way of establishing language
CHAPTER 4 FUNCTIONAL MRI 49

Figure 4–3. (Continued)

Figure 4–4. Motor functional magnetic resonance Figure 4–5. Motor functional magnetic resonance
imaging in a 49-year-old woman with a low-grade imaging in a 25-year-old woman with a left
glioma, seen as gyral fullness just anterior to the main parasagittal meningiomas, seen here to be just
site of activation in the right frontal lobe. anteromedial to the left primary motor cortex.
50 SECTION I TECHNIQUES

dominance in the hemisphere contralateral to the recurrent parasagittal meningiomas, showing how fMRI
planned resection, but as noted, is not adequate, at can be used to identify the motor cortex and measure
present, for precise spatial localization of the main areas the amount of radiation it receives.
controlling expressive and receptive speech. If language
fMRI is acquired in such patients, it should be supple-
mented with intraoperative cortical mapping 䉴 PITFALLS OF fMRI

The accuracy and reliability of fMRI may be adversely


affected by various factors. Among these are patient
STEREOTACTIC RADIOSURGERY movement, inadequate cooperation in task performance
and alternating rest cycles, and statistical manipulation
fMRI offers the possibility of more precisely defining (Fig. 4–7). Increasing or decreasing the threshold of sta-
the safe limits of stereotactic radiosurgery (SRS) when tistical significance may result in under- or overestimat-
applied to targets in or near cerebral cortex.33 Given ing the volume of functional cortex, respectively. Like-
the location of many SRS targets that may be within wise, the presence of adjacent tumor mass or edema may
the cerebellar hemispheres or elsewhere in the poste- decrease the BOLD signal changes, presumably because
rior fossa, fMRI of brainstem nuclei may be of use in of impaired regional blood flow.37 As with any other pre-
certain patients.34 Identification of “eloquent” areas can operative dataset, brain shift will invalidate the spatial
be used to direct excess dose toward less critical lo- accuracy of fMRI, making reliance on surgical naviga-
cations. If routinely used, fMRI for SRS could provide tion potentially hazardous.27 Of course, as a task-based
useful information about the tolerance of functionally method, fMRI cannot be done if the patient has a rele-
important brain to various dose levels of irradiation given vant deficit (e.g., aphasia not allowing for language map-
in single or several sessions. These limits are fairly well ping). This does not necessarily mean that the function
understood for most cranial nerves, but we are still in is irretrievably lost, so surgical plans should not rely on
the early stages of understanding the dose limits of SRS the absence of activation in such patients.
for specific areas of brain function.35,36 Figure 4–6 is a fMRI is a method for identifying areas of functionally
radiosurgical dose plan in a 64-year-old woman with a important gray matter. Lack of attention to the relevant
white matter tracts can lead to new neurological deficits
despite the most careful imaging and avoidance of cortex
or nuclei. Use of diffusion tensor imaging tractography,
possibly in conjunction with subcortical mapping, can
help to avoid such complications.38,39
There is often a certain circularity of reasoning ap-
plied to fMRI. For instance, motor area activation looks
accurate “because that’s where the motor cortex is sup-
posed to be.” The surgeon should avoid being caught
up in this dubious logic and if any question remains re-
garding the safety of relying on fMRI alone, the surgeon
should be prepared to perform intraoperative cortical
mapping.40,41

䉴 FUTURE DIRECTIONS

fMRI has not quite fulfilled its promise as a routine clin-


ical tool. Acquisition and analysis still requires some
degree of specialized expertise. This should change
with the availability of standardized paradigms and
systems that record patient activity, in conjunction
with services that will perform the statistical analysis
Figure 4–6. Radiosurgical dose plan. The (http://www.neurognostics.com/). The goal of such sys-
prescription dose is delivered to the meningiomas tems is to make fMRI as routine as anatomical imaging.
target (yellow). Functional magnetic resonance
imaging activation of the left primary motor cortex is
One obstacle to further acceptance of fMRI is the
shaded blue, and isodose lines indicate that a delay between scanning and the availability of the acti-
relatively small dose of radiation is received by that vation maps. For patients who live some distance from
area. imaging facilities, and/or are having surgery too soon to
CHAPTER 4 FUNCTIONAL MRI 51

Figure 4–7. Attempted motor functional magnetic resonance imaging, registered for
surgical navigation, in a 56-year-old man with a single metastatic tumor adjacent to
the right primary motor cortex. Scattered random areas of activation resulted from
suboptimal patient task performance.

allow for imaging and data analysis, fMRI is impractical. sis will be a reality, another requirement for fMRI in the
The ability to acquire valid real-time fMRI will avoid this operating room). Exceptions to this might be sensory or
problem. Preliminary results indicate that data so gener- visual stimulation, which can be presented to patients
ated are as accurate as those done with “conventional” under anesthesia. Benefits of intraoperative fMRI could
methods.42,43 include compensation for brain shift and avoidance of
It may be possible to bring fMRI into the operat- prolonged preoperative data acquisitions.47,48 Still, in-
ing room. Despite concerns regarding chemical shifts vestigators pursuing this option will have to prove a clear
and other sources of decreased signal, the feasibility of advantage to it over other means of updating informa-
acquiring fMRI even with a low-field strength (0.15 T) tion during surgery.
intraoperative magnet has been demonstrated.44 Other With fMRI, as with other technological advances in
studies have described intraoperative fMRI with high- neurosurgery, the question arises: how much is really
field strength magnets45,46 The main obstacle to prac- necessary? And how much does it benefit the patient
tical implementation of this technique is the need for in terms of improved outcome or at least greater ease,
activity and rest states—that is, as noted above, for most lower cost, and so forth? Data that definitively establish
paradigms, the patient has to be awake and cooperative fMRI, even for robust paradigms, as superior to cortical
for the acquisition (assuming that true real-time analy- stimulation are hard to come by. But we are still relatively
52 SECTION I TECHNIQUES

early in the fMRI era. As the speed of data processing im- parison with electrophysiological localization. J Neurosurg
proves and we get closer to reliable real-time fMRI that 1995;83(2):262-270.
can be acquired in nonspecialized settings, this technol- 14. Mueller WM, Yetkin FZ, Haughton VM. Functional mag-
ogy truly will move out of its purely scientific phase and netic resonance imaging of the somatosensory cortex. Neu-
become a routine clinical tool in neurosurgery. rosurg Clin N Am 1997;8(3):373-381.
15. Vingerhoets G, Deblaere K, Backes WH, et al. Lessons
for neuropsychology from functional MRI in patients with
epilepsy. Epilepsy Behav 2004;5(Suppl 1):S81-S89.
䉴 CONCLUSIONS 16. Harrington GS, Buonocore MH, Farias ST. Intrasubject re-
producibility of functional MR imaging activation in lan-
Ongoing refinements and acceptance of fMRI will con- guage tasks. AJNR Am J Neuroradiol 2006;27(4):938-944.
tinue to make it a regular part of 21st century intracranial 17. Balsamo LM, Gaillard WD. The utility of functional mag-
netic resonance imaging in epilepsy and language. Curr
neurosurgery. In the near future, it will be as routine as
Neurol Neurosci Rep 2002;2(2):142-149.
acquiring an anatomical study. Noninvasive functional
18. Sabbah P, Chassoux F, Leveque C, et al. Functional MR
mapping of the brain with fMRI will be seen as an indis- imaging in assessment of language dominance in epileptic
pensable neurosurgical tool. patients. Neuroimage 2003;18(2):460-467.
19. Danks RA, Rogers M, Aglio LS, Gugino LD, Black PM. Pa-
tient tolerance of craniotomy performed with the patient
REFERENCES under local anesthesia and monitored conscious sedation.
Neurosurgery 1998;42(1):28-34; discussion 34-36.
1. Belliveau J, Kennedy D, McKinstry R, et al. Functional map- 20. Meyer M, Zysset S, von Cramon DY, Alter K. Distinct fMRI
ping of the human visual cortex by magnetic resonance responses to laughter, speech, and sounds along the hu-
imaging. Science 1991;(254):716-719. man peri-sylvian cortex. Brain Res 2005;24(2):291-306.
2. Frackowiak RS. Forthergillian lecture. Imaging human 21. Powell HW, Duncan JS. Functional magnetic resonance
brain function. Trans Med Soc Lond 2000;117:53-63. imaging for assessment of language and memory in clinical
3. Gabrieli JD. Cognitive neuroscience of human memory. practice. Curr Opin Neurol 2005;18(2):161-166.
Annu Rev Psychol 1998;49:87-115. 22. Keightley ML, Winocur G, Graham SJ, Mayberg HS,
4. Li X, Lu ZL, Tjan BS, Dosher BA, Chu W. Blood oxygena- Hevenor SJ, Grady CL. An fMRI study investigating cog-
tion level-dependent contrast response functions identify nitive modulation of brain regions associated with emo-
mechanisms of covert attention in early visual areas. Proc tional processing of visual stimuli. Neuropsychologia
Natl Acad Sci U S A 2008;105(16):6202-6207. 2003;41(5):585-596.
5. Kennepohl S, Sziklas V, Garver KE, Wagner DD, 23. Zrinzo L. The role of imaging in the surgical treat-
Jones-Gotman M. Memory and the medial temporal ment of movement disorders. Neuroimaging Clin N Am
lobe: hemispheric specialization reconsidered. Neuroim- 2010;20(1):125-140.
age 2007;36(3):969-978. 24. Baumann SB, Noll DC, Kondziolka DS, et al. Comparison of
6. Au WL, Adams JR, Troiano A, Stoessl AJ. Neuroimaging in functional magnetic resonance imaging with positron emis-
Parkinson’s disease. J Neural Transm Suppl 2006(70):241- sion tomography and magnetoencephalography to iden-
248. tify the motor cortex in a patient with an arteriovenous
7. Jech R, Urgosik D, Tintera J, et al. Functional magnetic malformation. J Image Guid Surg 1995;1(4):191-197.
resonance imaging during deep brain stimulation: a pilot 25. Joliot M, Papathanassiou D, Mellet E, et al. FMRI and PET
study in four patients with Parkinson’s disease. Mov Disord of self-paced finger movement: comparison of intersubject
2001;16(6):1126-1132. stereotaxic averaged data. Neuroimage 1999;10(4):430-447.
8. Murphy KJ, Brunberg JA. Adult claustrophobia, anxiety and 26. McDonald JD, Chong BW, Lewine JD, et al. Integration of
sedation in MRI. Magn Reson Imaging 1997;15(1):51-54. preoperative and intraoperative functional brain mapping
9. Ogawa S, Lee TM, Kay AR, Tank DW. Brain magnetic res- in a frameless stereotactic environment for lesions near elo-
onance imaging with contrast dependent on blood oxy- quent cortex. Technical note. J Neurosurg 1999;90(3):591-
genation. Proc Natl Acad Sci USA 1990;87(24):9868-9872. 598.
10. Schulder M, Maldjian JA, Liu WC, et al. Functional image- 27. Dorward N, Alberti O, Velani B, et al. Postimaging
guided surgery of intracranial tumors located in or near the brain distortion: magnitude, correlates, and impact on
sensorimotor cortex. J Neurosurg 1998;89(3):412-418. neuronavigation. J Neurosurg 1998;88(4):656-662.
11. Fellner C, Schlaier J, Muller E, Fellner F. Functional MRI 28. Nabavi A, Black PM, Gering DT, et al. Serial intraoperative
of the motor cortex using a conventional gradient system: magnetic resonance imaging of brain shift. Neurosurgery
comparison of FLASH and EPI techniques. Magn Reson 2001;48(4):787-797; discussion 97-98.
Imaging 1998;16(10):1171-1180. 29. Atlas S, Howard RI, Maldjian J, et al. Functional MRI of re-
12. Spiegel J, Tintera J, Gawehn J, Stoeter P, Treede RD. Func- gional brain activity in patients with intracerebral gliomas:
tional MRI of human primary somatosensory and motor findings and implications for clinical management. Neuro-
cortex during median nerve stimulation. Clin Neurophys- surgery 1996;38:329-338.
iol 1999;110(1):47-52. 30. Hall WA, Liu H, Truwit CL. Functional magnetic resonance
13. Puce A, Constable RT, Luby ML, et al. Functional mag- imaging-guided resection of low-grade gliomas. Surg Neu-
netic resonance imaging of sensory and motor cortex: com- rol 2005;64(1):20-27; discussion 27.
CHAPTER 4 FUNCTIONAL MRI 53

31. Maldjian J, Atlas SW, Howard RS II, et al. Functional 40. Duffau H, Lopes M, Arthuis F, et al. Contribution of intra-
magnetic resonance imaging of regional brain activity operative electrical stimulations in surgery of low grade
in patients with intracerebral arteriovenous malforma- gliomas: a comparative study between two series with-
tions before surgical or endovascular therapy. J Neurosurg out (1985–96) and with (1996–2003) functional mapping
1996;84:477-483. in the same institution. J Neurol Neurosurg Psychiatry
32. Ojemann G, Ojemann J, Lettich E, Berger M. Cortical lan- 2005;76(6):845-851.
guage localization in left, dominant hemisphere. An elec- 41. Krishnan R, Raabe A, Hattingen E, et al. Functional
trical stimulation mapping investigation in 117 patients. magnetic resonance imaging-integrated neuronavigation:
J Neurosurg 1989;71(3):316-326. correlation between lesion-to-motor cortex distance and
33. Schulder M, Vega J, Narra V, et al. Functional magnetic reso- outcome. Neurosurgery 2004;55(4):904-914; discusssion
nance imaging and radiosurgical dose planning. Stereotact 914-915.
Funct Neurosurg 1999;73(1-4):38-44. 42. Feigl GC, Safavi-Abbasi S, Gharabaghi A, et al. Real-time
34. Komisaruk BR, Mosier KM, Liu WC, et al. Functional lo- 3T fMRI data of brain tumour patients for intra-operative
calization of brainstem and cervical spinal cord nuclei in localization of primary motor areas. Eur J Surg Oncol
humans with fMRI. Am J Neuroradiol 2002;23(4):609-617. 2008;34(6):708-715.
35. Maruyama K, Kamada K, Shin M, et al. Optic radia- 43. Kesavadas C, Thomas B, Sujesh S, et al. Real-time func-
tion tractography integrated into simulated treatment plan- tional MR imaging (fMRI) for presurgical evaluation of pae-
ning for Gamma Knife surgery. J Neurosurg 2007;107(4): diatric epilepsy. Pediatr Radiol 2007;37(10):964-974.
721-726. 44. Azmi H, Biswal B, Salas S, Schulder M. Functional
36. Aoyama H, Kamada K, Shirato H, et al. Integration of func- imaging in a low-field, mobile intraoperative magnetic
tional brain information into stereotactic irradiation treat- resonance scanner: expanded paradigms. Neurosurgery
ment planning using magnetoencephalography and mag- 2007;60(1):143-148; discussion 148-149.
netic resonance axonography. Int J Radiat Oncol Biol Phys 45. Hall WA, Liu H, Martin AJ, Pozza CH, Maxwell RE,
2004;58(4):1177-1183. Truwit CL. Safety, efficacy, and functionality of high-
37. Liu WC, Feldman SC, Schulder M, et al. The effect of tu- field strength interventional magnetic resonance imaging
mour type and distance on activation in the motor cortex. for neurosurgery. Neurosurgery 2000;46(3):632-641; dis-
Neuroradiology 2005;47(11):813-819. cussion 641-642.
38. Nimsky C, Ganslandt O, Fahlbusch R. Implementation 46. Nimsky C, Ganslandt O, von Keller B, Fahlbusch R. Intraop-
of fiber tract navigation. Neurosurgery 2006;58(4 Suppl erative high-field MRI: anatomical and functional imaging.
2):ONS292-ONS303; discussion ONS303-ONS304. Acta Neurochir Suppl 2006;98:87-95.
39. Berman JI, Berger MS, Chung SW, Nagarajan SS, Henry RG. 47. Nabavi A, Gering DT, Kacher DF, et al. Surgical navigation
Accuracy of diffusion tensor magnetic resonance imaging in the open MRI. Acta Neurochir Suppl 2003;85:121-125.
tractography assessed using intraoperative subcortical stim- 48. Schulder M, Azmi H, Biswal B. Functional magnetic reso-
ulation mapping and magnetic source imaging. J Neurosurg nance imaging in a low-field intraoperative scanner. Stereo-
2007;107(3):488-494. tact Funct Neurosurg 2003;80(1-4):125-131.
This page intentionally left blank
Chapter 5
Neuropsychological Testing:
Understanding Brain–behavior
Relationships
Mario F. Dulay1 , Corwin Boake 2 , Daniel Yoshor 3 , and Harvey S. Levin4
1
Department of Neurosurgery, The Methodist Hospital Neurological Institute,
Houston, Texas
2
Department of Physical Medicine and Rehabilitation, The University of Texas
Medical School, Houston, Texas
3
Department of Neurosurgery, Baylor College of Medicine, and Neuroscience Center, St. Luke’s
Episcopal Hospital, Houston, Texas
4
Departments of Physical Medicine and Rehabilitation, Pediatrics, Neurosurgery and Neurology,
Baylor College of Medicine, Houston, Texas

䉴 INTRODUCTION AND neuropsychological tests are often designed or validated


HISTORICAL PERSPECTIVE to quantify the impact of general or focal brain damage
on neurobehavioral functioning.
Neuropsychology is a field of science focused on There are many types of referral questions that can
the study of the neuroanatomical correlates of overt be addressed in different medical settings with a neu-
cognitive, behavioral, or emotional processes.1 Neu- ropsychological assessment including (1) what are the
ropsychological tests are noninvasive, psychometrically cognitive profiles associated with different disease states,
validated measures of cognition. Subspecialties that use (2) what is the severity of a specific disease, (3) what is
neuropsychological tests to understand brain–behavior the baseline cognitive status of a patient with an acute
relationships include cognitive neuroscience, experi- brain injury and what is the prognosis for cognitive re-
mental neuropsychology, cognitive neuropsychology, covery compared to how others have recovered after
psycholinguistics, neuropsychiatry, and clinical neu- injury, (4) what is the functional impact of brain damage
ropsychology. Clinical neuropsychology applies the on academic or vocational functioning, (5) what spe-
knowledge of human brain–behavior relationships to cific recommendations can be made for treatment of
clinical problems thereby bridging the gap between the identified cognitive deficits, and (6) how can we match
laboratory and the clinic. patients’ cognitive limitations to rehabilitation services?
Historically, a primary goal of a neuropsychological When addressing these questions, clinical neuropsychol-
evaluation was to quantify behavior in order to local- ogists are less concerned with localization and more in-
ize structural lesions.2 Today, neuropsychologists are no terested in determining diagnosis, prognosis, and treat-
longer asked to localize brain lesions based on testing ment planning.
given that structural imaging techniques such as comput- However, there are medical settings where refer-
erized tomography (CT) or magnetic resonance imaging ral questions directly relate to understanding brain–
(MRI) are effective at identifying neuroanatomical ab- behavior relationships, for example in settings when
normalities. Though neuropsychological assessment is the goal is the neurosurgical treatment of medically in-
not generally used for localizing brain lesions, validity of tractable epilepsy or a brain tumor. In these settings, re-
neuropsychological testing techniques is often based on ferral questions might include (1) what is the functional
whether or not there is concordance of test results with status of an area where a lesion is identified by CT or
focal abnormalities or neurologic dysfunction.3,4 That is, MRI, (2) what is the functional status of an area identified

55
56 SECTION I TECHNIQUES

by electroencephalography (EEG) as a seizure focus in tailed neuroimaging findings made it possible to study
the absence of an MRI identified lesion, (3) is there a pat- the cognitive effects of focal lesions affecting subcorti-
tern of cognitive test performance indicating lateralized cal structures (e.g., amnesia resulting from fornix and
hemispheric dysfunction that is consistent with EEG and thalamic lesions). Until recently, knowledge of human
imaging findings, (4) what is the risk of cognitive impair- brain–behavior relationships was based largely on re-
ment associated with removal of neural tissue, and (5) search with neurologic patients. The cognitive functions
what is the longitudinal impact of an identified growing of a given brain structure could be studied indirectly, to
or changing focal lesion on behavior? To address these the extent that neurologic disorders affecting that struc-
questions, it is assumed that distinct cognitive abilities ture led to changes in the function of interest. Interpre-
either map on to specific brain regions or represent an tation of these findings was complicated by many con-
interaction of specific brain regions. founding influences, such as recovery of function caused
by compensatory strategies or functional brain reorgani-
zation. The advent of functional neuroimaging has re-
䉴 HISTORY moved this major barrier by allowing direct observation
of cerebral activation while engaged in cognitive tasks.
Relationships of brain pathology to cognitive and par-
ticularly language disorders have been discussed since
the ancient medical literature.5 A critical insight into 䉴 UNDERLYING PRINCIPLES
the relationship between cognitive disorders and brain
pathology was prompted by case reports by Broca PHILOSOPHICAL CONSIDERATIONS IN
and others during the late 1800s, demonstrating that CLINICAL MAPPING
speech production was impaired selectively by stroke
and trauma affecting the frontal region of the left cere- A major assumption underlying clinical mapping is that
bral hemisphere.6 Following this discovery, numerous there are quantifiable behaviors that are associated with
cognitive disorders associated with focal brain pathology brain neuroanatomy and physiology. Different perspec-
were described. For example, the major syndromes of tives exist in neuropsychology regarding the extent to
aphasia were identified more than a century ago, mostly which there are localized functions in the brain. Tradi-
on the basis of case reports of stroke or brain trauma tional localizationists assume that several distinct brain
in which damage could be visualized during surgery regions are involved in the processing of specific be-
or postmortem. Advances in neurosurgery during the havioral functions and interact through associative path-
early 1900s created opportunities for the study of be- ways. This is supported by data that show that focal le-
havioral consequences of penetrating brain injuries and sions lead to a predictable pattern of motor, emotional,
ablative surgery. This research clarified the sensory and and cognitive impairments. This perspective is similar
cognitive functions of the temporal, parietal, and occip- to the concept of neurologic specificity and anatomic
ital lobes. The paradoxical results of pathology in the modularity.9,10 Traditional generalists assume parallel
prefrontal area, producing minimal change on conven- and/or hierarchical processing of cognitions/behaviors
tional cognitive testing, were also demonstrated.7 During that do not solely map onto one brain region and do not
the later 1900s, rapid progress in neuropsychology was work in isolation, but rather are produced as a result of
due to combined advances in neuroimaging and cog- an interaction of the parts or of the whole unit.11 The
nitive assessment. Cognitive tests were developed with idea of equipotentiality, or the proposal that a different
greater sensitivity and specificity to selective cognitive region will resume functions of a damaged or excised
disorders (e.g., amnesia, visual neglect). For example, region of the brain, provides support for the generalist
based on observations that patients with dysfunction of approach and is supported by research that shows that
the right cerebral hemisphere had relatively more dif- larger lesions lead to greater impairment.12 Traditional
ficulty performing visual perception tasks, studies us- connectionists assume that there are both specialized re-
ing refined tests to compare patients with different le- gions of functioning in the brain that are interrupted with
sion locations discovered that abnormality in the right focal damage, as well as areas away from the damaged
temporal–parietal area produced a selective impairment region that are interrupted due to a disruption of neural
in perceiving spatial relationships.8 The measurement interconnectivity.13,14
of brain abnormalities was revolutionized by comput- Which approach would we support? This is an em-
erized structural neuroimaging. A major consequence pirical question open to debate. However, while the
of this technology was to facilitate the investigation of basic orientation in some subspecialties within neu-
cognitive disorders associated with diffuse brain pathol- ropsychology is to assume localization of function, a
ogy. For example, patterns of cerebral atrophy in pa- strict localization of function view is unsupported. That
tients with degenerative dementia were compared quan- is, behaviors, emotions, and cognitions do not consis-
titatively to impairments in memory. In addition, de- tently map on to specific brain regions and there is
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 57

increasing evidence for the role of distributed neural net-


works mediating complex cognitive functions. Even so, X
neuropsychological tests are often developed and val-
idated to measure specific cognitive constructs that are Y
tied to specific brain processes and regions. That is, there Association
are robust associations that have been found when using
neuropsychological tests to lateralize and localize cogni-
tive functions when focal damage is known to exist (dis-
cussed later in this chapter). An important point to note
is that an identified lesion may not result in complete
loss of functions, but rather a degree of loss.15 Thus, A Task A Task B

the prominent sign of brain impairment may be the de-


gree of inefficiency of processing instead of total loss of
functioning. In some patient populations, neuropsycho- X
logical tests have data supporting their sensitivity to focal
brain dysfunction, for example, in patients with com-
plex partial seizures. In other patient populations such Y Dissociation
as traumatic brain injury due to closed head trauma, the
effects of multifocal or diffuse neuropathology often pre-
dominate and assessment of cognitive deficit associated
with a focal lesion is of less importance.
B Task A Task B

THE LESION METHOD,


ASSOCIATIONS, AND DISSOCIATIONS
Y
The efficacy of neuropsychological assessment in clin-
ical mapping to quantify the functional status of the
Double dissociation
brain is its use in conjunction with structural imaging X

techniques. The lesion method is an approach to under-


standing brain–behavior relationships based on the de-
gree to which brain damage or selective removal of neu-
ral tissue impairs some cognitive abilities while sparing
other abilities. The approach is guided by the assump- C Task A Task B

tion that if a specific brain region contributes to ade- Figure 5–1. Association, dissociation, and double
quate performance on a neuropsychological task, then dissociation. (Adapted from Bauer RM, Leritz E,
structural damage to that region should lead to func- Bowers D. Research methods in neuropsychology. In
tional impairment.7 The approach also assumes that cog- Comprehensive Handbook of Psychology, vol. 2,
nitive domains can be damaged separately from each edited by JA Schinka and WF Velicer. New York: John
Wiley & Sons, 2003, pp. 282–322.)
other. The material or domain specificity hypothesis, a
concept routinely discussed in the context of memory,
refers to the belief that specific tests and procedures can
be constructed that are sensitive to impairments in spe- memory (Task B in Fig. 5–1A) difficulties often co-occur.
cific regions of the brain.16 For example, verbal memory Association of deficits might occur because the process-
impairment is associated with language dominant uni- ing of abilities is anatomically close in the brain (even
lateral hippocampal damage and nonverbal memory though the systems might be functionally distinct) or be-
impairment is associated with nonlanguage dominant cause a deficit in one ability leads to impairment in the
unilateral hippocampal damage. 17 other ability. For example, a patient should be able to at-
Within the lesion method, three main approaches tend to episodic information to be remembered in order
are used to evaluate the relationship between neuropsy- for the information to be encoded and later retrieved.
chological data and lesion locations Fig. 5–1: (1) associa- A single dissociation occurs when a patient with a focal
tion of deficits, (2) single dissociation of deficits, and (3) lesion is impaired on one task (Task B in Fig. 5–1B for
double dissociation.9 Association of deficits refers to the patient X is impaired (higher is poorer), but performs
study of the correlation between two cognitive impair- normally on another task (Task A is intact for patient X
ments that occur together, for example, poor attention and patient Y, and patient Y performs within normal lim-
(Task A in Fig. 5–1A for patients X and Y) and verbal its for Task B). For example, nonverbal memory is intact
58 SECTION I TECHNIQUES

but there is impaired verbal memory associated with lan- for impairment. Evaluating multiple domains also serves
guage dominant (often left) hippocampal damage in a to address the concern that a single neuropsychologi-
single patient. A double dissociation occurs when a focal cal test does not solely measure one cognitive function,
lesion impairs a specific ability while other abilities are but rather a single test often involves multiple cognitive
left intact in one person (Task A is impaired but Task B is abilities. For example, adequate performance on a neu-
intact for patient X in Fig. 5–1C), and a focal lesion in an- ropsychological test requiring one to remember a story
other area in another person impairs a different specific involves the ability to attend to the story, understand in-
ability while leaving other abilities intact (Task B is im- structions, strategically encode the information, and re-
paired but Task A is intact for patient Y in Fig. 5–1C). For trieve the story. Poor performance revealed by an inabil-
example, the dissociation noted previously with a single ity to retrieve the story may result from a breakdown of
case would become a double dissociation when another any of these independent cognitive abilities. This issue
patient is found to have the opposite cognitive presen- is akin to the problem in functional MRI (fMRI) research
tation. That is, verbal memory may be intact but there of finding adequate control or baseline tasks for a block
is impaired nonverbal memory associated with nonlan- design to negate the behaviors that are not of interest,
guage dominant (often right) hippocampal damage in a or why event-related designs usually identify a network
separate patient. of brain regions activated during performance on a task.
Testing multiple cognitive domains may also help to ex-
plain why a person performs within normal limits on a
䉴 METHODS task when imaging data suggests that the person should
be impaired on the particular ability. For example, a per-
Clinical neuropsychologists use a variety of validated son may be able to compensate for a weakness such as
tests to evaluate brain functions. There are several books verbal memory difficulties related to identified left hip-
that provide extensive readings on the different types of pocampal sclerosis because of good working memory
neuropsychological tests that are available.18−20 A thor- and visual memory strategies that he or she uses to help
ough neuropsychological evaluation is routinely based compensate for the structural damage.
on at least four types of information: (1) medical history, There are several strategic approaches that a clinical
(2) clinical interview, (3) behavioral observations during neuropsychologist may take when evaluating cognitive
the interview and testing, and (4) the test battery.19 An functions. One approach is the use of a fixed battery
effective neuropsychological assessment begins with ob- of neuropsychological tests that covers most cognitive
taining relevant medical history including imaging and domains of interest. The approach could be used to cal-
neurologic exam results. These data, along with the culate both individual and global scores in a multivari-
referral question and patient complaints, often guide ate approach to determining a patient’s strengths and
which neuropsychological tests will be chosen for the weaknesses. Given that multiple domains are assessed
assessment. A clinical interview with the patient and representing different brain regions, this approach lends
family members is conducted to obtain relevant demo- itself well to evaluating lateralized and localized relative
graphic, social, educational, language learning, occupa- weaknesses associated with focal brain damage. This ap-
tional, and psychiatric history, as well as to obtain a proach also provides a uniform database that could be
patient’s level of activities of daily living in order to bet- used to study patterns of findings in relation to patient
ter interpret neuropsychological test results. Also, these variables, surgery, or pharmacologic treatment. Another
data provide ecologically valid clues to real world dys- approach is to use a flexible battery where a core bat-
function that may be correlated with neuropsychologi- tery of neuropsychological tests is used to screen for
cal impairments. Information obtained in the interview impairment and then the assessment is refocused on the
is also useful for modifying the selection or administra- identified cognitive problems. In this approach, empha-
tion of neuropsychological tests to accommodate spe- sis on test selection is often strongly guided by the dis-
cific disabilities, other limitations, and to gauge fluency ease state or type of neurologic impairment, or to ac-
in English for patients who primarily speak a foreign lan- commodate individual differences such as educational
guage. Behavioral observations are an important part of level or disabilities. However, in each approach, tests
gathering qualitative data during a neuropsychological may be chosen to measure previously identified seque-
assessment that can provide evidence for or disconfirm lae associated with the specific disease state based on
impressions garnered from the objective testing. clinical and research literature. Finally, a common ap-
In this section, we describe neuropsychological proach is to begin with a fixed battery and add additional
tests that represent different cognitive domains. A neu- tests when deficits in one domain are determined, or add
ropsychological assessment should evaluate multiple tests to help confirm impressions (e.g., the multiple fixed
functional domains. Evaluating multiple domains allows battery approach,13 ). Regardless of the approach, the
a clinician to determine the pattern of cognitive strengths chosen tests should have known normal standard values
and weaknesses, and often helps to clarify the reason and known psychometric properties in order to improve
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 59

the validly and reliability of testing. Qualitative methods performance to known chronologic age, gender and/or
are also useful. The process approach involves the iden- education-based norms improves the likelihood that the
tification and quantification of qualitative behaviors rep- interpretation of scores identified as impaired will be a
resentative of possible brain dysfunction. The approach valid indicator of brain dysfunction. We now define the
involves testing the limits21 to help explain how a per- different cognitive constructs that compose a neuropsy-
son performed poorly on a particular test and is usually chological assessment.
conducted after giving the standardized version of the
test.
Intelligence
Intelligence is a global composite of different mental
SUBJECT PREPARATION abilities mental, such as the capacities to solve prob-
lems, plan, comprehend ideas and language, remember
Several factors should be controlled for prior to and dur- and learn. An intelligence quotient (IQ) below 70 is a
ing an evaluation to maintain the validity and reliability criterion of mental retardation. One of the most widely
of the testing session. First, a testing session should be used measures of intelligence is the Wechsler Adult In-
conducted in an environment free of distractions (e.g., telligence Scale.23 The WAIS-IV has a composite score
noise). Second, it should be determined whether or not that is an indicator of general overall intellectual func-
a patient has visual or auditory acuity problems that will tioning (Full scale IQ), as well as subscales that estimate
affect cognitive testing. Third, a standard introduction verbal intelligence (known as verbal IQ (VIQ); e.g., fund
to testing should be provided to the patient that ex- of knowledge, vocabulary level) and nonverbal intelli-
plains what to expect. The introduction partly serves to gence (known as performance IQ (PIQ); e.g., visual con-
minimize patient anxiety, as too much anxiety can af- struction, spatial reasoning). The WAIS-IV manual pro-
fect neuropsychological test performance.22 Fourth, as a vides standard score values that indicate the significant
part of behavioral observations and during testing, eval- difference that exists between a VIQ-PIQ split suggest-
uate whether or not there are other patient issues such ing lateralized impairment with left hemisphere damage
as poor comprehension level, difficulty with attention, more likely to lead to lower VIQ scores and right hemi-
poor motivation, patient response style, and poor mood sphere damage more likely to lead to lower PIQ scores.23
state that can affect the validity of testing. Instructions
should be simplified or repeated if it appears through
Memory
behavioral observation or testing that a patient has dif-
ficulty comprehending instructions or difficulty attend- Figure 5–2 shows a taxonomy of the different types of
ing long enough to encode instructions. Poor effort, low memories that may exist.3 A wide variety of tests exist
motivation or malingering can be evaluated through the that measure the explicit memory domain. Episodic ver-
use of validated tests of malingering. Behavioral obser- bal memory tests commonly involve verbally presenting
vations may also suggest that a patient is giving poor a story or a list of words to a patient and then have the
effort (e.g., statements such as “I quit because I cannot patient repeat the story or list in a short duration (e.g.,
do it”), although it is often difficult to differentiate frus- within 30–45 seconds) or a longer duration (typically
tration with an inability to perform a task due to brain over 25–30 minutes). The most commonly used episodic
dysfunction from poor effort due to low self-esteem or verbal memory tests include the California Verbal Learn-
depressed mood. Taking these factors into account will ing Test, subtests of the Wechsler Memory Scales, the Rey
help to create a testing environment that promotes op- Auditory Verbal Learning test, and Warrington’s Recog-
timal performance. Noting these patient limitations will nition Memory Test for words.24−27 Episodic nonver-
also help clarify the nature of true cognitive impairments bal memory tests commonly involve visually present-
that reflect neuroanatomical degradation from misinter- ing faces, spatial locations or an abstract design to a
pretation of poor test scores due to confounding factors. patient and then have the patient draw what she saw,
point to the location of the objects, or match the objects
in a forced-choice paradigm. The most commonly used
TOOLS OF INVESTIGATION episodic nonverbal memory tests include the Warrington
Recognition Memory Test for faces, the Rey-Osterrieth
Neuropsychological tests should be chosen that have Complex Figures test, and the Brief Visual-Spatial Mem-
known psychometric properties. The validity and reli- ory Test.26,28−30 Comparison of results on verbal versus
ability of the most commonly used neuropsychological nonverbal memory tests may point to lateralization of
tools of assessment are published.18−20 Tests are given brain pathology. Implicit memory refers to unintentional
either by standard pencil and paper or computer ad- learning of material that someone was exposed to and is
ministration. Giving tests in a standardized fashion im- measured with tests of perceptual priming (e.g., word-
proves the reliability of the testing session. Comparing stem completion or lexical decision tasks), procedural
60 SECTION I TECHNIQUES

Long-term
memory

Explicit Implicit
(declarative) (nondeclarative)

Episodic Semantic (factual Procedural (motor, Perceptual


(events) knowledge) cognitive skills) priming

CC and operant Nonassociative


conditioning learning

Figure 5–2. Model of long-term memory. (Adapted from Squire LR. Memory systems
of the brain: A brief history and current perspective. Neurobiol Learn Mem 2004;82:
171-177.)

memory (e.g., learning to ride a bike), and nonasso- the Digit Span Backwards/Sequencing and Spatial Span
ciative learning (e.g., learning unrelated and irrelevant subtests of the Wechsler Memory Scale.27
information in the process of learning a target task).
Visual Perception
Language Visual perception refers to identification of stimuli and
localizing them in space. Measures of visual perception
Language is broadly defined by many different abilities
involve a variety of abilities including copying an object,
including comprehension, repetition, naming, fluency,
recognizing faces in a forced choice format, and judging
reading, writing, and prosody. Confrontation naming, or
line orientations. Common measures include the copy
a verbal response to visually presented objects or pic-
portion of the Clock Drawing Test, Benton’s Face Recog-
tures, is one of the most commonly tested domains using
nition Test, and Benton’s Judgment of Line Orientation
measures such as the Boston Naming Test.31 Standard-
Test.37,38
ized batteries such as the Multilingual Aphasia Examina-
tion and Boston Diagnostic Aphasia Examination have
been created that sample the wide range of language Cognitive Speed
abilities.32,33
Cognitive speed has been measured by many types
of tasks. The most common measures require visual-
motor tracking as a person quickly connects numbers
Attention and Working Memory
(e.g., Trails A; Army Individual Test,39 quickly naming
Attention refers to our ability to process certain informa- words (e.g., Stroop Word),40 or orally names numbers
tion selectively and continuously while ignoring other that match abstract geometric designs (e.g., Symbol Digit
information and is often evaluated by timed tests that Modalities Test).41 The main idea of these types of tasks
emphasize both speed and accuracy of performance. is to quantify how fast individuals are able to process
Standardized measures of different aspects of attention information and exert cognitive control as in the Stroop
(e.g., selective attention, focused attention and divided task, which involves suppressing a well-established
attention) include the Digit Span subtest of the WAIS-IV, response (reading) while performing a more effortful re-
the Ruff 2 and 7 Selective Attention Test and the Paced sponse (naming the color of print that conflicts with a
Auditory Serial Attention Test.23,34−36 Working memory color word such as “blue” appearing in red print). Cog-
refers to the ability to mentally manipulate and store in- nitive control and cognitive speed are less related to lat-
formation received either from visual or auditory sensory eralized and localized dysfunction and more sensitive
inputs or from long-term memory stores.20 Commonly to generalized white matter injury or disease and other
used standardized measures of working memory include neuropathology that results in motor or visual-motor
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 61

tracking impairments. Cognitive control also pertains to Once the data are scored, there are several meth-
executive functions as described in the following sec- ods to interpret neuropsychological test scores that re-
tion. quire clinical judgment. These methods include (1) in-
terpreting the level of performance (normal/abnormal,
below average above average) on a particular test, (2)
Executive Functions
analyzing the pattern of performance on test scores,
Executive functions refer to complex behavioral pro- (3) comparison of tests that purportedly measure op-
cesses that involve initiation, planning, problem solv- posite sides of the hemispheres and (4) the evalu-
ing and adaptation, purposive action and goal seeking, ation of pathognomonic signs indicative of localized
the ability to inhibit responses, self-monitoring of be- impairment.49 Analysis of level of performance involves
havior, and avolition.42 As such, there are diverse sets interpreting how poorly someone performs on a test (or
of tools that can be used. Several neuropsychological a group of tests in the same domain) in comparison
tests thought to be symptomatic of problems with ex- to a standard, usually normative data from a sample of
ecutive functions have been validated that quantify the the general population. Impaired performance is often
extent of loss in different types of executive function- defined by cut-off scores or percentile rank. Percentile
type behaviors.43 The most common tasks include the ranks below a certain threshold (e.g., 10%, 5%) are used
Ruff Figural Fluency Test (RFFT; a measure of the abil- to define impairment based on the referral context. An-
ity to generate spatial designs; sustained attention, self- alyzing the pattern of test scores helps to determine
monitoring to avoid repetition, regulation of responses, patient strengths and weaknesses and helps provide par-
cognitive processing speed),36 the Stroop Color-Word allel support for functional impairment when an identi-
task (ability to inhibit a response, selective attention, fied lesion exists. Use of continuous data also helps to
cognitive processing speed),40 the Trail Making Test part determine impairment on a continuum if the cognitive
B (visual conceptual and visuomotor tracking, set shift- function relies on several brain regions since, as previ-
ing, ability to inhibit responses; Army Individual Test),39 ously mentioned, impairments are not usually absolute
Wisconsin Card Sorting Test (assess abstract behavior, but rather relative. Comparing how someone performs
ability to use feedback, ability to shift sets),44 and FAS on tests representing the left or right hemisphere is
Letter Fluency (word knowledge base, organization of a standard tool in neurologic exams. Neuropsycholo-
verbal retrieval, cognitive flexibility, ability to inhibit a gists can use the evaluation of sensory, motor, memory,
response).37 Newer standardized measures of executive language, and perceptual-motor skills to discuss the rel-
functioning include the Iowa gambling task45 and the ative efficiency of the two hemispheres. Finally, use
Delis–Kaplan Executive Function System.46 of pathognomonic signs can help to interpret data by
identifying specific types of behaviors characteristic of
specific brain dysfunction. Classic examples include
DATA ANALYSIS AND paraphasic errors (indicative of language dominant
INTERPRETATION hemisphere impairment), as well as perserverative re-
sponses and confabulations (indicative of probable
Data analysis is straightforward and begins by scoring frontal lobe impairment).
information (e.g., response accuracy, response speed) Interpretation of neuropsychological data should be
using standard criteria and using the raw scores to deter- approached with caution given that several factors can
mine the z-score or percentile rank compared to norma- contribute to the misclassification of functional impair-
tive data from samples of nonbrain injured controls. The ment. Individual differences in level of comprehension,
goal of using normative data is to understand whether visual and auditory acuity, IQ, sedative medications, and
or not poor performance on a test is actually impaired severe depression are examples of factors that can lead
or if performance is expected compared to others with to false lateralizing and localizing information that mis-
similar demographic characteristics. For example, two lead conclusions drawn from neuropsychological test-
individuals of the same age may differ in their abil- ing and therefore need to be taken into account to
ity to name pictures of objects if their education lev- increase the likelihood of validly assessing neuropsy-
els differ, reflecting experiential differences rather than chological results. Also, patients for whom English is a
functional brain impairment. There are several sources second language may perform below expectation on
that provide normative data for the most commonly tests that involve processing complex language. Another
used and well-validated measures of neuropsycholog- issue, which is a limit of the lesion method and any
ical functioning.18,19,20 Data are routinely adjusted for clinical mapping approach, is that the same lesion in
chronological age since expected performance on many the same place does not always equate to the same
neurocognitive tasks change with age.47 There are also observable behavior due to individual factors such as
normative data for some tests that adjust for race, sex, chronologic age, education, sex, culture, and other fac-
and level of education.48 tors. Also, disease characteristics may lead to functional
62 SECTION I TECHNIQUES

reorganization of abilities.50 Normative data help to par- Global Versus Local Processing
tially address this concern by providing a definition of of Information
typical performance. Thus, a goal of much neuropsy- Previous research suggests that the two hemispheres are
chological research has been to determine what factors specialized in the perceptual organization of visual in-
account for individual differences. formation with the right hemisphere integrating many
features of an object or scene as a whole (called global
processing, also known as holistic or contextual or con-
䉴 APPLICATION figurational gestalt) and the left hemisphere processing
local (fine details of a whole object, a.k.a. analytic or
The following section mainly focuses on research re- featural) features of an object or scene in healthy hu-
garding frontal or temporal lobe epilepsy/surgery pa- man subjects.59 Presenting information to both visual
tients given that the bulk of studies have reported data fields, Doyon and Milner60 found faster processing of
on these groups, and we only briefly discuss the litera- local information for patients who had undergone right
ture regarding patients with parietal and occipital lobe anterior temporal lobectomy (ATL) compared to subjects
epilepsy. We present these data with particular focus post left-ATL. The authors interpreted this finding as re-
on the neuroanatomical correlates of neuropsychologi- flecting the damaged right temporal lobe’s minimized
cal tests. There is an extensive history of the study of the focus on the processing of global characteristics of vi-
neuroanatomical correlates of neuropsychological test sual information. Other researchers have used complex
performance with pre- and posttemporal lobectomy pa- visual information with responses quantified using free-
tients with intractable epilepsy as neuropsychological as- hand drawing when stimuli were presented to full vi-
sessment methods play a valuable role in the evaluation sual fields in either presurgical or postsurgical patients
of patients presenting at epilepsy treatment centers.51 with epileptic seizures. The results of these studies are
Surgical candidates provide the closest approximation of equivocal with two studies providing some evidence for
lesion studies in animal models given that pre- to post- global processing impairments in individuals with right
surgical evaluations can be conducted on patients that TLE,61,62 but another study not finding evidence for dis-
receive similar excision of structures, mostly anterior sociation between hemispheres.63 Finally, applied to the
temporal lobe, which helps minimize a criticism of the processing of unfamiliar faces, research suggests that the
lesion method by increasing the similarity among patient right hemisphere processes features of a face as a whole
lesions. and the left hemisphere relies on a parts-based decoding
of faces.64,65

LATERALIZATION OF FUNCTION: LEFT LOCALIZATION OF FUNCTION: THE


VERSUS RIGHT HEMISPHERE TEMPORAL LOBE

Verbal Versus Performance IQ Episodic Verbal Memory


The lateralizing value of material specific IQ indices has The largest amount of neuropsychological research on
been studied in patients with epilepsy. Double disso- individuals with epilepsy has focused on the study of
ciations have been reported such that left hemisphere memory. Modern theories of medial temporal lobe func-
damage is more likely to lead to lower VIQ scores, and tioning agree that the hippocampus and parahippocam-
right hemisphere damage is more likely to lead to poorer pal gyrus are important for memory of factual knowledge
PIQ scores.52−56 For example, using the mean score dif- and episodic events.3,66 Evidence supporting the role
ference between the VIQ and PIQ, Akanuma et al.52 of the language-dominant hippocampus in the encod-
found that patients with video-EEG defined left-sided ing, short-term storage and retrieval of verbal material
temporal lobe epilepsy (TLE) were more likely to have is well validated. Verbal recall and recognition memory
poorer VIQ scores compared to individuals with right- impairments have been found in patients with video-
sided TLE attributable to difficulties with language pro- EEG defined left-TLE on measures of declarative mem-
cesses. In nonepilepsy patient populations, the VIQ–PIQ ory tested with story recall, verbal paired associates,
split is more prominent in males57 as lateralization of and list-learning formats (see Bell and Giovagnoli for
cerebral specialization (i.e., into verbal and visuo-spatial) a review).67 Further, the presence of imaging abnor-
is less characteristic of females than males. Larrabee58 malities suggestive of left-hippocampal mesial temporal
suggested that the lateralizing utility of the VIQ–PIQ sclerosis (MTS) is associated with poorer performance
split may be diminished by language impairments that on measures of verbal memory compared to individuals
affect performance on the supposed “nonverbal” PIQ without left-MTS or patients with right-MTS.68−70 Further,
subtests.58 decreased left lateral temporal lobe hypometabolism,
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 63

based on positron emission tomography (PET), is asso- based on fMRI,86 and in patients with higher presurgi-
ciated with impaired verbal memory in individuals with cal verbal memory ability,74,87−89 which all are hypothe-
left-TLE.71 Recently, Pauli et al.72 found that neuronal sized to reflect functional (e.g., nonimpaired) tissue that
cell loss within the internal limb of the dentate gyrus was ultimately resected. There is also evidence that re-
of the hippocampus was the strongest predictor of the sults of Wada testing (intracarotid injection of amobar-
ability of a patient to acquire new verbal memories. bital to test for language and memory) indicating poor
Other evidence supporting the role of the left lan- memory ability in the contralateral temporal lobe pre-
guage dominant temporal lobe in the processing of ver- dicts postsurgical verbal memory impairment because
bal memories comes from research in patients who un- the contralateral hemisphere is not able to compen-
derwent unilateral ATL. Left-ATL leads to a precipitous sate for the ipsilateral removal of functional tissue.90
drop in verbal recall and learning ability.73−80 Figure Other factors found to account for individual differ-
5–3A shows the rate of impaired verbal learning across ences in the extent of decline in verbal memory after a
trials for left-ATL patients compared to better perfor- language dominant/left-temporal lobe resection include
mance for right-ATL patients on the Buschke Verbal Se- more extensive left-ATL,74 older age at ATL surgery,91,92
lective Reminding Test from data from our series of pa- poor seizure control after surgery,93,94 male gender, as
tients at the Baylor College of Medicine.81 Figure 5–3B women have a more bilateral representation of verbal
shows the magnitude of impaired performance for the memory,95 and depressed mood state either before96 or
sum across trials between the two groups. The decline after surgery.97,98
in verbal memory abilities after surgery is especially ap-
parent in patients without presurgical imaging abnor-
malities or hippocampal sclerosis,75,82−85 in patients with
Episodic Nonverbal Memory
greater presurgical left medial temporal lobe activation
Material specific nonverbal memory impairments have
been associated with nonlanguage dominant temporal
lobectomy. Table 5–1 shows the extant evidence be-
12 Left-sided tween studies that assessed nonverbal memory for ab-
Number of words recalled

11 Right-sided
10
stract designs, faces and spatial locations after right-ATL
9 compared to patients who underwent left-ATL. For tests
8 of memory for abstract designs, there are many stud-
7 ies that show both impairments and no impairments
6
after right-ATL compared to left-ATL. Data are more con-
5
4 vincing using memory for faces with all studies show-
3 ing impaired performance in individuals who underwent
2 right-ATL. Moran et al.105 was the only study that found
1 that the impairment was not greater for right-ATL pa-
0
A 0 1 2 3 4 5 6 7 8 9 10 11 12 tients compared to left-ATL patients because both groups
Trial showed impaired performance on face memory after
surgery. Spatial learning tests also provide some evi-
120 dence for a role in the nonlanguage dominant hemi-
Total number of words recalled

p < .001 sphere in nonverbal memory functioning, but there are


110 still several studies that show no post-ATL impairments.
The lack of evidence relating the right temporal lobe
100 to nonverbal memory tasks from some studies has been
attributed to several reasons including (1) visual informa-
90 tion to be remembered is often not complex enough to
elicit the deficit, (2) visual information can sometimes be
80
easily verbalized so more abstract and less familiar infor-
mation should be used to elicit anterior right-hemisphere
functions, and (3) the contralateral hemisphere is able to
70
B Left Right compensate effectively for deficiencies when the visual
information is presented to both visual fields.
Surgery side
Nonverbal visual memory impairments are more
Figure 5–3. (A) Shows the rate of verbal learning
likely to occur in patients with right-MTS or smaller
across trials by side of anterior temporal lobectomy right-sided hippocampal volumes.82,133 New postsurgi-
(ATL). (B) Bar graph shows the total recall of words cal nonverbal memory impairments are predicted by re-
from 12 trials divided by side of ATL. section of a relatively large right hippocampus,85 better
64 SECTION I TECHNIQUES

䉴 TABLE 5–1. STUDIES OF VISUAL MEMORY WITH POSTSURGICAL GROUP DATA COMPARING
RIGHT TO LEFT-ANTERIOR TEMPORAL LOBECTOMY

Type of Nonverbal Memory Significantly Poorer Performance vs. Left-ATL No Significant Difference vs. Left-ATL

Abstract design memory Abrahams et al.99 Gleissner et al.a100


Doyon and Milner60 Chelune et al.87
Goldstein and Polkey101 Dulay et al.102
Graydon et al.103 Katz et al.76
Ivnik et al.104 Moran et al.a105
Jones-Gotman106 Nunn et al.107 (wnl)
Jones-Gotman et al.108 Rausch et al.109
Helmstaedter et al.110 Selwa et al.111
Kimura112 Spiers et al.113
Rausch and Babb114
Face memory Chiaravalloti and Glosser115 Moran et al.a105
Chiaravalloti et al.116
Doss et al.117
Dulay et al.102
Milner118,119
Morris et al.120
Spiers et al.113
Spatial learning and memory Abrahams et al.99 Chiaravalloti and Glosser115 (wnl)
Corkin121 Feigenbaumb and Morris122 (wnl)
Dulay et al.102 Goldstein et al.a,c123
Milner16 Maguire et al.a124
Nunn et al.107,125 Owen et al.a126
Parslow et al.127
Petrides128
Pigott and Milner129
Pillon et al.130
Smith and Milner131
Spiers et al.113
Worsley et al.132
a
Indicates memory test performance was impaired for both left and right ATL compared to normative data or significantly poorer than a
normal control group.
b
There was no significant difference between left/right or controls when using an egocentric spatial memory task, but the right-ATL
group performed significantly poorer than controls using an allocentric spatial memory task.
c
Goldstein et al.123 found no significant difference among between left/right or controls when using an egocentric spatial memory task,
but the right- and left-ATL group performed significantly poorer than controls using an allocentric spatial memory task.
Wnl, indicates within normal limits compared to normative data or a normal control group.

presurgical nonverbal memory ability,100 right sided patients with TLE or post-ATL. Anatomical distinctions
presurgical fMRI activation during a nonverbal task,86,134 between explicit/episodic memory and implicit memory
larger right-lateral neocortex and mesial temporal originally came from research with patient H.M. who
excision,106,119 and later age at onset of seizures,101 had a bilateral temporal lobectomy. He was found to
which all suggest the removal of functional right hip- have impaired explicit memory with the ability to learn
pocampal tissue. Later age at onset is a predictor because new information implicitly suggesting that mesial tempo-
individuals who have seizures that begin in adulthood ral lobe structures are not essential in the processing of
have a greater likelihood of developing adequate non- implicit memories.78 In research with other patient pop-
verbal memory abilities that are at risk for loss when ulations, implicit memory impairments have been tied to
functional hippocampus is resected. basal ganglia, cerebellum, occipital lobe, and association
cortex damage depending on the characteristics of the
implicit memory task.136,137 However, implicit memory
impairments have been demonstrated in individuals with
Implicit Memory
a unilateral TLE or post-ATL138−141 though there is also
Relatively few studies have evaluated the biologic basis research that has not found impairment.142 To address
of implicit memory tests (measures of unintended or in- this discordance, Leritz et al.135 hypothesized that hip-
direct learning) in patients with epileptic seizures135 (see pocampus is important for the formation of new memo-
for a review). In almost every case, the focus has been on ries regardless of the explicit/implicit nature of the task
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 65

because the important factor in explaining the variability more sensitive to left temporal lobe damage than con-
is the degree to which the tests that were used required frontation naming.160−162 Finally, the presence of phone-
effort or intention. Thus, implicit memory tasks that re- mic paraphasias, but not semantic paraphasias, occurs
quire more effort for remembering (even though it is at a greater rate in left TLE146,163,164 suggesting an un-
unintended learning) will be more dependent on mesial derlying phonological deficit (vs. a semantic deficit) to
temporal lobe functioning. There is also debate regard- identified naming difficulties.
ing lateralized processing of material-specific verbal im-
plicit memory tasks as some have found no differences
between left- and right-TLE patients,140 whereas others
have found differences.138,139 Interestingly Zaidel DW, Processing of Faces
et al.,141 found a significant association between poorer
Previous research with pre- and postsurgical epilepsy pa-
performance on a verbal implicit memory task and re-
tients indicates greater impairments in face recognition
duced left-CA1 hippocampal cell densities.
memory ability for unfamiliar faces,115,165 face recogni-
tion memory for famous faces,166 and identification of
Language fearful facial expressions167,168,169 for right-TLE patients
compared to left-TLE patients or normal controls. The
Different types of naming tasks are impaired in individu-
amygdala likely plays a primary role in the emotional la-
als with left-TLE compared to individuals with right-TLE.
beling of faces,170 though the effects of recurrent seizures
Confrontation naming tasks, or the ability to provide a
in patients with right-TLE or the effects of right-ATL are
one or two word verbal label for a picture, has been
not specific enough to be able to differentiate the differ-
the most commonly studied type of naming task. Con-
ent roles of mesial temporal lobe structures (e.g., amyg-
frontation naming impairments in individuals with left-
dala vs. hippocampus, hippocampus vs. entorhinal cor-
TLE is associated with the presence of left hippocampal
tex). A role of both the left and right hemispheres has
atrophy or sclerosis,143,144 reduced left hippocampal cell
been hypothesized to exist for the processing of face
density,145 postictal language disturbance,146 and more
stimuli when task instructions require the lexical or se-
lateral lesions (vs. mesial).147 Research also demonstrates
mantic naming of familiar or famous faces with simulta-
a precipitous drop in confrontation naming ability after
neous visual processing of the face.166,171,172−175
surgical resection of the left temporal lobe that is pre-
dicted by more extensive resection of lateral temporal
cortex,148 better presurgical naming ability,149 absence
of hippocampal sclerosis144,150,151 and later age at onset
Olfaction
of seizures,149,150,152−154 which are indicative of a func-
tional left hemisphere. Further, Bell et al.155 found that In the field of epilepsy research, olfactory loss has been
object names learned later in childhood are more likely consistently found with seizure focus and damage.176
to be lost after left ATL. Table 5–2 displays studies that assessed olfactory func-
Other test material-specific language difficulties tioning in patients with epilepsy, the olfaction-cognition
have been associated with the language-dominant left tasks employed, and the region implicated in loss of abil-
temporal lobe. For example, Hermann et al.156 reported ity. These studies indicate poor performance on mea-
that left-TLE patients performed significantly poorer on sures of olfactory function as a result of temporal lobe
tests of sentence repetition, listening comprehension and disruption, damage, or surgery. Previous research has
reading comprehension compared to right-TLE patients. found conflicting results regarding lateralization when
Further, left-ATL patients have greater difficulty nam- assessing odor discrimination (ability to discriminate be-
ing living things (e.g., animals) versus naming nonliving tween two or more odors) and odor recognition memory
things (e.g., tools) after surgery compared to right-ATL performance (the ability to remember and discriminate
patients157,158 providing support for the idea that there an odor among a group of other odors after a delay) in
exists focal and lateralized category specific naming re- patients with epilepsy. The right temporal lobe is con-
gions of the brain. In addition, Glosser and Donofrio159 sistently implicated in all of the studies; nonetheless, the
found that left-ATL patients had a selective deficit for left temporal lobe is also implicated in several studies
naming “pictured objects,” but not “pictured actions” (see West and Doty for a review).189 On the other hand,
compared to right-ATL patients, which provided evi- most of the studies that have assessed odor identifica-
dence of specialization of left temporal lobe structures tion ability (the ability to discriminate between odors
for processing nouns but not verbs. Auditory naming and retrieval of an odor label) indicated deficits with im-
tasks that assess the ability to provide definitions of pairment to either the left or right temporal lobe. Other
common animals and objects (without visual stimuli, but researchers have found odor identification deficits in
rather a cued verbal label) have also been shown to be patients with orbitofrontal resection and frontal lobec-
sensitive to left temporal lobe impairment, and may be tomies that impinged on orbitofrontal cortex.176,185,188
66 SECTION I TECHNIQUES

䉴 TABLE 5–2. STUDIES THAT REPORT DEFICITS ON OLFACTORY MEASURES OF COGNITIVE


FUNCTIONING IN PATIENTS WITH EPILEPSY AS A FUNCTION OF SIDE OF SURGERY

Task Number of Patients with Impairment and Location of Seizures or Surgery

Odor recognition memory impairment


Right-sided impairment only:
Eskenazi et al.177 16 anterior right temporal lobe resection
Jones-Gotman and Zatorre178 36 right temporal lobe and 8 right orbitofrontal/frontal lobectomy (ipsilateral)a
Martinez et al.179 11 right medial temporal lobe resection (ipsilateral)a
Rausch and Serfetinides180 1 patient with right temporal lobe resection
Left or right-sided impairment:
Henkin et al.181 4 left and 7 right temporal lobectomy
Rausch et al.182 7 left and 7 left anterior temporal lobectomy
Eskenazi et al.183 7 language dominant and 10 nonlanguage dominant side temporal lobectomy
Odor identification impairment
Right-sided impairment only:
Eskenazi et al.177 16 anterior right temporal lobe resection
Martinez et al.179 11 right medial temporal lobe resection (ipsilateral)a
Left or right-sided impairment:
Eichenbaum et al.184 Patient H.M., bilateral temporal lobe resection
Eskenazi et al.183 7 language dominant and 10 nonlanguage dominant side temporal lobectomy
Jones-Gotman and Zatorre185 36 left and 35 right temporal lobe lobectomy
8 left frontal-orbital, 12 right frontotemporal, and 8 right lobectomy
Jones-Gotman et al.108 11 left and 14 right amygdalohippocampectomy (ipsilateral)a ;
11 left and 12 right anterior temporal lobe resection (ipsilateral)a ;
12 left and 10 right temporal neocorticectomy (ipsilateral)a
Lehrner et al.186 Case study on a patient with amygdalohippocampectomy
Odor discrimination impairment
Right-sided impairment only:
Abraham and Mathai187 14 right temporal lobectomy
Martinez et al.179 11 right medial temporal lobe resection (ipsilateral)a
Left or right-sided impairment:
Eichenbaum et al.184 Patient H.M., bilateral temporal lobe resection
Eskenazi et al.183 7 language dominant and 10-nonlanguage dominant side temporal lobectomy
Zatorre and Gotman188 31 left temporal and 31 right temporal lobectomy
9 left frontal (ipsilateral),a 10 right orbitofrontal, and 10 right frontal lobectomy
a
Ipsilateral refers to the finding that there was olfactory impairment only for the nostril ipsilateral to the side of the seizure focus or
surgery.

LOCALIZATION OF FUNCTION: THE spatial working memory tasks (vs. verbal working mem-
FRONTAL LOBE ory) after right frontal lobectomy compared to left frontal
lobectomy,196,197 but this laterality effect of spatial work-
Attention and Working Memory ing memory tasks has not been seen in nonsurgical
epilepsy candidates with a unilateral seizure focus.125
Previous research indicates that attention and working
memory are impaired in individuals with frontal lobe
seizure foci, or post unilateral frontal lobe resection;190
Design and Verbal Fluency
(see Helmstaedter et al. for reviews).191,192 For example,
Helmstaedter et al.192 found that patients with frontal The ability to generate words (phonemic or semantic
lobe epilepsy (FLE) performed poorer on a measure of fluency) and create novel designs (design fluency) is
sustained attention compared to patients with TLE (see associated with frontal lobe functioning.198−203 For ex-
also Auclair et al.).193 Further, McDonald et al.194 found ample, Jones-Gotman and Milner200 originally found a
that FLE patients performed below controls or patients double dissociation with poorer performance on a mea-
with TLE on a measure of selective attention. Within the sure of design fluency for individuals with right-TLE
frontal lobe, damage to dorsolateral prefrontal cortex has compared with left-TLE patients, and greater verbal flu-
been specifically associated with impaired performance ency impairments for left-TLE patients compared to
on measures of attention and working memory.195 Oth- right-TLE patients. In a follow-up study with postsur-
ers have found greater material specific impairment on gical data, Jones-Gotman204 found that centrally located
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 67

right frontal resections (vs. right frontal lateral resections) is associated with poorer executive functioning221 pos-
were more likely to lead to the design fluency impair- sibly reflecting greater chronicity of disease or possible
ment. Others have reported that verbal and design flu- re organization of these behavioral functions.
ency tasks were sensitive in differentiating individuals There is also literature demonstrating executive dys-
with FLE from individuals with TLE.202,203 Left frontal function in individuals with TLE compared to healthy
lobectomy is associated with declines in phonemic flu- controls Table 5–3. Most of the research to date has
ency ability after surgery and design fluency after right focused on performance on the Wisconsin Card Sort-
frontal lobectomy when the resection was large (e.g., ing Test (WCST), a measure of abstraction, sorting
including frontal pole and orbitofrontal cortex).201 strategy, attention shifting, indicating impaired per-
Not all of the data is consistent concerning the later- formance compared to normal controls or normative
ality of phonemic versus design fluency.153,205 McDonald data.221,222,224,227,230 Others have found impaired perfor-
et al.206 found that design fluency impairments could be mance on tests of mental flexibility222 in patients with
demonstrated in patients with left frontal lobe lesions. TLE. Laterality differences have been found on measures
Further, Davidson et al.207 found the presence of ver- of executive functioning such that individuals with left-
bal fluency deficits after right frontal resection. Davidson TLE performed more poorly on the WCST compared
et al.207 suggested that executive dysfunction explains with individuals with right-TLE208,221 or left-TLE com-
the presence of the verbal fluency impairments after pared with healthy controls.226
right-sided surgery while both linguistic and executive Several theories have been proposed that attempt
impairments may explain the verbal fluency deficits after to explain why executive functions would be dimin-
left-sided surgery. Of note, Helmstaedter et al.191 found ished in individuals with TLE. For example, research has
phonemic fluency impairments after left frontal lobec- provided support for the hypothesis that electrical dis-
tomy only when transient expressive aphasia difficulties charges propagate to the frontal lobe thereby interfer-
occurred when surgery included left frontal premotor/ ing with frontal-type functions.226,229,235 Other research
supplemental motor areas. has provided support for the idea that hippocampal
dysfunction impairs the registration or retrieval of in-
formation important for adequate performance on tests
Executive Functioning
of executive function.208,223 For instance, poorer perfor-
Performance on different aspects of executive func- mance on measures of executive functioning is asso-
tioning is diminished in individuals with FLE or af- ciated with hippocampal sclerosis compared to those
ter unilateral frontal lobectomy. Specifically, impair- without sclerosis.208,222 Corcoran and Upton223 found
ments have been found on cognitive tests designed that performance on a measure of concept formation
to measure response inhibition,16,191,192,203,208,209 mental if poorer for TLE patients with hippocampal sclerosis
flexibility,190,207,210,211 concept formation,190 estimation compared to patients with FLE.
of abilities,212 humor appreciation,213 proverb interpreta-
tion214 and motor programming and coordination192
compared to individuals with TLE or normal con-
trols. Individuals with FLE are also more likely to LOCALIZATION OF FUNCTION: THE
demonstrate memory difficulties due to poor en- PARIETAL AND OCCIPITAL LOBES
coding strategies215,216 or difficulties with proactive
interference,217 have loss of set when performing a Relatively few studies have been published evaluating
task,210 and demonstrate perseverative responding.211,218 neuropsychological performance associated with pari-
In an interesting study, McDonald et al.216 recently found etal lobe epilepsy.236 On the basis of case studies and/or
that patients with left-FLE benefit the most from struc- behavioral observation without formal neuropsycholog-
tured encoding of verbal information while right-FLE ical testing, several studies report that parietal lobe
patients do not. lesions are associated with visual perception and con-
Several factors are associated with greater extent of structional difficulties, as well as disturbances of body
executive dysfunction in individuals with frontal lobe image (e.g., asomatognosia or a lack of awareness of the
epilepsy. For example, executive dysfunction is associ- condition of part of one’s body) and visual illusions.237
ated with depressed mood110,219,220 possibly reflecting Salanova et al.238 reported that 9 of 30 presurgical pari-
frontostriatal dysfunction. In addition, larger frontal lobe etal lobe epilepsy patients showed impaired visuocon-
resections have been associated with difficulties in in- struction and spatial impairments, and 7 of 27 had new
hibiting responses and a perseverative response style201 onset apraxia and face recognition problems after resec-
compared to smaller resections. Further, Helmstaedter tion of parietal lobe seizure foci. Djordjevic and Jones-
et al.191 found distinct impairments in initiation of ac- Gotman239 describe a series of patients with right parietal
tions when frontal lobe resection included supplemen- lobe focal damage who had poor or distorted copying
tal motor areas. Finally, earlier age at onset of seizures of a complex figure, and they found left parietal seizure
68 SECTION I TECHNIQUES

䉴 TABLE 5–3. STUDIES THAT ASSESS COMMON MEASURES OF EXECUTIVE FUNCTIONS IN


INDIVIDUALS WITH EPILEPSY

Study Sample Measures Results

Allegri et al.222 TLE WCST, Trails B TLE poorer than NC


TLE with sclerosis poorer vs.
w/out
Corcoran and Upton223 FLE, TLE WCST TLE with sclerosis worse than
FLE
LTLE poorer than RTLE
Drake et al.224 TLE and GE WCST categories Larger percentage of poor EF
with TLE
Giovagnoli208 LTLE, RTLE, LF, RF WCST LF and TLE with sclerosis
perform poorest
Giovagnoli and Avanzini225 LTLE and RTLE WCST categories, No between-group differences
perseverations Phonemic Fluency, Trails B
Helmstaedter et al.191 L/RFLE, L/RTLE Letter fluency, FLE worse on all vs. TLE
Maze test (response No LTLE vs. RTLE
inhibition) differences
Hermann et al.226 LTLE, RTLE, GTC WCST RTLE more PEs vs. GTC
LTLE more PEs vs. GTC
RTLE poorer than LTLE
Hermann et al.219 LTLE and RTLE WCST Depression and WCST
performance related in LTLE
Horner et al.227 LTLE and RTLE WCST Both groups perform poorly on
WCST
Klein et al.228 epilepsy with gliomas Stroop Color-Word Poorer performance vs. NC
Concept Shifting Test EF related to seizure severity
and AED
Martin et al.229 CPS WCST Comorbid GE with CPS worse
than
CPS alone
Martin et al.230 LTLE vs. RTLE WCST category, PE No between-group differences.
Trails B, COWAT 40% RTLE impaired on WCST,
38%
LTLE; COWA similarly impaired.
Martin et al.231 TLE letter fluency LTLE worse than RTLE, RTLE
poorer than NC
Moore and Baker232 LTLE and RTLE Stroop Color-Word No lateralized differences
Paradiso et al.220 LTLE, RTLE WCST, Trails B, COWAT Depressed with TLE have
poorer
COWAT and TMT
Prevey et al.233 CPS vs. GTC vs. NC Stroop Color-Word, COWAT GTC worse on both vs. CPS
and NC
Strauss et al.221 LTLE vs. RTLE WCST Interaction between side of
focus and
Age of seizure onset with LTLE
and younger age poorest
Suchy et al.202 LTLE, RTLE FAS, Figural Fluency No lateralization; no between
group FAS
LFLE, RFLE, FLE poorer on Figural vs. TLE
Upton and Corcoran234 RLTE WCST Poor sorting performance vs.
NC

TLE, temporal lobe epilepsy; WCST, Wisconsin Card Sorting Test; NC, normal controls; FLE, frontal lobe epilepsy; GE, generalized
epilepsy; CPS, complex partial seizures, LTLE, left temporal lobe epilepsy; RTLE, right temporal lobe epilepsy; EF, executive function;
RFLE, right frontal lobe epilepsy; GTC, generalized tonic-clonic epilepsy; PE, perseverative errors; AED, antiepileptic drugs; TMT, trail
making test, FAS, letter fluency for letters FAS.
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 69

foci to be associated with inefficient reading abilities. Fi- as PET, magnetoencephalography (MEG) and fMRI that
nally, Djordjevic and Jones-Gotman239 reported impaired identify the brain regions involved in a cognitive task,
tactile discrimination ability for the hand contralateral to but do not indicate if the region is vital to competent
the parietal lobe seizure focus. performance on a task. That is, many brain regions are
Few studies have also been published describing implicated during performance of a task using functional
the cognitive sequelae associated with occipital lobe neuroimaging techniques even though the regions may
epilepsy. Visual agnosia has been consistently reported not be required. Transcranial magnetic stimulation (TMS)
in case studies of individuals with occipitotemporal lobe and electrical stimulation (ES) also show that a brain re-
seizure foci.240−242 Seizure semiology consisting of vi- gion is required to perform a task by creating a transient
sual hallucinations (e.g., unfamiliar faces or scenes), vi- lesion, but ES is very invasive and only used on select
sual illusions, blindness, seeing colored dots, and field samples, and TMS can both excite and inhibit cognitive
defects have also been reported.243,244 In a sample functions leading to interpretation difficulties.
of 11 individuals with occipital lobe epilepsy, Chilosi Second, neuropsychological assessment allows re-
et al.245 found that 8 of the 11 had poorer PQ abili- searchers to hypothesize about how impairment occurs
ties compared to VIQ abilities (laterality of seizures were because multiple domains are evaluated to create a cog-
not reported), partly attributable to difficulties in com- nitive profile of strengths and weaknesses. This is a ben-
petently completing the highly loaded visual-perceptual efit in clinical settings versus neuroimaging techniques
subtests that make up PQ.246 They also found impaired because the profile may be used to separate cognitive
performance on the Judgment of Line Orientation test impairment correlated with structural lesions from im-
and Benton Face Recognition task in a subgroup of the pairments associated with individual disease-related and
occipital lobe epilepsy patients. In a sample of 28 teens demographic differences. For example, a right-handed
and adults with “posterior cortex” lesions (5 parietal, 5 patient with identified right hippocampal sclerosis may
occipital, 14 temporal-occipital and 2 temporal-parietal perform poorly both on measures of verbal and nonver-
occipital), Luerding et al.247 found that PIQ showed a bal memory, and a neuropsychological assessment can
small but significant decline after posterior cortex focal suggest that the verbal memory impairment may be as-
resection while VIQ showed significant improvement. In sociated with severe attentional difficulties (e.g., associ-
a sample of 21 children with occipital lobe epilepsy (side ated with poly anti-epileptic medication use). Neuropsy-
of focus not identified), Gulgonen et al.246 found signif- chological assessment can also be advantageous versus
icant impairments compared to a normal control group neuroimaging techniques in a clinical setting because
on tests of PIQ, visual memory, and visual planning and rehabilitation, vocational, and academic recommenda-
problem solving. They also unexpectedly found poorer tions can be made based on a cognitive profile. A closely
performance on measures of VIQ and verbal memory related third strength is that neuropsychological assess-
leading the authors to suggest that the impact of oc- ment is the best approximation of how individuals act in
cipital lobe epilepsy in children (or the treatment for the real world settings. Neuroimaging experimental design
seizures) leads to generalized deficits and does not read- is naturally limited by the setting (e.g., scanner noise)
ily dissociate into verbal versus visual domains. Finally, and requires many different assumptions when analyz-
Fleischman et al.136 revealed a dissociation on a test of ing the data that make the techniques further removed
implicit visual memory with intact explicit visual mem- from real world situations. Among the different mapping
ory in a 30-year-old right handed male who underwent techniques in this book, only the ecological validity of
right occipital lobectomy of Brodmann’s areas 17, 18 and neuropsychological functioning in predicting level of ac-
a portion of 19 suggesting that the right occipital lobe tivities of daily living, academic achievement and every
may be intricately involved in the processing of visual day attentional and memory abilities in individuals with
implicit memories. focal brain damage has been well studied.249,250
A fourth strength of neuropsychological assessment
compared to other techniques in a clinical setting is
that the normal range of test scores is known, and can
䉴 RELATIVE STRENGTHS AND be statistically verified (e.g., impaired performance be-
WEAKNESSES COMPARED TO low fifth percentile rank), for the most commonly used
OTHER MAPPING TECHNIQUES tests compared to age, education, and/or sex-based nor-
mative data. Functional imaging uses control groups,
There are several strengths to the use of neuropsycho- but methods vary from center to center, demographic-
logical assessment in a clinical setting when evaluat- equated control groups do not minimize problems with
ing the neuroanatomical correlates of cognition. First, individual variability when sample sizes are small, and
correlating structural damage with neuropsychological normative data are not yet routinely available, mak-
deficits shows that the brain region is required to per- ing clinical utility limited when trying to define func-
form the task.248 This is in contrast to techniques such tional impairment. In addition, the quantitative nature of
70 SECTION I TECHNIQUES

neuropsychological test results facilitates longitudinal niques that can require the patient to passively view im-
testing (e.g., pre- and postsurgical testing). ages or scenes to determine brain function. However, all
Finally, we argue that neuropsychological testing is clinical mapping techniques are limited by the patient’s
a different way of looking at certain aspects of the brain– comprehension of instructions, motivation to perform
behavior relationship that may be helpful to physicians. at an optimal level, and visuomotor abilities to execute
The power of mapping in a clinical setting lies in the necessary movements.
ability of clinicians to use multiple mapping techniques A final limitation is that interpretation of neuropsy-
that provide converging validity to explicate the role of chological test results is more complex when applied to
a region of the brain. For example, for a single patient, patients from different language, learning, and educa-
the efficacy of neuropsychological assessment may be tional backgrounds. Neuropsychological test normative
demonstrated when neuropsychological impairment in data and standardized instructions are lacking in many
a specific domain is correlated with a structural lesion, developing countries.
hypometabolism with PET, MEG reduction and/or de-
creased fMRI BOLD signal. For research purposes across
patients, evidence for the neuroanatomical substrates of 䉴 CASE EXAMPLE
a cognitive function may be demonstrated by concurrent
evidence of neuropsychological impairment, increased We now present data from a surgical candidate with
functional activity in healthy patients (MEG, PET, fMRI), intractable epileptic seizures to demonstrate the prin-
and deficits induced by TMS or electrical stimulation. ciples discussed in this chapter. The patient is a
There are also several limitations of neuropsy- 30-year-old right-handed female with 12 years of edu-
chological assessment as a tool to understand brain– cation, referred for neuropsychological assessment as
behavior relationships. First, replication of a relationship part of a diagnostic work-up for possible surgical in-
between structural damage and cognitive impairment is tervention for medically intractable seizures. Prolonged
limited by the inherent variability in the extent of an MRI video-EEG scalp with scalp electrodes revealed electro-
identified lesion or the extent of surgical resection of the graphic seizure activity originating over the left temporal
patient population. This creates a problem because mul- lobe with nearly immediate spread to the right tempo-
tiple structural areas tied to multiple functional domains ral lobe. Seizure semiology consisted of staring, chew-
may be impaired simultaneously. Other issues that in- ing movements, loss of awareness and responsiveness
troduce variability include: a lesion in the same location for 1–2 minutes, picking at her arm, and increased agi-
does not always lead to the same functional deficits be- tation or violent behavior after the event. The postictal
tween individual subjects, a lesion in the same location behavior led to animosity among family members. In-
can cause multiple deficits, and lesions in different brain tracarotid sodium amobarbital testing indicated mixed
regions can lead to a similar cognitive impairment. These language dominance, and her right hemisphere was
are also limitations of other lesion methods such as found to support memory functions. Structural MRI sug-
TMS and electrical stimulation. Understanding structure/ gested left hippocampal sclerosis. Neurologic exam was
function relations with any mapping technique is con- normal. Medical history was negative for other neuro-
founded by the variables of individual differences (hand- logic disorders or other medical conditions. She was be-
edness, demographics, poor motivation, poor attention ing treated for major depression. Her complex partial
level, low IQ, depression), and there are always excep- seizure disorder began at the age of 6-years-old, and at
tions to robust structure/function relationships due to the time of her evaluation, seizures were occurring at a
reorganization of function or plasticity after injury. rate of 1–2 per week at the time of the evaluation. She
An additional limitation is that neuropsychological graduated from high school with average grades, and
assessment does not determine what cognitive function never attended special education services. She stopped
is supported by the lesioned area, but rather it estimates working as a manager at a store in the year prior to
how the system processes information when the area is the evaluation because of difficulties with transportation
damaged or excised. Therefore, we cannot differentiate to work, interpersonal difficulties associated with anger
whether a brain region is the primary source involved in management, and on the job memory difficulties.
a specific function from whether or not the region serves As part of her neuropsychological assessment, there
as a pathway that, when damaged, leads to a disconnec- were no noted irregular behavioral observations during
tion from other regions that are intricately involved in a the clinical interview or during testing other than notice-
specific function. We can only demonstrate that the area able word finding difficulties and the presence of para-
is required because, when damaged, task performance phasic errors during conversational speech. Neuropsy-
is impaired. chological testing was conducted covering all major
Additionally, the validity of a neuropsychological cognitive areas. Testing revealed average overall intel-
assessment is limited by the ability of the patient to par- lectual functioning with a significantly poorer VIQ com-
ticipate in testing. This is contrast to neuroimaging tech- pared to her non-VIQ (12 point difference). There were
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 71

mild defects in recent memory, verbal recall and recog- REFERENCES


nition memory, letter and category fluency, confronta-
tion naming with paraphasic errors, and auditory nam- 1. American Academy of Clinical Neuropsychology (AACN)
ing suggesting left temporal lobe involvement. She had practice guidelines for neuropsychological assessment
relative strengths for perceptual constructional skills, and consultation. Clin Neuropsychol 2007;21:209-231.
2. Loring DW. History of neuropsychology through epilepsy
cognitive speed, sustained attention, and visual reten-
eyes. Arch Clin Neuropsychol 2010;25:259-267.
tive memory. Planning and problem solving were intact, 3. Squire LR, Stark CE, Clark RE. The medial temporal lobe.
except that she would lose set when performing dif- Annu Rev Neurosci 2004;27:279-306.
ferent tasks. Given the already existing impairments in 4. Stuss DT, Levine B. Adult clinical neuropsychology:
the processing of verbal information, the patient was lessons from studies of the frontal lobes. Annu Rev Psy-
considered to be at minimal risk for sustaining addi- chol 2002;53:401-433.
tional memory and language impairments after standard 5. Benton AL, Joynt RJ. Early descriptions of aphasia. Arch
left-ATL (consisting of resection of lateral temporal neo- Neurol 1960;3:205-221.
cortex from superior or middle temporal gyrus to the 6. Finger S. Origins of neuroscience: A history of explo-
collateral fissure, as well as anterior hippocampal for- rations into brain function. New York: Oxford University
mation, subiculum, parahippocampal gyrus, and infe- Press, 1994.
7. Damasio H, Damasio AR. Lesion analysis in neuropsy-
rior amygadala). The patient underwent left-ATL and
chology. New York: Oxford University Press, 1989.
was seizure-free thereafter. She noticed some additional 8. Benton A. Neuropsychology: past, present, and future.
memory difficulties immediately after surgery with re- In Handbook of Neuropsychology, edited by F Boller
turn to baseline after several months. Language abilities and J. Grafman. Amsterdam: Elsevier Science, 1988
improved to the point that paraphasic errors were no pp. 3-27.
longer apparent. She was able to resume driving, but 9. Coltheart M. Assumptions and methods in cognitive neu-
did not return to work because of her verbal memory ropsychology. In The handbook of cognitive neuropsy-
difficulties. She was referred for rehabilitation services in cholgy: What deficits reveal about the human mind, edited
order to help her to compensate for the continued verbal by B Rapp. Philadelphia: Taylor & Francis, 2001.
memory difficulties and to help her learn to more fully 10. Fodor JA. The modularity of mind: An essay on faculty
exploit her cognitive strengths in non-VIQ, nonverbal psychology. Cambridge, MA: MIT Press, 1983.
11. Luria AR. The Working Brain. An Introduction to Neu-
memory, processing speed, and planning and problem
ropsychology. New York: Basic Books, 1973.
solving. 12. Orbach J. The neuropsychological theories of Lashley and
Hebb. Lanham MD: University Press of America, 1998.
13. Goldstein G. Functional considerations in neuropsychol-
䉴 CONCLUSION ogy. In Ecological validity of neuropsychological testing,
edited by RJ Sbordone and CJ Long. Delray Beach, FL: GR
Press/St. Lucie Press, 1996.
Our review shows that complex relationships exist 14. Matlin MW. Cognition, 6th edition. Hoboken, NJ: Wiley,
among multiple factors that combine to influence per- 2005.
formance on any neuropsychological test. The main 15. Shallice T. From Neuropsychology to mental structure.
strength of neuropsychological assessment in a clinical New York: Cambridge University Press, 1988.
setting for brain mapping is its use in conjunction with 16. Milner B. Visually-guided maze learning in man: effects
other mapping methods to provide converging validity of bilateral hippocampal, bilateral frontal, and unilateral
about the role of a region of the brain in overt behav- cerebral lesions. Neuropsychologia 1965;3:317-338.
ior. Testing methods are also useful for creating a risk– 17. Milner B, Johnsrude I, Crane J. Right medial temporal-lobe
benefit profile when estimating the possible postsurgical contribution to object-location memory. In Parietal and
decrements associated with excision of eloquent areas hippocampal contributions to spatial cognition, edited by
JM O’Keefe and N Burgess, Oxford: Oxford University
of the brain. We believe that neuropsychological assess-
Press, 1998, pp. 247-58.
ment provides an important contribution to multidisci- 18. Lezak MD, Howieson DB, Loring DW. Neuropsychologi-
plinary teams because unique information is provided cal Assessment, 4th edition. New York: Oxford University
about how focal damage relates to a patient’s level of Press, 2004.
functioning in their everyday lives. 19. Mitrushina MN, Boone K, Razani J, et al. Handbook of
Normative Data For Neuropsychological Assessment, 2nd
edition. New York: Oxford University Press, 2005.
20. Strauss E, Sherman EMS, Spreen O. A Compendium
䉴 ACKNOWLEDGEMENT of neuropsychological tests: Administration, norms, and
commentary, 3rd edition. New York: Oxford University
The authors would like to acknowledge the generous Press, 2006.
support of The Moody Foundation for a grant on 21. Milberg WP, Hebben NA, Kaplan E. The Boston pro-
epilepsy research. cess approach to neuropsychological assessment. In
72 SECTION I TECHNIQUES

Neuropsychological Assessment of Neuropsychiatric Dis- ness edited by S Salloway, PF Malloy, JD Duffy. Washing-
orders, 1st edition, edited by I Grant and KM Adams. New ton, DC: American Psychiatric Press, Inc, 2001, pp. 125-137.
York: Oxford University Press, 1986. 44. Grant DA, Berg EA. A simple objective test for measuring
22. Castaneda AE, Tuulio-Henriksson A, Marttunen M, et al. A flexibility in thinking. J Gen Psychol 1948;39:15-22.
review on cognitive impairments in depressive and anxi- 45. Bechara AR, Damasio AR, Damasio H, et al. Insensitivity
ety disorders with a focus on young adults. J Affect Disord to future consequences following damage to prefrontal
2008;106:1-27. cortex. Cogn 1994;50:7-15.
23. Wechsler D. WAIS-IV administration and scoring manual. 46. D-KEFS; Delis-Kaplan Executive Function Scale. The Psy-
San Antonio, TX: Pearson, 2008. chological Corporation, 2000.
24. Delis DC, Kramer JH, Kaplan E, Ober BA. California Verbal 47. Park HL, O’Connell JE, Thomson RG. A systematic review
Learning Test, 2nd edition. San Antonio, TX: Psychologi- of cognitive decline in the general elderly population. Int
cal Corporation, 2000. J Geriatr Psychiatry 2003;18:1121-1134.
25. Rey A. L’Examen Clinique en Psychologie. Paris: Press 48. Heaton RK, Miller SW, Taylor MJ et al. Revised compre-
Universitaire de France, 1964. hensive norms for an expanded Halstead-Reitan Battery:
26. Warrington EK. Warrington, Recognition memory test. Demographically adjusted neuropsychological norms for
Windsor, UK: NFER-Nelson, 1984. African American and Caucasian adults Scoring Program.
27. Wechsler D. Wechsler Memory Scale, 4th edition. San An- Odessa, FL: Psychological Assessment Resources, Inc,
tonio, TX: Pearson, 2009. 2004.
28. Benedict RHB. Brief Visuospatial Memory Test—Revised. 49. Hannay HJ, Lezak MD. The neuropsychological examina-
Odessa, FL: Psychological Assessment Resources, Inc, 1997. tion: Interpretation. In Neuropsychological Assessment,
29. Rey A. L’examen psychologique dans les cas d’encephalo- 4th edition, edited by MD Lezak, DB Howieson and DW
pathie traumatique. Archives de Psychologie 1941;28:215- Loring. New York: Oxford University Press, 2004.
285. 50. Ewing-Cobbs L, Barnes MA, Fletcher JM. Early brain injury
30. Osterrieth PA. Filetest de copie d’une figure complex: in children: development and reorganization of cognitive
Contribution a l’etude de la perception et de la memoire. function. Dev Neuropsychol 2003;24:669-704.
Archives de Psychologie 1944;30:286-356. 51. Helmstaedter C: Neuropsychological aspects of epilepsy
31. Kaplan E, Goodglass H, Weintraub S. Boston naming test: surgery. Epilepsy Behav 2004;5:S45-55.
Revised edition. Philadelphia: Lea & Febiger, 1983. 52. Akanuma N, Alarcon G, Lum F, et al. Lateralising value
32. Benton AL, Hamsher K, Sivan AB. Multilingual Aphasia of neuropsychological protocols for presurgical assess-
Examination 3rd edition. San Antonio, TX: Psychological ment of temporal lobe epilepsy. Epilepsia 2003;44:408-
Corporation, 1994. 418.
33. Goodglass H, Kaplan E, Barresi B. The Assessment of 53. Bornstein RA. Verbal IQ-performance IQ discrepancies on
Aphasia and Related Disorders, 3rd edition. Baltimore: the Wechsler Adult Intelligence Scale-Revised in patients
Lippincott, Williams and Wilkins, 2001. with unilateral or bilateral cerebral dysfunction. J Consult
34. Gronwall D. Paced auditory serial addition task: A mea- Clin Psychol 1983;51:779-780.
sure of recovery from concussion. Percep Motor Skills 54. Kaufman AS, Lichtenberger EO: Assessing adolescent and
1977;44:367-373. adult intelligence, 2nd edition, Boston: Allyn & Bacon,
35. Paschler HE. The Psychology of Attention. Cambridge, 2002.
MA: MIT Press, 1997. 55. Satz P. Specific and nonspecific effects of brain lesions in
36. Ruff RM, Allen CC. Ruff 2 & 7 Selective Attention Test. Pro- man. J Abnorm Psychol 1966;71:65-70.
fessional Manual. Psychological Assessment Resources, 56. Hermann BP, Gold J, Pusakulich R, et al. Wechsler
1996. adult intelligence scale-revised in the evaluation of an-
37. Benton AL, Sivan AB, Hamsher K, et al. Contributions to terior temporal lobectomy candidates. Epilepsia 1995;36:
Neuropsychological Assessment. New York: Oxford Uni- 480-487.
versity Press, 1994. 57. Bornstein RA, Matarazzo JD. Wechsler VIQ versus PIQ dif-
38. Shulman KI, Shedletsky R, Silver I. The challenge of time: ferences in cerebral dysfunction: a literature review with
clock drawing and cognitive function in the elderly. Int J emphasis on sex differences. J Clin Neuropsychol 1982;4:
Geriatr Psychiatry 1986;1:135-140. 319-334.
39. Army Individual Test Battery. Manual of directions and 58. Larrabee GJ. Another look at VIQ-PIQ scores and unilat-
scoring. Washington, DC: War Department, Adjutant Gen- eral brain damage. Int J Neurosci 1986;29:141-148.
eral’s Office, 1944. 59. Van Kleeck MH. Hemispheric differences in global versus
40. Golden CJ. Stroop color and word test: A manual for clini- local processing of hierarchical visual stimuli by normal
cal and experimental uses. Wood Dale, IL: Stoelting, 1978. subjects: new data and a meta-analysis of previous stud-
41. Smith A. Symbol Digits Modalities Test, Western Psycho- ies. Neuropsychologia 1989;27:1165-1178.
logical Services, Los Angeles, 1982. 60. Doyon J, Milner B. Right temporal-lobe contribution to
42. Damasio AR, Anderson SW. The frontal lobes. In Clinical global visual processing. Neuropsychologia 1991;29:343-
Neuropsychology, edited by KM Heilman, E Valenstein, 360.
4th edition. New York: Oxford University Press, 2003, pp. 61. Breier JI, Plenger PM, Castillo R, et al. Effects of temporal
404-446, 716. lobe epilepsy on spatial and figural aspects of memory
43. Malloy PF, Richardson ED. Assessment of frontal lobe for a complex geometric figure. J Int Neuropsychol Soc
functions. In The frontal lobes and neuropsychiatric ill- 1996a;2:535-540.
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 73

62. Loring DW, Lee GP, Meador KJ. Revising the Rey- ral lobectomy using the Wechsler Memory Scale-Revised.
Osterrieth: rating right hemisphere recall. Arch Clin Neu- Arch Clin Neuropsychol 1993;8:381-395.
ropsychol 1988;3:239-47. 80. Novelly RA, Augustine EA, Mattson RH, et al. Selective
63. McConley R, Martin R, Baños J, et al. Global/local scoring memory improvement and impairment in temporal lobec-
modifications for the Rey-Osterrieth Complex Figure: re- tomy for epilepsy. Ann Neurol 1984;15:64-67.
lation to unilateral temporal lobe epilepsy patients. J Int 81. Buschke H, Fuld PA. Evaluation of storage, retention,
Neuropsychol Soc 2006;12:383-390. and retrieval in disordered memory and learning. Neu-
64. Rhodes G. Configural coding, expertise, and the right rol 1974;11:1019-1025.
hemisphere advantage for face recognition. Brain Cogn 82. Baxendale, SA, Van Paesschen W, Thompson, PJ et al.
1993;22:19-41. Hippocampal cell loss and gliosis: relationship to preop-
65. Rossion B, Dricot L, Devolder A, et al. Hemispheric asym- erative and postoperative memory function. Neuropsychi-
metries for whole-based and part-based face processing in atry Neuropsychol Behav Neurol 1998;11:12-21.
the human fusiform gyrus. J Cogn Neurosci 2000;12:793- 83. Bell BD, Davies KG. Anterior temporal lobectomy, hip-
802. pocampal sclerosis, and memory: recent neuropsycholog-
66. Moscovitch M, Nadel L, Winocur G, et al. The cognitive ical findings. Neuropsychol Rev 1998;8:25-41.
neuroscience of remote episodic, semantic and spatial 84. Sawrie S, Martin RC, Knowlton R, et al. Relationships
memory. Curr Opin Neurobiol 2006;16:179-190. among hippocampal volumetry, proton magnetic reso-
67. Bell BD, Giovagnoli AR. Recent innovative studies of nance spectroscopy, and verbal memory in temporal lobe
memory in temporal lobe epilepsy. Neuropsychol Rev epilepsy. Epilepsia 2001;42:1403-1407.
2007;17:455-476. 85. Trenerry MR, Jack CR, Ivnik RJ, et al. MRI hippocampal
68. Sass KJ, Spencer DD, Kim JH, et al. Verbal memory impair- volumes and memory function before and after temporal
ment correlates with hippocampal pyramidal cell density. lobectomy. Neurol 1993;43:1800-1805.
Neurol 1990;40:1694-1697. 86. Powell HW, Richardson MP, Symms MR, et al. Preoper-
69. Lencz T, McCarthy G, Bronen RA. Quantitative magnetic ative fMRI predicts memory decline following anterior
resonance imaging in temporal lobe epilepsy: relationship temporal lobe resection. J Neurol Neurosurg Psychiatry
to neuropathology and neuropsychological function. Ann 2008;79:686-693.
Neurol 1992;31:629-637. 87. Chelune G, Naugle RI, Luders HO, Awad IA. Prediction
70. Rausch R. Anatomical substrates of interictal memory of cognitive change as a function of preoperative ability
deficits in temporal lobe epileptics. Int J Neurol 1987;21- status among temporal lobectomy patients seen at six-
22:17-32. month follow-up. Neurol 1991;41:399-404.
71. Rausch R, Henry TR, Ary CM, et al. Asymmetric interictal 88. Davies KG, Hermann BP, Bush AJ, et al. Prediction
glucose hypometabolism and cognitive performance in of episodic memory loss following anterior temporal
epileptic patients. Arch. Neurol 1994;51:139-144. lobectomy using a multiple regression model. Epilepsia
72. Pauli E, Hildebrandt M, Romstöck J, et al. Deficient mem- 1996;37:130.
ory acquisition in temporal lobe epilepsy is predicted by 89. Jokeit H, Ebner A, Holthausen H, et al. Individual
hippocampal granule cell loss. Neurology 2006;67:1383- predication of change in delayed recall of prose pas-
1389. sages after left-sided anterior temporal lobectomy. Neurol
73. Dulay MF, Levin HS, York MK, et al. Changes in individual 1997;49:481-487.
and group spatial and verbal learning characteristics af- 90. Bell BD, Davies KG, Haltiner AM, et al. Intracarotid amo-
ter anterior temporal lobectomy. Epilepsia 2009;50:1385- barbital procedure and prediction of postoperative mem-
1395. ory in patients with left temporal lobe epilepsy and hip-
74. Helmstaedter C, Elger CE. Cognitive consequences of two- pocampal sclerosis. Epilepsia 2000;41:992-997.
thirds anterior temporal lobectomy on verbal memory in 91. Hermann B, Seidenberg M, Haltiner A, et al. Relation-
144 patients: A three-month follow-up study. Epilepsia ship of age at onset, chronological age, and adequacy of
1996;7:171-180. preoperative performance to verbal memory change after
75. Hermann BP, Wyler AR, Somes G et al. Pathological sta- anterior temporal lobectomy. Epilepsia 1995;36:137-145.
tus of the mesial temporal lobe predicts memory out- 92. Loring D, Lee GP, Meador KJ, et al. Age effects on ver-
come from left anterior temporal lobectomy. Neurosurg bal and visuospatial memory after temporal lobectomy.
1992a;31:652-656. Epilepsia 1993;34:72.
76. Katz A, Awad IA, Kong AK, et al. Extent of resection in 93. Hamberger MJ, Drake EB. Cognitive functioning following
temporal lobectomy for epilepsy. II. Memory changes and epilepsy surgery. Curr Neurol Neurosci Rep 2006;6:319-
neurologic complications. Epilepsia 1989;30:763-771. 326.
77. Lee TM, Yip JT, Jones-Gotman M. Memory deficits af- 94. Sanyal SK, Chandra PS, Gupta S, et al. Memory and in-
ter resection from left or right anterior temporal lobe in telligence outcome following surgery for intractable tem-
humans: a meta-analytic review. Epilepsia 2002;43:283- poral lobe epilepsy: relationship to seizure outcome and
291. evaluation using a customized neuropsychological bat-
78. Milner B, Corkin S, Teuber HL. Further analysis of the tery. Epilepsy Behav 2005;6:147-155.
hippocampal amnesic syndrome: 14 year follow-up study 95. Trenerry M, Jack CR, Cascino GD, et al. Gender differ-
of H.M. Neuropsychologia 1968;6:215-234. ences in post-temporal lobectomy verbal memory and
79. Naugle RI, Chelune GJ, Cheek R, et al. Detection of relationships between MRI hippocampal volumes and
changes in material-specific memory following tempo- preoperative verbal memory. Epilepsy Res 1995;20:69-76.
74 SECTION I TECHNIQUES

96. Dulay MF, Schefft BK, Fargo JD, et al. Severity of depres- 113. Spiers HJ, Burgess N, Maguire EA, et al. Unilateral tem-
sive symptoms, hippocampal sclerosis, auditory mem- poral lobectomy patients show lateralized topographical
ory, and side of seizure focus in temporal lobe epilepsy. and episodic memory deficits in a virtual town. Brain
Epilepsy Behav 2004;5:522-531. 2001;124:2476-2489.
97. Busch RM, Dulay MF, Kim KH, et al. Pre-surgical 114. Rausch R Babb TL. Hippocampal neuron loss and memory
mood predicts memory decline after anterior tempo- scores before and after temporal lobe surgery for epilepsy.
ral lobe resection for epilepsy. Arch Clin Neuropsychol Arch Neurol 1993;50:812-817.
2011;26(8):739-745. 115. Chiaravalloti ND, Glosser G. Memory for faces dissoci-
98. Dulay, MF, York, MK, Soety EM, et al. Memory, emotional ates from memory for location following anterior tempo-
and occupational outcome before and after anterior tem- ral lobectomy. Brain Cogn 2004;54:35-42.
poral lobectomy for complex partial seizures. Epilepsia 116. Chiaravalloti ND, Tulsky DS, Glosser G. Validation of the
2006;47:1922-1930. WMS-III Facial Memory subtest with the Graduate Hospi-
99. Abrahams S, Pickering A, Polkey CE, Morris RG. Spa- tal Facial Memory Test in a sample of right and left an-
tial memory deficits in patients with unilateral damage to terior temporal lobectomy patients. J Clin Exp Neuropsy-
the right hippocampal formation. Neuropsychologia 1997; chol 2004;26:484-497.
35:11-24. 117. Doss RC, Chelune GJ, Naugle RI. WMS-III performance
100. Gleissner U, Helmstaedter C, Schramm J, et al. Memory in epilepsy patients following temporal lobectomy. J Int
outcome after selective amygdalohippocampectomy: a Neuropsychol Soc 2004,10;173-179.
study in 140 patients with temporal lobe epilepsy. Epilep- 118. Milner B. Disorders of memory after brain lesions in man:
sia 2002;43:87-95. preface: material-specific and generalized memory loss.
101. Goldstein LH, Polkey CE. Short-term cognitive changes af- Neuropsychologia 1968;6:175-179.
ter unilateral temporal lobectomy or unilateral amygdalo- 119. Milner B. Visual recognition and recall after right
hippocampectomy for the relief of temporal lobe temporal-lobe excision in man. Epilepsy Behav 2003;4:
epilepsy. J Neurol Neurosurg Psychiatry 1993;56:135-140. 799-812.
102. Dulay MF, Levin HS, York MK, et al. Predictors of individ- 120. Morris RG, Abrahams S, Polkey CE. Recognition memory
ual visual memory decline after unilateral anterior tempo- for words and faces following unilateral temporal lobec-
ral lobe resection. Neurology 2009;72:1837-1842. tomy. Br J Clin Psychol 1995;34:571-576.
103. Graydon FJ, Nunn JA, Polkey CE, Morris RG. Neuropsy- 121. Corkin S. Tactually-guided maze learning in man:
chological outcome and the extent of resection in the uni- Effects of unilateral cortical excisions and bilat-
lateral temporal lobectomy. Epilepsy Behav 2001;2:140- eral hippocampal lesions. Neuropsychologia 1965;3:339-
151. 351.
104. Ivnik RJ, Sharbrough FW, Laws ER Jr. Effects of anterior 122. Feigenbaum JD, Morris RG. Allocentric versus egocentric
temporal lobectomy on cognitive function. J Clin Psychol spatial memory after unilateral temporal lobectomy in hu-
1987;43:128-137. mans. Neuropsychol 2004;18:462-672.
105. Moran M, Seidenberg M, Sabsevitz D, et al. The acquisition 123. Goldstein LH, Canavan AG, Polkey CE. Cognitive map-
of face and person identity information following anterior ping after unilateral temporal lobectomy. Neuropsycholo-
temporal lobectomy. J Int Neuropsychol Soc 2005;11:237- gia 1989;27:167-177.
248. 124. Maguire EA, Frackowiak RS, Frith CD. Learning to find
106. Jones-Gotman, M: Memory for designs: the hippocampal your way: a role for the human hippocampal formation.
contribution. Neuropsychologia 1986;24:193-203. Proc Biol Sci 1996;263:1745-1750.
107. Nunn JA, Polkey CE, Morris RG. Selective spatial mem- 125. Nunn JA, Graydon FJ, Polkey CE, Morris RG. Differ-
ory impairment after right unilateral temporal lobectomy. ential spatial memory impairment after right temporal
Neuropsychologia 1998;36:837-848. lobectomy demonstrated using temporal titration. Brain
108. Jones-Gotman M, Zatorre RJ, Olivier A, et al. Learning 1999;122:47-59.
and retention of words and designs following excision 126. Owen AM, Morris RG, Sahakian BJ, et al. Double disso-
from medial or lateral temporal-lobe structures. Neuropsy- ciations of memory and executive functions in working
chologia 1997;35:963-973. memory tasks following frontal lobe excisions, temporal
109. Rausch R, Kraemer S, Pietras CJ, et al. Early and late cogni- lobe excisions or amygdalo-hippocampectomy in man.
tive changes following temporal lobe surgery for epilepsy. Brain 1996;119:1597-1615.
Neurology 2003;60:951-959. 127. Parslow DM, Morris RG, Fleminger S, et al. Allocentric
110. Helmstaedter C, Van Roost D, Clusmann H, Urbach H, El- spatial memory in humans with hippocampal lesions. Acta
ger CE, Schramm J. Collateral brain damage, a potential Psychol 2005;118:123-147.
source of cognitive impairment after selective surgery for 128. Petrides M. Deficits on conditional associative-learning
control of mesial temporal lobe epilepsy. J Neurol Neu- tasks after frontal- and temporal-lobe lesions in man. Neu-
rosurg Psychiatry 2004;75:323-326. ropsychologia 1985;23:601-614.
111. Selwa LM, Berent S, Giordani B, et al. Serial cognitive test- 129. Pigott S, Milner B. Memory for different aspects of com-
ing in temporal lobe epilepsy: longitudinal changes with plex visual scenes after unilateral temporal- or frontal-lobe
medical and surgical therapies. Epilepsia 1994;35:743- resection. Neuropsychologia 1993;31:1-15.
749. 130. Pillon B, Bazin B, Deweer B, et al. Specificity of mem-
112. Kimura D. Right temporal-lobe damage. Perception of un- ory deficits after right or left temporal lobectomy. Cortex
familiar stimuli after damage. Arch Neurol 1963;8:264-271. 1999;35:561-571.
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 75

131. Smith ML, Milner B. The role of the right hippocam- 148. Hermann BP, Perrine K, Chelune GJ, et al. Visual con-
pus in the recall of spatial location. Neuropsychologia frontation naming following left anterior temporal lobec-
1981;19:781-793. tomy: a comparison of surgical approaches. Neuropsychol
132. Worsley CL, Recce M, Spiers HJ, et al. Path integration fol- 1999;13:3-9.
lowing temporal lobectomy in humans. Neuropsycholo- 149. Busch RM, Frazier TW, Haggerty KA, et al. Utility of
gia 2001;39:452-464. the Boston Naming Test in predicting ultimate side of
133. Breier JI, Leonard CM, Bauer RM, et al. Quantified vol- surgery in patients with medically intractable temporal
umes of temporal lobe structures in patients with epilepsy. lobe epilepsy. Epilepsia 2005;46:1773-1779.
J Neuroimaging 1996;6:108-114. 150. Hermann B, Davies K, Foley K, et al. Visual confronta-
134. Janszky J, Jokeit H, Kontopoulou K, et al. Functional MRI tion naming outcome after standard left anterior temporal
predicts memory performance after right mesiotemporal lobectomy with sparing versus resection of the superior
epilepsy surgery. Epilepsia 2005;46:244-250. temporal gyrus: a randomized prospective clinical trial.
135. Leritz EC, Grande LJ, Bauer RM. Temporal lobe epilepsy Epilepsia 1999;40:1070-1076.
as a model to understand human memory: the distinction 151. Seidenberg M, Hermann B, Wyler AR, et al. Neuropsycho-
between explicit and implicit memory. Epilepsy Behav logical outcome following anterior temporal lobectomy in
2006;9:1-13. patients with and without the syndrome of mesial tempo-
136. Fleischman DA, Vaidya CJ, Lange KL, et al. A dissoci- ral lobe epilepsy. Neuropsychol 1998;12:303-316.
ation between perceptual explicit and implicit memory 152. Ruff IM, Swanson SJ, Hammeke TA, et al. Predictors of
processes. Brain Cogn 1997;35:42-57. naming decline after dominant temporal lobectomy: age
137. Poldrack RA, Gabrieli JD: Functional anatomy of long- at onset of epilepsy and age of word acquisition. Epilepsy
term memory. J Clin Neurophysiol 1997;14:294-310. Behav 2007;10:272-277.
138. Blaxton, TA: Dissociations among memory measures in 153. Saykin AJ, Stafiniak P, Robinson LJ, et al. Language before
memory impaired subjects: Evidence for a processing ac- and after temporal lobectomy: specificity of acute changes
count of memory. Mem Cogn 1992;20:549-562. and relation to early risk factors. Epilepsia 1995;36:1071-
139. Savage GR, Saling MM, Davis CW, et al. Direct and in- 1077.
direct measures of verbal relational memory following 154. Schwarz M, Pauli E, Stefan H. Model based progno-
anterior temporal lobectomy. Neuropsychologia 2002;40: sis of postoperative object naming in left temporal lobe
302-316. epilepsy. Seizure 2005;14:562-568.
140. Zaidel DW, Esiri MM, Oxbury JM. Effects of surgery in 155. Bell BD, Davies KG, Hermann BP, et al. Confrontation
unilateral medial temporal lobe regions on verbal explicit naming after anterior temporal lobectomy is related to
and implicit memory. Neuropsychiatry Neuropsychol Be- age of acquisition of the object names. Neuropsychologia
hav Neurol 1994;7:104-108. 2000;38:83-92.
141. Zaidel DW, Esiri MM, Beardsworth ED. Observations on 156. Hermann BP, Seidenberg M, Haltiner A, et al. Adequacy
the relationship between verbal explicit and implicit mem- of language function and verbal memory performance in
ory and density of neurons in the hippocampus. Neu- unilateral temporal lobe epilepsy. Cortex 1992;28:423-433.
ropsychologia 1998;36:1049-1062. 157. Giovagnoli AR. Characteristics of verbal semantic im-
142. Billingsley RL, McAndrews MP, Smith ML. Intact percep- pairment in left hemisphere epilepsy. Neuropsychol
tual and conceptual priming in temporal lobe epilepsy: 2005;19:501-508.
neuroanatomical and methodological implications. Neu- 158. Luckhurst L, Lloyd-Jones TJ. A selective deficit for liv-
ropsychol 2002;16:92-101. ing things after temporal lobectomy for relief of epileptic
143. Alessio A, Bonilha L, Rorden C, et al. Memory and lan- seizures. Brain Lang 2001;79:266-296.
guage impairments and their relationships to hippocam- 159. Glosser G, Donofrio N. Differences between nouns and
pal and perirhinal cortex damage in patients with me- verbs after anterior temporal lobectomy. Neuropsychol
dial temporal lobe epilepsy. Epilepsy Behav 2006;8:593- 2001;15:39-47.
600. 160. Bell BD, Seidenberg M, Hermann BP, et al. Visual and
144. Davies KG, Bell BD, Bush AJ, et al. Naming decline auditory naming in patients with left or bilateral temporal
after left anterior temporal lobectomy correlates with lobe epilepsy. Epilepsy Res 2003;55:29-37.
pathological status of resected hippocampus. Epilepsia 161. Hamberger MJ, Tamny TR. Auditory naming and temporal
1998;39:407-419. lobe epilepsy. Epilepsy Res 1999;35:229-243.
145. Sawrie SM, Martin RC, Gilliam FG, et al. Visual confronta- 162. Hamberger MJ, Seidel WT. Auditory and visual nam-
tion naming and hippocampal function: A neural network ing tests: normative and patient data for accuracy, re-
study using quantitative (1)H magnetic resonance spec- sponse time, and tip-of-the-tongue. J Int Neuropsychol
troscopy. Brain 2000;123:770-180. Soc 2003;9:479-489.
146. Ramirez MJ, Schefft BK, Howe SR, et al. Interictal and 163. Fargo JD, Schefft BK, Dulay MF et al. Confrontation nam-
postictal language testing accurately lateralizes language ing in individuals with temporal lobe epilepsy: a quantita-
dominant temporal lobe complex partial seizures. Epilep- tive analysis of paraphasic error subtypes. Neuropsychol
sia 2008;49:22-32. 2005;19:603-611.
147. Giovagnoli AR, Erbetta A, Villani F, et al. Semantic mem- 164. Schefft, BK, Testa SM, Dulay, MF, et al. Preoperative
ory in partial epilepsy: verbal and non-verbal deficits assessment of confrontation naming ability and inter-
and neuroanatomical relationships. Neuropsychologia ictal paraphasia production in unilateral temporal lobe
2005;43:1482-1492. epilepsy. Epilepsy Behav 2003;4:161-168.
76 SECTION I TECHNIQUES

165. Bengner T, Malina T, Lindenau M, et al. Face memory 185. Jones-Gotman M, Zatorre RJ. Olfactory identification
in MRI-positive and MRI-negative temporal lobe epilepsy. deficits in patients with focal cerebral excision. Neuropsy-
Epilepsia 2006;47:1904-1914. chologia 1988;26:387-400.
166. Glosser G, Salvucci AE, Chiaravalloti ND. Naming and rec- 186. Lehrner J, Baumgartner C, Serles W, et al. Olfactory pro-
ognizing famous faces in temporal lobe epilepsy. Neurol dromal symptoms and unilateral olfactory dysfunction are
2003; 61:81-86. associated in patients with right mesial temporal lobe
167. Adolphs R, Tranel D, Damasio H. Emotion recognition epilepsy. Epilepsia 1997;38:1042-1044.
from faces and prosody following temporal lobectomy. 187. Abraham A, Mathai KV. The effect of right temporal
Neuropsychol 2001:15;396-404. lobe lesions on matching of smells. Neuropsychologia
168. Benuzzi F, Meletti S, Zamboni G, et al. Impaired fear pro- 1983;21:277-281.
cessing in right mesial temporal sclerosis: a fMRI study. 188. Zatorre RJ, Jones-Gotman M. Human olfactory discrimina-
Brain Res Bull 2004;63:269-281. tion after unilateral frontal or temporal lobectomy. Brain
169. Meletti S, Benuzzi F, Rubboli G, et al. Impaired facial emo- 1991;114:71-84.
tion recognition in early-onset right mesial temporal lobe 189. West SE, Doty RL. Influence of epilepsy and temporal lobe
epilepsy. Neurology 2003;60:426-431. resection on olfactory function. Epilepsia 1995;36:531-
170. Adolphs R, Tranel D, Damasio H, Damasio A. Fear and 542.
the human amygdala. J Neurosci 1995;15:5879-5891. 190. Exner C, Boucsein K, Lange C, et al. Neuropsychological
171. Fukatsu R, Fujii T, Tsukiura T, et al. Proper name anomia performance in frontal lobe epilepsy. Seizure 2002;1:20-
after left temporal lobectomy: a patient study. Neurol 32.
1999;52:1096-1099. 191. Helmstaedter C, Gleissner U, Zentner J, et al. Neuropsy-
172. Griffith HR, Richardson E, Pyzalski RW, et al. Memory chological consequences of epilepsy surgery in frontal
for famous faces and the temporal pole: functional imag- lobe epilepsy. Neuropsychologia 1998;36:681-689.
ing findings in temporal lobe epilepsy. Epilepsy Behav 192. Helmstaedter C, Kemper B, Elger, CE. Neuropsycholog-
2006;9:173-180. ical aspects of frontal lobe epilepsy. Neuropsychologia
173. Lah S, Grayson S, Lee T, et al. Memory for the past af- 1996;34:399-406.
ter temporal lobectomy: impact of epilepsy and cognitive 193. Auclair L, Jambaqué I, Dulac O, et al. Deficit of prepara-
variables. Neuropsychologia 2004;42:1666-1679. tory attention in children with frontal lobe epilepsy. Neu-
174. Seidenberg M, Griffith R, Sabsevitz D, et al. Recognition ropsychologia 2005;43:1701-1712.
and identification of famous faces in patients with unilat- 194. McDonald CR, Bauer RM, Filoteo JV, et al. Attentional in-
eral temporal lobe epilepsy. Neuropsychologia 2002;40: hibition in patients with focal frontal lobe lesions. J. Clin.
446-456. Exp. Neuropsychol 2005;27:485-503.
175. Tranel D, Damasio H, Damasio AR. A neural basis for 195. Kho KH, Rutten GJ, Leijten F, et al. Working memory
the retrieval of conceptual knowledge. Neuropsychologia deficits after resection of the dorsolateral prefrontal cortex
1997;35:1319-27. predicted by functional magnetic resonance imaging and
176. Savic I, Bookheimer SY, Fried I, Engel J Jr. Olfactory electrocortical stimulation mapping. Case report. J Neuro-
bedside test. A simple approach to identify temporo- surg 2007;106:501-505.
orbitofrontal dysfunction. Arch Neurol 1997;54:162-168. 196. Miotto EC, Bullock P, Polkey CE, et al. Spatial working
177. Eskenazi B, Cain WS, Novelly RA, Mattson R. Odor per- memory and strategy formation in patients with frontal
ception in temporal lobe epilepsy patients with and with- lobe excisions. Cortex 1996;32:613-630.
out temporal lobectomy. Neuropsychologia 1986;24:553- 197. Morris RG, Rowe A, Fox N, et al. Spatial working mem-
562. ory in Asperger’s syndrome and in patients with focal
178. Jones-Gotman M, Zatorre RJ. Odor recognition memory in frontal and temporal lobe lesions. Brain Cogn 1999;41:
humans: role of right temporal and orbitofrontal regions. 9-26.
Brain Cogn 1993;22:182-98. 198. Drane DL, Lee GP, Cech H, et al. Structured cueing on
179. Martinez BA, Cain WS, de Wijk RA, et al. Olfactory func- a semantic fluency task differentiates patients with tem-
tioning before and after temporal lobe resection for in- poral versus frontal lobe seizure onset. Epilepsy Behav
tractable seizures. Neuropsychol 1993;7:351-363. 2006;9:339-344.
180. Rausch R, Serafetinides EA. Specific alterations of olfactory 199. Henry JD, Crawford JR. A meta-analytic review of ver-
function in humans with temporal lobe lesions. Nature bal fluency performance following focal cortical lesions.
1975;255:557-578. Neuropsychol 2004;18:284-295.
181. Henkin RI, Comiter H, Fedio P, O’Doherty D. Defects 200. Jones-Gotman M, Milner B. Design Fluency: The inven-
in taste and smell recognition following temporal lobec- tion of nonsense drawings after focal cortical lesions. Neu-
tomy. Trans Am Neurol Assoc 1977;102:146-150. ropsychologia 1977;15:653-674.
182. Rausch R, Serafetinides EA, Crandall PH. Olfactory mem- 201. Risse GL. Cognitive outcomes in patients with frontal lobe
ory in patients with anterior temporal lobectomy. Cortex epilepsy. Epilepsia 2006;47:87-89.
1977;13:445-452. 202. Suchy Y, Sands K, Chelune, GJ. Verbal and nonverbal flu-
183. Eskenazi B, Cain WS, Novelly RA, Friend KB. Olfactory ency performance before and after seizure surgery. J Clin
functioning in temporal lobectomy patients. Neuropsy- Exp Neuropsychol 2003;25:190-200.
chologia 1983;21:365-374. 203. Upton D, Thompson PJ. General neuropsychological
184. Eichenbaum H, Morton TH, Potter H, Corkin S. Selective characteristics of frontal lobe epilepsy. Epilepsy Res
olfactory deficits in case H.M. Brain 1983;106:459-472. 1996;23:169-177.
CHAPTER 5 NEUROPSYCHOLOGICAL TESTING 77

204. Jones-Gotman M. Localiztion of lesions by psychological 222. Allegri RF, Drake M, Thomson A. Neuropsychological
testing. Epilepsia 1991;32:41-52. findings in patients with middle temporal lobe epilepsy.
205. Davies KG, Maxwell RE, Jennum P, et al. Language func- Rev Neurol 1999;29:1160-1163.
tion following subdural grid-directed temporal lobectomy. 223. Corcoran R, Upton D. A role for the hippocampus in card
Acta Neurologica Scandinavica 1994;90:201-206. sorting? Cortex 1993;29:293-304.
206. McDonald CR, Delis DC, Norman MA, et al. Discriminat- 224. Drake M, Allegri RF, Thomson A. Executive cognitive al-
ing patients with frontal-lobe epilepsy and temporal-lobe teration of prefrontal type in patients with mesial temporal
epilepsy: utility of a multilevel design fluency test. Neu- lobe epilepsy. Medicina (B Aires) 2000;60:453-456.
ropsychol 2005;19:806-813. 225. Giovagnoli AR, Avanzini G. Quality of life and memory
207. Davidson PS, Gao FQ, Mason WP, et al. Verbal flu- performance in patients with temporal lobe epilepsy. Acta
ency, Trail Making, and Wisconsin Card Sorting Test Neurol Scand 2000;101:295-300.
performance following right frontal lobe tumor resection. 226. Hermann BP, Wyler AR, Richey ET. Wisconsin Card Sort-
J Clin Exp Neuropsychol 2007;11:1-15. ing Test performance in patients with complex partial
208. Giovagnoli AR. Relation of sorting impairment to hip- seizures of temporal-lobe origin. J Clin Exp Neuropsychol
pocampal damage in temporal lobe epilepsy. Neuropsy- 1988;10:467-476.
chologia 2001;39:140-50. 227. Horner MD, Flashman LA, Freides D, et al. Temporal lobe
209. McDonald CR, Delis DC, Norman MA, et al. Response epilepsy and performance on the Wisconsin Card Sorting
inhibition and set shifting in patients with frontal lobe Test. J Clin Exp Neuropsychol 1996;18:310-313.
epilepsy or temporal lobe epilepsy. Epilepsy Behav 2005; 228. Klein M, Engelberts NH, van der Ploeg HM, et al. Epilepsy
7:438-446. in low-grade gliomas: the impact on cognitive function
210. McDonald CR, Delis DC, Norman MA, et al. Is impair- and quality of life. Ann Neurol 2003;54:514-520.
ment in set-shifting specific to frontal-lobe dysfunction? 229. Martin RC, Sawrie SM, Gilliam FG, et al. Wisconsin
Evidence from patients with frontal-lobe or temporal-lobe Card Sorting performance in patients with temporal lobe
epilepsy. J Int Neuropsychol Soc 2005;11:477-481. epilepsy: clinical and neuroanatomical correlates. Epilep-
211. Risse GL, Mercer DK, Penovich PE, et al. Cognitive out- sia 2000;41:1626-1632.
come in patients undergoing surgical resection of the 230. Martin RC, Sawrie SM, Edwards R, et al. Investigation
frontal lobe. Neurol 1996;46:A213. of executive function change following anterior tempo-
212. Smith ML, Milner B. Differential effects of frontal-lobe le- ral lobectomy: selective normalization of verbal fluency.
sions on cognition estimation and spatial memory. Neu- Neuropsychol 2000;14:501-508.
ropsychologia 1984;22;697-705. 231. Martin RC, Loring DW, Meador KJ, Lee GP. The effects of
213. Farrant A, Morris RG, Russell T, et al. Social cognition in lateralized temporal lobe dysfunction on formal and se-
frontal lobe epilepsy. Epilepsy Behav 2005;7:506-516. mantic word fluency. Neuropsychologia 1990;28:823-829.
214. McDonald CR, Delis DC, Kramer JH, et al. A componential 232. Moore PM, Baker GA. The neuropsychological and emo-
analysis of proverb interpretation in patients with frontal tional consequences of living with intractable tempo-
lobe epilepsy and temporal lobe epilepsy: Relationships ral lobe epilepsy: implications for clinical management.
with disease-related factors. Clin Neuropsychol 2007;26: Seizure 2002;11:224-230.
1-17. 233. Prevey ML, Delaney RC, Cramer JA, Mattson RH. Com-
215. Kemper B, Helmstaedter C, Elger CE. Kognitive Profile plex partial and secondarily generalized seizure patients:
von prachirurgischen Patienten mit Frontal- und Tempo- cognitive functioning prior to treatment with antiepileptic
rallappenepilepsie. In Epilepsie ’91, edited by D medication. VA Epilepsy Cooperative Study 264 Group.
Scheffner. Reinbeck: Einhorn Presse Verlag, 1992, pp. 345- Epilepsy Res 1998;30:1-9.
350. 234. Upton D, Corcoran R. The role of the right temporal lobe
216. McDonald CR, Bauer RM, Filoteo JV, et al. Episodic mem- in card sorting: a case study. Cortex 1995;31:405-409.
ory in patients with focal frontal lobe lesions. Cortex 235. Lieb JP, Dasheiff RM, Engel J Jr. Role of the frontal lobes in
2006;42:1080-1092. the propagation of mesial temporal lobe seizures. Epilep-
217. McDonald CR, Bauer RM, Grande L, et al. The role of the sia 1991;32:822-837.
frontal lobes in memory: evidence from unilateral frontal 236. Smith ML, Billingsley RL. Neuropsychology of Parieto-
resections for relief of intractable epilepsy. Arch Clin Neu- occipital Epilepsy. In Neuropsychology of Childhood
ropsychol 2001;16:571-585. Epilepsy. Advances in Behavioral Biology, volume 50,
218. Tárnok Z, Barsi P, Gádoros J, et al. Executive dysfunc- New York: Kluwer Academic/Plenum Press, 2002, pp.
tion in frontal lesions and frontal epilepsy. Ideggyogy Sz 113-120.
2006;59:269-280. 237. Jokeit H, Schacher M. Neuropsychological aspects of type
219. Hermann BP, Seidenberg M, Haltiner A, Wyler AR. Mood of epilepsy and etiological factors in adults. Epilepsy Be-
state in unilateral temporal lobe epilepsy. Biol Psychiatry hav 2004;5:S14-20.
1991;30:1205-1218. 238. Salanova V, Andermann F, Rasmussen T, et al. Tumoural
220. Paradiso S, Hermann BP, Blumer D, et al. Impact of de- parietal lobe epilepsy. Clinical manifestations and out-
pressed mood on neuropsychological status in temporal come in 34 patients treated between 1934 and 1988. Brain
lobe epilepsy. J Neurol Neurosurg Psychiatry 2001;70:180- 1995;118:1289-1304.
185. 239. Djordjevic, J, Jones-Gotman, M. Psychological testing in
221. Strauss E, Hunter M, Wada J. Risk factors for cognitive presurgical evaluation of epilepsy. In The Treatment of
impairment in epilepsy. Neuropsychol 1995;9:457-463. Epilepsy, 2nd edition, edited by S Shorvon, E Perucca,
78 SECTION I TECHNIQUES

D Fish and E Dodson. Oxford: Blackwell Science Ltd, 245. Chilosi AM, Brovedani P, Moscatelli M, et al. Neuropsy-
2004, pp. 699-715. chological findings in idiopathic occipital lobe epilepsies.
240. Eriksson K, Kylliäinen A, Hirvonen K, et al. Visual agnosia Epilepsia 2006;47:76-78.
in a child with non-lesional occipito-temporal CSWS. 246. Gulgonen S, Demirbilek V, Korkmaz B, et al. Neuropsy-
Brain Dev 2003;25:262-267. chological functions in idiopathic occipital lobe epilepsy.
241. Guerrini R, Bonanni P, Parmeggiani L, Belmonte A. Epilepsia 2000;41:4054-11.
Adolescent onset of idiopathic photosensitive occipital 247. Luerding R, Boesebeck F, Ebner AJ. Cognitive changes af-
epilepsy after remission of benign rolandic epilepsy. ter epilepsy surgery in the posterior cortex. Neurol Neu-
Epilepsia 1997;38:777-781. rosurg Psychiatry 2004;75:583-587.
242. Mottron L, Mineau S, Décarie JC, et al. Visual agnosia 248. Rorden C, Karnath HO. Using human brain lesions to infer
with bilateral temporo-occipital brain lesions in a child function: A relic from a past era in the fMRI age? Nat Rev
with autistic disorder: a case study. Dev Med Child Neu- Neurosci 2004;5:813-819.
rol 1997;39:699-705. 249. Chaytor N, Schmitter-Edgecombe M. The ecological va-
243. Lee L, Lee K, Chung C. Occipital Lobe Epilepsy: Clinical lidity of neuropsychological tests: a review of the lit-
Characteristics, Surgical Outcome, and Role of Diagnostic erature on everyday cognitive skills. Neuropsychol Rev
Modalities. Epilepsia 2005;46:688-695. 2003;13:181-197.
244. Pfaender M, D’Souza WJ, Trost N, et al. Visual distur- 250. Spooner DM, Pachana NA. Ecological validity in neu-
bances representing occipital lobe epilepsy in patients ropsychological assessment: a case for greater consider-
with cerebral calcifications and coeliac disease: a case se- ation in research with neurologically intact populations.
ries. J Neurol Neurosurg Psychiatry 2004;75:1623-1625. Arch Clin Neuropsychol 2006;21:327-337.
Chapter 6
The Wada Test: Intracarotid Injection
of Sodium Amobarbital to Evaluate
Language and Memory
Brian D. Bell 1,2 , Bruce P. Hermann1 , and Paul Rutecki 1,2
1
Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
2
Department of Neurology, W.S. Middleton Memorial Veterans Hospital, Madison, Wisconsin

䉴 INTRODUCTION, HISTORICAL The Wada procedure has a fascinating history.


PERSPECTIVE AND, While working in his native Japan in the late 1940s, Juhn
UNDERLYING PRINCIPLES A. Wada, MD, saw patients treated with unilateral elec-
tric shock therapy for psychiatric disorders. Striving to
The Wada test or the procedure of intracarotid injection prevent cognitive deficits that could result from bilateral
of sodium amobarbital to evaluate language and mem- convulsions, he arrived at the idea of injecting amobar-
ory is used most often with patients being considered bital into the common carotid artery to anesthetize the
for epilepsy surgery, especially unilateral anterior tem- dominant hemisphere and thereby prevent the spread
poral lobectomy (ATL) for the treatment of intractable of the seizure from the contralateral hemisphere. How-
mesial temporal lobe epilepsy (TLE).1 TLE is among the ever, before unilateral amobarbital injection was first im-
most common forms of epilepsy and the one of the most plemented for this purpose, Wada was presented with a
pharmacoresistant. ATL is a well-established surgery that boy in status epilepticus unresponsive to medication. He
reportedly eliminates disabling seizures in about 70% made a left common carotid amobarbital injection that
of TLE patients and reduces seizure frequency in an- arrested the seizure and had only transitory neurological
other 15%.2–4 The success of ATL in terms of seizure side effects. Intracarotid amobarbital injection for cessa-
outcome and neuropsychological morbidity is depen- tion of unremitting status epilepticus soon was used else-
dent on careful selection of surgery candidates. Mem- where with success and, in fact, its routine use in such
ory and object naming are the cognitive abilities most cases was recommended.11 But most importantly, this
vulnerable to ATL, especially when the surgery is per- early success with unilateral amobarbital injection led
formed on the dominant temporal lobe.5 The Wada test Wada to consider the potential of the procedure for iden-
typically is a component of a comprehensive preopera- tification of the speech dominant hemisphere in brain
tive work up that also includes electroencephalographic surgery candidates.12,13
(EEG)-video monitoring, structural and functional imag- In 1955, during sabbatical years in North America,
ing, and a clinical neuropsychological assessment.6 Ide- Wada introduced what became known as the Wada test
ally, these techniques provide concordant evidence of a to the Montreal Neurological Institute (MNI) and soon
focal seizure locus and rule out risk factors that would published the results of amobarbital injections in mon-
preclude surgery.7–9 keys and in 20 human surgical candidates.14 The latter
The Wada test is a means of studying brain func- were tested because their handedness or seizure pat-
tion unilaterally. Amobarbital (sodium amytal) injection tern raised questions about the speech dominant hemi-
into the right or left internal carotid artery (ICA) creates sphere. This early research clearly demonstrated that
a temporary “pharmacological lesion” or “chemical (1) unilateral amobarbital injection resulted in a con-
lobectomy”10 in the anterior two-thirds of one hemi- tralateral flaccid paralysis and hemianopsia, as well as
sphere. This permits a presurgical evaluation of the rel- other neurological and EEG changes, and (2) dominant
ative contribution of each temporal lobe to vital lan- hemisphere injection typically also resulted in speech ar-
guage and memory functions, with the goal of predicting rest and then the production of dysphasic errors as lan-
level of risk of aphasia and, especially, amnesia, should guage functions gradually returned over the course of a
surgery be performed. few minutes.13,14 The Wada test added to knowledge

79
80 SECTION I TECHNIQUES

of localization of speech functions acquired from the occurrence of memory impairment during the Wada
postmortem findings and provided complementary in- test was not dependent on filling of the posterior cere-
formation to intraoperative speech mapping data.15,16 bral artery (PCA). To better control and understand the
Examination of language dominance via the Wada distribution of the drug, further developments included
test quickly spread to other countries.17–22 Wada and femoral catheterization to replace the common carotid
Rasmussen14 concluded their 1960 paper by recom- injection, the pre-Wada test control angiogram, estab-
mending the amobarbital injection technique “. . . as a lishing an optimal range for the amobarbital dose, and
preliminary to operations in the vicinity of the Syl- a slower rate of injection.27,35,36 After the institution of
vian area in left-handed and ambidextrous patients, and the memory procedure, no cases of postoperative global
in right-handed patients in whom any doubt exists as amnesia were encountered in its early years of use at
to which cerebral hemisphere is dominant for speech” MNI.26,27 In 1962, Milner and colleagues26 concluded
(p. 282). from their preliminary findings that “. . . the amytal tech-
However, clinicians became well aware that lan- nique may be of value in reducing the incidence of
guage was not the only cognitive function that could be memory loss after temporal lobectomy in patients with
disrupted permanently by temporal lobe surgery. For ex- bilateral EEG abnormality.” Its purpose expanded and
ample, during the 1950s, bilateral temporal lobe surgery great variation developed across institutions in the ad-
resulted in global amnesia in the well-known case of ministration and interpretation of both the language and
H.M. memory tasks. But the test continues to be an integral
In addition, in the absence of imaging techniques to part of the pre-ATL work up and to bear Wada’s name,
determine the integrity of the contralateral mesial tempo- in recognition of his pioneering work.37
ral region,23,24 it became readily apparent that, if the con-
tralateral temporal lobe was damaged already, patients
undergoing unilateral ATL could become amnestic;25–27 䉴 METHODS
defined as memory loss for events and other informa-
tion that is disproportionate to other cognitive deficits, SUBJECT PREPARATION
present across modalities, evident in assessment of re-
call and recognition, and quite apparent clinically.28 The Wada test procedures are explained to the patient
Theodore Rasmussen, suggested in 1959 that the Wada hours or days in advance. The safety of the procedure
test might be successfully extended to the assessment and the range and temporary nature of the amobarbital’s
of memory in order to identify patients who would neurological effects should be highlighted to minimize
be at risk for amnesia.27 Brenda Milner, the eminent the patient’s anxiety.37–39 Clinicians also may administer
Canadian neuropsychologist and Rasmussen’s colleague, tasks at this introduction to acquire baseline language
then developed a memory component for the Wada and memory data and for the purpose of assessing ret-
test. She soon determined that failure on free recall rograde memory during the Wada test. Alternatively, the
testing was common during the Wada test, even when baseline data without anesthetization can be acquired
the hemisphere ipsilateral to the lesion was injected. hours after the Wada test has been completed.40 The
Thereafter, recognition memory results became the pri- patient fasts the night before the test. It has been rec-
ority for the procedure.29 Milner, Rasmussen, and col- ommended that, if a generalized seizure occurs within
leagues (Wada had begun a career at the University of the 24 hours preceding the test, the procedure should
British Columbia by this time) quickly made the im- be postponed, although there is no consensus about
portant observation that memory and language perfor- this issue.6,16 See Rausch and Risinger,16 and Dodrill41
mance were dissociable during the Wada test. Aphasia for additional clinical guidelines. The Wada test session
did not necessarily cause poor recognition memory and may be videotaped for the purpose of later review and
memory could be impaired even after nondominant precise scoring.42
hemisphere injection.26,27 Thus, with selection of ap- On the day of the test, the patient is brought to
propriate stimuli, memory assessment during the Wada the radiological suite, bilaterally symmetric EEG scalp
procedure is “designed to anticipate and cope with dys- electrodes are placed, and several minutes of baseline
phasia” (p. 813).30 EEG data are obtained before the procedure begins.
In the first series of MNI patients who were admin- With the patient supine, the right femoral artery is punc-
istered the Wada recognition memory protocol, tempo- tured after local anesthesia and a guide wire and catheter
rary memory impairment was unexpectedly rare (22%) are advanced under fluoroscopic guidance through the
after injections contralateral to the lesion and there were femoral artery and aorta to final placement just distal to
other question marks about memory functioning during the origin of the ICA. Angiography is carried out for ex-
the test.31 These issues led to further Wada test research amination of the vascular anatomy. After ruling out prob-
and refinements at MNI and elsewhere.10,17,27,30,32–34 lematic vascular anomalies and determining whether
This work resulted, for example, in the discovery that there is any excessive interhemispheric cross-flow or
CHAPTER 6 THE WADA TEST 81

filling of the posterior cerebral artery via the posterior memory failure, and so the timing of test administration
communicating artery, the patient is prepared for the is a subjective but critical matter.10 A patient is said to be
amobarbital injections. At different centers, the hemi- confused or obtunded when “unable to maintain atten-
sphere with the epileptic focus or the one without the tion on a single object or person, except transiently.”50
focus is injected first in all cases, the left or right hemi- This state may pass relatively quickly, allowing the test
sphere is injected first, or the order of injection side is to proceed.
alternated across patients.37,43–45 Most centers perform The majority of patients do not show depression or
both injections on the same day, 30–60 minutes apart. euphoria after an amobarbital injection. When such a re-
Others recommend carrying out the two injections on action does occur, depressive reactions are more likely
consecutive days,,15,46,47 but this recommendation has after left hemisphere injection, whereas euphoria or dis-
been questioned.41,43,48,49 inhibition is more likely after injection on the right. But
Before the amobarbital is injected and while still not all investigators report observing a clear lateraliza-
supine, of course, the patient is asked to raise his or tion effect.37
her arms in the air with elbows flexed, with palms Extreme and prolonged emotional reactions that
turned rostrally and fingers spread, and to count out seriously disrupt the Wada assessment appear to oc-
loud. Hemiplegia or hemiparesis quickly becomes ev- cur in less than or equal to 6% of patients undergo-
ident in the contralateral arm, which falls to the bedside. ing the test.38,39 There are no clear predictor variables
The patient often keeps the arm ipsilateral to injection in for extreme emotional reactions, but partial restraint,
the air and it may be guided to the bedside.50 The patient manipulation of the inguinal region for the femoral
is instructed to count because the overlearned nature of catheterization, a predominantly male staff, and
this sequence is easier to resume when speech suddenly amobarbital-induced disinhibition of emotions may all
stops immediately after injection. If normal counting be factors in some cases with a history of sexual
quickly resumes after a prompt, this suggests language trauma.39 In one case, such behavior began or esca-
disruption has not occurred. A potential drawback of the lated after an initial period of adequate cooperation.38
counting instruction is that after a nondominant hemi- It has been suggested that continuous cognitive testing
sphere injection, the patient may perseverate on count- during the entire period of recovery from the drug may
ing despite instructions to discontinue. An alternative is help suppress strong emotional reactions.53 See O’Shea
to begin conversing with the patient before the injec- et al.54 for a novel use of the Wada test in studying disin-
tion. With a nondominant injection, the conversation is hibited behavior and predicting response to frontal lobe
expected to continue, although the patient’s speech is surgery in patients with frontal lobe injury.
likely to be dysarthric and there may be some garru-
lousness. Within seconds after a dominant hemisphere
injection, speech arrest and a period of global apha-
sia is typical. For cases with mixed dominance, where INSTRUMENTATION
both hemispheres contribute to speech, variations in the
prototypical language changes may occur.51 Amobarbital The sodium salt of amobarbital is very lipid soluble
induced deficits are “transient and in continuous resolu- and so it crosses the blood–brain barrier easily and has
tion” (p. viii).37 The nature and resolution of the domi- a rapid anesthetic effect.45 Injection into the ICA via
nant hemisphere language changes are described more a catheter placed with a transfemoral artery approach
fully later in this chapter. causes inactivation of areas in the distribution of the ip-
When the contralateral hemiparesis and expected silateral anterior and middle cerebral arteries and the
EEG changes (see later) are confirmed and it is estab- anterior choroidal artery. Thus, the amobarbital perfuses
lished that the patient is demonstrating an appropri- frontal and temporal lobe language areas and the ante-
ate level of alertness and visual attention, the memory rior one-third of the hippocampus, modeling to a degree
stimuli are presented to the ipsilateral visual field, be- the effects of surgery on the injected hemisphere.45,55
cause presentation of the target stimuli must take into The carotid angiography preceding the Wada test
account gaze deviation and hemianopsia caused by the confirms catheter placement in the ICA rather than the
amobarbital.17,30,37 In some cases, it may be necessary external carotid or vertebral artery, in addition to deter-
to hold open the patient’s eyelids.50 mining whether there is cross-flow of amobarbital into
To avoid the possibility of a false-negative result, the contralateral hemisphere, usually via the anterior
memory testing should be conducted during the height communicating artery, perfusion of the posterior cere-
of the drug effect. It is not necessary to wait for speech bral artery, or atypical neurovascular architecture that
to return before presenting the memory stimuli, as it might result in a dangerous distribution of the drug.56 In
has been demonstrated that registration in memory can regard to the latter issue, a vascular variation in which
occur even when aphasia is present.29,37,43,44,50,52 On there is a persistent trigeminal artery precludes the Wada
the other hand, obtundation can lead to a false-positive test because of the risk of anesthetizing the brain stem.57
82 SECTION I TECHNIQUES

It should be noted that the distributions of the con- frequently reported dosage, with a range of 3–5 cc of so-
trast medium during angiography and the amobarbi- lution and a 1–2 cc/s manual injection speed being most
tal may differ due to differences in injection methods, common. But individual patient variation in pharmaco-
with amobarbital usually less extensively distributed.6,28 dynamics and arterial anatomy are quite important.59
On the other hand, angiography may underestimate the Centers vary in setting criteria for a sufficient anestheti-
extent of bilateral perfusion of the drug when com- zation effect based on tone and grip strength changes
pared to single-photon emission computed tomography and EEG slowing.6
(SPECT).58 Most epilepsy centers use surface EEG during the
The dosage and concentration of amobarbital, the Wada test to monitor the strength and lateralization of
volume of the drug and saline mixture, and the rate the drug effect.6 There are a few different advantages
and method (manual vs. automated injection) of deliv- to simultaneous EEG monitoring.16,60 The EEG can (1)
ery vary widely.6 A faster rate or a greater volume of identify possible seizure activity, (2) reveal bilateral EEG
injection may perfuse a larger area with widespread ef- abnormalities that may coincide with an obtunded state
fects on cognition, whereas a slower rate or smaller vol- resulting from bilateral perfusion, and (3) establish the
ume of injection can lead to a more intense or persistent duration of the amobarbital effect and the window for
drug effect.6,45 Generally speaking, the amount of the testing. Unilateral injection of the amobarbital results in
medication used appears to have been reduced over the a brief period of bilateral EEG delta waves (Fig. 6–1),
past few decades.29 Most centers administer the drug in but afterward the delta activity becomes predomi-
a single bolus, but others use incremental injections as nantly ipsilateral.28,30 The EEG effects typically last for
necessary.29,59 On the basis of a survey of epilepsy cen- 7–8 minutes.29,30,61 Amobarbital may also result in an
ters, Rausch et al.6 reported that 125 mg was the most increase in spike discharges, as revealed by depth

Figure 6–1. (A) Electroencephalographic (EEG) obtained following left intracarotid


injection with 100 mg of sodium amobarbital. The injection started approximately
7 seconds before the start of the EEG record shown in the figure. Note the asymmetry
of fast activity and the development of rhythmic delta activity over the left hemisphere
leads. Delta slowing is also noted in the right frontal central regions. The angiogram
showed minimal cross-filling. Associated with the injection was a flaccid right arm
weakness and global aphasia. The annotation regarding the left arm down refers to
the examiner guiding the arm down. Visual stimuli were presented during the patient’s
aphasia, which showed partial recovery approximately 3.5 minutes after the injection.
Difficulty repeating sentences was present until 6.5 minutes after the injection. At
10 minutes after the injection, full strength and language function had returned. At that
time, the patient recalled 7 of the 8 objects presented without any false-positive
responses for a net score of 7.0.
CHAPTER 6 THE WADA TEST 83

Figure 6–1 (Continued ). (B) EEG obtained 10 seconds following injection of right
internal carotid artery with 100 mg of sodium amobarbital. As with the other injection,
there is increased fast activity and development of rhythmic delta activity over right
hemisphere leads and left frontal regions. Left arm weakness occurred and no
language dysfunction was identified. During the period of weakness, eight different
target objects were presented visually and the patient could name them. At 10
minutes following the injection, the patient identified all eight objects presented and
mistakenly chose one of the sixteen foils as a target object, so that the net score was
7.5. Thus, the EEG during the Wada test documented focal slowing with some
contralateral frontal slowing typically observed following unilateral injection. This Wada
study demonstrated intact memory function of both left and right hemispheres as well
as left hemisphere language dominance. These findings were expected in this
right-handed patient with a cavernous angioma near the left angular gyrus. The
patient has been seizure free for 14 months since a left temporal lobe lesionectomy
performed after intraoperative language mapping.

electrode recordings.62 As implied previously, attention assessing memory after presentation of the stimuli and a
to the EEG helps distinguish global aphasia due to a uni- distracter task, all while the patient still is under the ef-
lateral amytal effect from obtundation that might be due fect of the drug. With discrete item presentation, target
to a temporary bilateral effect and it also helps avoid items are presented during the drug effect and memory
false-negative data by assuring that language and mem- is then assessed about 10–15 minutes later or when the
ory stimuli are being presented while the amobarbital is amobarbital effect has dissipated.
still active.30 Loring and colleagues37 developed the following
protocol that uses discrete presentation of real objects
followed by a language assessment and subsequent
RECORDING TECHNIQUES measurement of recognition memory for the target ob-
jects. Real objects are presented rather than drawings or
“Protocols vary widely in almost every detail.”56 This verbal items so that the stimuli are minimally biased to-
statement holds true for both the language and memory ward either hemisphere and easily encoded. The Loring
aspects of the Wada test. In fact, protocol changes some- et al. procedure, initiated at the Medical College of Geor-
times are made over time even within epilepsy centers. gia, now is used with some variations in a number of
For example, the type of stimuli, the timing of their pre- other epilepsy centers. Prior to the procedure, the patient
sentation, and the assessment technique all vary.6 Wada is informed of its purpose, and is instructed to remem-
memory assessment can involve continuous testing or ber a sentence and line drawings of two common ob-
discrete item presentation. Continuous testing involves jects. After the preparatory work of the neuroradiologists
84 SECTION I TECHNIQUES

is completed, the procedure starts with a single bo- DATA ACQUISITION AND
lus injection of 100 mg amobarbital (5% solution) by DATA ANALYSIS
hand over about a 4-second interval. As the injection is
made, the patient holds his or her hands raised in the air Language
with fingers spread and counts repeatedly from 1 to 20.
Following evaluation for hemiplegia and eye gaze devi- “The dominant hemisphere can be distinguished from
ation, a simple motor command is given. The neuropsy- the nondominant . . . by the appearance of language
chologist then presents eight common objects (utensils, disturbances that in the space of 5–7 minutes repro-
plastic animals, etc.) to the eye field ipsilateral to the in- duce all the known varieties of receptive and expres-
jection, and the patient’s eyelids are held open when sive aphasia . . . ”22 The systematic assessment of object
necessary. After the eight memory stimuli have been naming and the gradual recovery from dysnomia dur-
presented, language tasks are administered: a modified ing the Wada test was described decades ago by Fedio
token test or comprehension test,63 object naming, and Weinberg53 for left hemisphere speech dominant pa-
sentence repetition, and reading. A nonverbal discrimi- tients. No significant naming errors occurred after right-
nation task is also presented. The language and discrim- side injection. After left injection, dysnomia persisted
ination tasks serve as “late memory items,” in contrast to after vocalization ability returned. Complete recovery
the objects, which are “early memory items” presented from dysnomia generally occurred within 5 minutes, al-
during maximal amobarbital effect. though some patients had persisting dysnomia for 6–
The counting, comprehension, naming, and read- 7 minutes. Errors of substitution and omission were
ing performances are scored on a 0 to 2, 0 to 3, or 0 equally common, whereas perseverative errors were
to 4 scale, with zero representing normal function. Free much rarer. Rausch et al.64 also examined language func-
recall and recognition memory are tested after the amo- tioning after each injection in small groups of left and
barbital effect has cleared, as demonstrated by further right TLE patients, all of whom had left hemisphere lan-
brief motor and language testing and EEG normaliza- guage dominance. In this study, the time elapsed until
tion. A yes/no recognition memory test for the target naming and reading abilities returned to baseline was
objects is the primary memory measure. It involves pre- not reported, but it was established that the time to
sentation of the eight objects randomly interspersed with the first correct response for each of these abilities was
sixteen foils. A correct recognition of a target receives a more prolonged for left-side injections. Any deficits af-
score of 1.0. Any false-positive errors are multiplied by ter the right hemisphere injection resolved quickly. An-
0.5 and that number is subtracted from the total num- other finding was a consistent interaction between side
ber correct. False-positive errors are rare. The maximum of injection and side of seizure focus. After left injection,
and minimum scores are 8.0 and -8.0, respectively, with delay to naming and reading lasted longer in right TLE
consistent guessing expected to achieve a score of 0.0. patients. The reverse was true for right-side injection,
Different target objects and foils are used after each in- but to a lesser degree. Similarly, EEG slowing in the
jection. If less than two target objects are recognized hemisphere ipsilateral to the injection was more pro-
after the injection ipsilateral to the planned surgery, longed when the epileptic focus was in the contralat-
the test may be repeated for that side. Memory testing eral hemisphere. Thus, dysfunction in the epileptogenic
for the “late memory items” follows the object mem- hemisphere interfered with behavioral and physiologi-
ory assessment and the data is used to help determine cal recovery of the contralateral, injected hemisphere. In
whether or not an injection is repeated on one side. See fact, Fedio and Weinberg53 cautioned that an injection
Loring et al. (1992)37 for more procedural and scoring contralateral to a hemisphere with diffuse cortical injury
details. can result in several minutes of unresponsiveness.
Over the course of the past two decades, Loring, More recently, Ravdin et al.65 provided a compre-
Meador, Lee, and colleagues have published a wide hensive description of the gradual recovery from global
range of reports based on their procedure that have ex- aphasia following dominant hemisphere injection of
tended knowledge of lateralized cerebral function and either 100 mg or 125 mg of amobarbital, with some pa-
helped secure the Wada test as a standard compo- tients receiving additional incremental injections if nec-
nent of the pre-ATL work up. Their Wada test reports essary to maintain hemiplegia. All 41 patients in this
have covered topics that include optimizing amobarbi- study were judged to be either left hemisphere (95%)
tal dosage and stimulus presentation timing, retrograde or right hemisphere dominant, with no mixed domi-
memory, false-positive recognition memory errors, com- nance cases present. After memory test items were pre-
parison of Wada language findings with cortical stim- sented, the authors tested language functions serially at
ulation mapping, prediction of post-ATL amnesia, and increasing levels of difficulty. For example, the naming
hemispheric specialization for language representation, items included visual confrontation naming and naming
memory, emotional expression, eye gaze mechanisms, to definition, and the range of difficulty for the latter
tactile attention, and motor organization. included “color of the sky” versus “hard outside part
CHAPTER 6 THE WADA TEST 85

of bread.” Comprehension was tested with basic com- operational definition of bilateral speech varies among
mands such as “touch your nose” as well as syntacti- epilepsy centers.68 In addition, the possible contribution
cally complex questions that included, “If the lion was to language of the posterior temporal lobe(s), which is
eaten by the tiger, which animal died?” Items requiring not perfused by the amobarbital, cannot be determined
repetition were interspersed with the naming and com- by Wada testing. This is an issue because early damage
prehension items and ranged from the patient’s name to the anterior temporal lobe may result in either an in-
to “the phantom soared across the foggy heath.” Para- trahemispheric or interhemispheric transfer of language.
phasic and dysnomic errors were monitored. Baseline
testing prior to the Wada procedure determined the high-
Memory
est levels of difficulty that would be employed during
the procedure. Recovery was defined as a successful re- Initially, Wada memory testing emphasized the perfor-
sponse to the most difficult item administered to a par- mance of the hemisphere contralateral to the side of
ticular patient. It is notable that linguistic errors persisted the proposed lobectomy and evaluation of this hemi-
after the return of motor functions, consistent with other sphere remains vital.7,17,30,69–73 Failed memory test-
reports.43,66 In almost 75% of patients, the pattern of lan- ing after amobarbital injection ipsilateral to the seizure
guage recovery was described as a stereotypical progres- focus suggests the contralateral temporal lobe is dys-
sion, in which return of vocalization was followed in turn functional and that surgery might result in amnesia. Such
by naming, comprehension, and then repetition. Thus, contralateral temporal lobe dysfunction could be the re-
muteness typically occurred and was followed by intel- sult of a diffuse or multifocal initial precipitating injury
ligible vocalizations at an average of 3.5 minutes postin- that caused not only a seizure focus in one temporal lobe
jection, although paraphasic errors invariably remained but also dysfunction in the other. Meningitis, encephali-
at that point. The return of imperfect vocalizations was tis, and traumatic brain injuries are examples of this type
followed by full return of naming at 8.5 minutes, com- of injury. Another possible explanation of contralateral
prehension at 10 minutes, and repetition at 12.5 minutes. memory impairment is the deleterious effect of seizures
Ravdin et al.65 noted that anomia often is the most persis- across time.40,74
tent deficit in aphasia due to stroke, whereas a repetition A false-negative Wada test memory result, in which
deficit or conduction aphasia was most persistent until a patient passes memory testing after ipsilateral injection
full recovery from the amobarbital. They hypothesized and then develops severe amnesia after ATL, is rare.6,28
that the latter may represent a specific amobarbital effect However, some patients who show adequate contralat-
on complex polysynaptic neuronal networks integrating eral Wada memory may show significant post-ATL de-
verbal comprehension and verbal expression. Finally, it cline in memory functioning, as opposed to global
is worth noting that two left TLE patients in this study amnesia.75 The possible association between extent of
demonstrated speech arrest after a right hemisphere in- hippocampal resection and postoperative memory de-
jection but in these cases this effect was not considered cline received a good deal of early attention. But poor
evidence of right hemisphere language. One patient was outcome has more to do with the integrity of the ipsilat-
lethargic, abulic, and akinetic as well as mute, the other eral hippocampus.76–80 The ideal result from Wada mem-
initially was obtunded, and both showed rapid recovery ory testing is an intact score for the side contralateral to
of all language functions upon return of vocalizations. the epileptogenic temporal lobe and a relatively low ip-
It was concluded that (1) “patterns representative of the silateral score, in other words, a memory asymmetry. A
classic acquired aphasias are usually present following poor score for the ipsilateral, epileptogenic hemisphere
dominant hemisphere injection and typically show an after an injection contralateral to it indicates a probable
evolution from a global aphasic syndrome to a longer abnormality of the hippocampus planned for resection.
lasting conduction aphasia,” and (2) “recovery of lan- This likelihood is, of course, preferable to resection of
guage functions uncomplicated by linguistic errors is a well-functioning hippocampus.37,70 Loring et al.81 re-
suggestive of nonlinguistic (i.e., motor, attentional) me- ported that left ATL patients who declined greater than
diated aspects of vocal production.” one standard deviation pre- to postsurgery on a verbal
Meador67 discussed some of the considerations in selective reminding test or story memory test had a sig-
interpreting Wada language testing. For example, he nificantly smaller mean Wada memory asymmetry score
pointed out that agitation or obtundation may compli- (left injection score minus right injection score) com-
cate or preclude language assessment and absence of pared to patients who did not decline postsurgically. The
aphasia after both injections can be due to either inade- authors also applied a memory asymmetry cutoff score
quate amobarbital effects or bilateral language represen- of 2.5, where the score for each hemisphere ranges from
tation. When language is in fact bilaterally represented, −8.0 to 8.0, to predict decline at the individual level.
there can be dissociations among various speech func- Patients who had an asymmetry at least this large experi-
tions, but the exact contribution of each hemisphere may enced significantly less decline on the two verbal mem-
be difficult to determine.42,51 It should be noted that the ory measures administered postsurgically compared to
86 SECTION I TECHNIQUES

those who had an asymmetry score less than or equal strated poor memory after amobarbital injection con-
to 2.0. tralateral to the side of proposed surgery. Hamberger
In this study then, neither left nor right hemisphere et al.87 reported that a group of patients with lat-
memory scores in isolation were significantly related to eral TLE had a significantly smaller memory asymmetry
postsurgical memory outcome. It was the combination score compared to a group with mesial TLE, consistent
of functional reserve in the contralateral mesial tempo- with the expectation that a neocortical temporal seizure
ral lobe and the functional adequacy of the ipsilateral focus would interfere less with new learning than a
side that predicted outcome. Thus, when hippocampal mesial temporal focus. On the other hand, Spencer
sclerosis is present in the epileptogenic hemisphere, the et al.86 reported that five of seven patients with mesial
Wada memory score after the contralateral injection is frontal lobe epilepsy demonstrated bilateral Wada mem-
likely to be relatively low.27,49 This finding in combi- ory impairment. They suggested that bilateral failure
nation with an intact contralateral Wada memory score should raise suspicion of mesial frontal epilepsy in pa-
suggests a relatively low risk of significant postsurgery tients who perform normally on clinical memory tests
decline on clinical memory testing and a greater likeli- and are thought to have mesial TLE.
hood of seizure freedom.49,51,77,81–83 Significant memory In a unique criterion validity study, two patients
decline after surgery now often is defined by reliable with global amnesia unrelated to epilepsy were admin-
change index data, which are derived from assessing istered a Wada recognition memory procedure without
nonsurgical patients on two different occasions.84 injection of amobarbital.94 They failed the test by scor-
ing near or below the chance level, supporting the idea
that Wada memory failure in ATL candidates is capable
䉴 APPLICATION of identifying patients who are at risk for a post-ATL
global amnesia. In another study, the Wada test was ad-
HEALTHY SUBJECTS ministered to three patients before they underwent an
amygdalohippocampectomy (2 left, 1 right) and then
Because it is an invasive procedure, the Wada test is not a second time before resection of more tissue on the
carried out on healthy subjects. same side by ATL.95 As would be predicted, all three
patients showed at least a slight decline in Wada mem-
ory performance when the contralateral hemisphere was
DISEASE STATES injected after the initial surgery.
Some patients are refused surgery or undergo a tem-
As noted, the Wada test is used most often in individuals poral lobe surgery in which the hippocampus is spared
with TLE who are candidates for ATL.56 There contin- because they failed memory testing after an ipsilateral
ues to be variation among centers as to whether the test Wada test injection. It cannot be determined how many
is administered to all candidates or only a select group of such cases are actually false positives.51,59 That is,
of patients.6,14,15,23,30,37,41,56,85,86 Some institutions apply while the Wada test can prevent post-ATL amnesia by
it only in cases where cerebral dominance is uncertain identifying patients who should not have surgery, it also
(e.g., left-handers or early left hemisphere damage), clin- probably mistakenly excludes some patients with in-
ical neuropsychological test results bring into question tractable epilepsy from surgery.27,37,51,94 See Simkins-
the laterality or possible bilateralism of memory impair- Bullock29 for an extended discussion of this issue. To
ment, or there is a discrepancy between other measures avoid the scenario where a patient could be unnecessar-
such as EEG and imaging.51 ily excluded from surgery, Loring et al.37 recommended,
“We suggest that the Wada memory results not be con-
sidered as absolute and should be interpreted within the
䉴 VALIDATION OF THE TECHNIQUE entire clinical context of preoperative epilepsy surgery
evaluation.” In some cases where the Wada test results in
There has been very little systematic research on the prolonged obtundation, anomalous data, or some other
relative contribution of the Wada test to clinical decision- question about its reliability, the test is repeated, possi-
making in the neurosurgical referral process.8,9 How- bly with a smaller dose.6,29,96 An alternative is to carry
ever, the validity of the Wada is supported by studies out a selective or superselective Wada test in which a
that have revealed a correlation between Wada mem- vessel that supplies a more limited cerebral territory is in-
ory results and lateralization of the seizure focus, lateral jected. However, this type of injection is associated with
versus mesial temporal lobe localization of the epilepto- greater risks.28,37,97 For a discussion of selective Wada
genic zone, hippocampal volume and pathology, mesial procedures, see Weissenborn et al.98 and Wieser et al.99
temporal glucose metabolism, age of seizure onset, and A false-positive memory finding could result from
post-ATL seizure outcome.23,49,74,77,87–93 For example, procedures in which verbal stimuli such as written words
Rausch et al.90 reported that 63% of TLE patients demon- predominate among the memory test items.40,100–102 This
CHAPTER 6 THE WADA TEST 87

approach conflates language and memory, so that a are not without drawbacks themselves.57 For example,
left hemisphere injection in a patient with left TLE and functional MRI (fMRI) allows observation of changes re-
left hemisphere language dominance may result in an lated to regional blood flow during the performance of
apparent poor contralateral hemisphere memory score. cognitive tasks. It uses widely available technology, has
In fact, the contralateral score may be worse than the received the most research attention, can provide both
ipsilateral memory score, which constitutes a reverse lateralization and localization information, and is the
asymmetry or “wrong-way Wada.” In one study, more leading candidate to replace the Wada test. But it is an
than one-third of the left TLE patients showed this re- expensive procedure that requires a lengthy period of re-
verse asymmetry.101 However, the failure to recognize stricted movement in a confined space on the part of the
the verbal stimuli after the drug effect has worn off could patient and a great deal of data processing.57,109 Magne-
represent left hemisphere aphasia rather than right hemi- toencephalography directly measures neurophysiologi-
sphere amnesia, which would explain the absence of cal activation and is noninvasive, relatively fast, and free
a relationship between Wada data and postoperative of risk. But it has not yet been thoroughly tested as a
memory outcome.101,102 For a memory Wada procedure replacement for the Wada and is not widely available.57
that employed dually encodable objects, the wrong-way
Wada occurred less frequently and was associated with
poor memory outcome, as would be expected.103
䉴 EXAMPLES IN CLINICAL
Because the amobarbital does not typically per-
fuse the posterior hippocampus, the exact effect of in- PRACTICE
tracarotid injection on memory functioning has been a
Andelman et al.7 described a relatively complex left TLE
matter of debate.26,28,30 Some SPECT and depth elec-
trode recordings have led to the conclusion that the case for which Wada data were useful. A 30-year-old,
amobarbital effect on a large part of the temporal lobe left-handed man with a high school education had a his-
produces a functional deafferentation of the posterior tory of early childhood febrile seizures and onset of com-
hippocampus.61,104,105 Moreover, a number of studies re- plex partial seizures at age 22. His seizures were resistant
to polytherapy with antiepilepsy drugs. Video-EEG, ictal
ported no relationship between posterior cerebral artery
SPECT, and MRI were consistent with left TLE. How-
(PCA) filling and memory performance.44,106,107 One
ever, clinical neuropsychological testing revealed both
study found a limited relationship between the presence
verbal and visual memory impairment, which could in-
of PCA filling and Wada memory performance, but the
dicate bilateral dysfunction, as well as a deficit in visual-
authors concluded that lack of PCA filling does not inval-
perceptual ability. The Wada test (125 mg injections)
idate the procedure.108 On the basis of the relationship
was consistent with right hemisphere language domi-
between Wada memory findings and hippocampal imag-
nance, impaired left temporal lobe memory (0/10 cor-
ing and pathology data, Jones-Gotman51 concluded,
rect), and intact right temporal lobe memory (10/10).
“. . . we are succeeding in testing hippocampal function
After a left ATL, the patient was seizure-free except for
with amobarbital memory tests. We infer further that re-
auras of déjà vu and showed no postoperative cogni-
sults from the tests do allow us to predict postoperative
tive decline. The authors concluded that language and
memory loss, although this remains difficult or impossi-
memory functions had transferred early in life to the
ble to prove.”
right hemisphere and in so doing created a “crowding”
effect, with visual-perceptual abilities suffering as a re-
sult. The authors observed, “The critical contribution of
䉴 RELATIVE STRENGTHS AND the Wada test results to neurosurgical decision-making
WEAKNESS COMPARED TO was in its ability to demonstrate intact memory function
in the hemisphere contralateral to surgery. Demonstrated
OTHER MAPPING TECHNIQUES
right hemisphere language and memory functioning al-
lowed a more liberal left hemisphere resection . . . ”
Morbidity is low with the Wada test and its associated
arteriogram.6,14,15,30,41,57 But it is invasive, uncomfort-
able for the patient, and difficult to apply to children,
and interpretation can be complicated by issues such as 䉴 PEARLS AND PITFALLS
agitation, chronic cognitive impairments, or incomplete
sedation.109 In rare cases, it is necessary to physically re- Because there is no standardized Wada test, all of its
strain a patient for a brief period due to disinhibition or aspects vary from center to center, including the num-
agitation.38 It has been predicted, even by Wada himself, ber and type of stimuli presented and the strategies for
that the test eventually will be replaced by less invasive interpreting the data.6,17,29,60 Almost four decades after
procedures.12,29 Some of these techniques can provide the memory component of the Wada test was initiated,
localization as well as lateralization of functions, but they Milner27 wrote “. . . the question of how we define failure
88 SECTION I TECHNIQUES

on these tests remains to be satisfactorily resolved.” This study of language, memory, and other neuropsycholog-
is in line with the cautionary note by Loring et al.37 that ical functions and the careful selection of candidates for
Wada test memory results should not be considered in epilepsy surgery.
isolation.
It recently has been reported that carbonic an-
hydrase inhibitors, which include topiramate and
zonisamide, can reduce the amobarbital effect.110 Barbi- REFERENCES
turate compounds such as phenobarbital also may affect
the cellular uptake of amobarbital.16,59 Because of past 1. Synder PJ, Harris LJ. The intracarotid amobarbital proce-
shortages of amobarbital, other drugs have been used dure: an historical perspective. Brain Cogn 1997;33:18.
as a replacement.111 These include etomidate, propofol, 2. Engel J Jr. A greater role for surgical treatment of epilepsy:
and methohexital, all of which have half-lives shorter why and when? Epilepsy Curr 2003;3:37.
than amobarbital.57 Etomidate is a nonbarbiturate with a 3. Engel J Jr, Wiebe S, French J, et al. Practice parame-
short duration of action that is administered by bolus and ter: temporal lobe and localized neocortical resections for
subsequent infusion to maintain the drug effects. How- epilepsy. Neurol 2003;60:538.
ever, this medication was associated with adrenal insuf- 4. McIntosh AM, Wilson SJ, Berkovic SF. Seizure outcome
after temporal lobectomy: current research practice and
ficiency in critically ill patients (not Wada test patients)
findings. Epilepsia 2001;42:1288.
who received a dose when they required tracheal intuba- 5. Bell BD, Davies KG. Anterior temporal lobectomy, hip-
tion. In one Wada study,112 , about one-third of patients pocampal sclerosis, and memory: recent neuropsycholog-
experienced side effects from propofol that included in- ical findings. Neuropsychol Rev 1998;8:25.
creased muscle tone and twitching and rhythmic move- 6. Rausch R, Silfvenius H, Wieser H-G, et al. Intraarterial
ments or tonic posturing. These were more likely to amobarbital procedures. In Surgical Treatment of the
occur in older patients who received a higher than aver- Epilepsies, edited by J Engel Jr., 2nd edition. Philadelphia:
age dose. Methohexital commonly is used for the Wada Lippincott-Raven, 1993, p. 341.
test in France and South America and has been used suc- 7. Andelman F, Neufeld MY, Fried I. Contribution of neu-
cessfully in the United States.113 This drug is associated ropsychology to epilepsy surgery. Isr J Psychiatry Relat
with a shorter duration of action and less sedation than Sci 2004;41:125.
8. Uijl SG, Leitjen FS, Parra J, et al. What is the current
amobarbital and so two or more successive injections are
evidence on decision-making after referral for temporal
made, with language tested after the first and memory af- lobe epilepsy surgery? A review of the literature. Seizure
ter the second.113 Methohexital does have epileptogenic 2005;14:534.
potential with high doses45,113 and has been in short sup- 9. Uijl SG, Leitjen FS, Arends JB, et al. Decision-making in
ply itself at times.111 In addition, it recently was reported temporal lobe epilepsy surgery: the contribution of basic
that methohexital resulted in a higher memory score af- non-invasive tests. Seizure 2008;17:364.
ter the second, contralateral hemisphere injection in one 10. Klove H, Trites RL, Grabow JD. Intracarotid sodium amytal
group of patients compared to a second group that re- for evaluating memory function. Electroencephalogr Clin
ceived amobarbital.114 Neurophysiol 1970;28:418.
The future of the Wada test has been discussed for 11. Bladin PF. Intra-carotid treatment of status epilepticus.
many years. It is possible that advances in structural Epilepsia 1963;4:151.
12. Wada JA. Youthful season revisited. Brain Cogn 1997;33:7.
and functional imaging, as well as our understanding of
13. Wada JA. Clinical experimental observations of carotid
the relationship between a range of epilepsy variables artery injections of sodium amytal. Brain Cogn 1997;33:11.
and prognosis,73,85,115 , will make the test obsolete rel- Translated from J. Wada: Igaku to Seibutsuqaku 1949;
atively soon.57,97,109,111,116–118 One advantage the Wada 14:221.
test maintains over most other approaches is its ability 14. Wada JA, Rasmussen T. Intracarotid injection of sodium
to directly assess each hemisphere unilaterally.67,116,119 amytal for the lateralization of cerebral speech domi-
And, of course, both its strengths and limitations now nance. J Neurosurg 1960;17:266.
are well understood. But its future does indeed appear 15. Branch C, Milner B, Rasmussen T. Intracarotid sodium
to be growing shorter because of the promise shown amytal for the lateralization of cerebral speech domi-
by fMRI and other noninvasive techniques.109,120–124 The nance. J Neurosurg 1964;21:399.
Wada test will remain widely used until it is established 16. Rausch R, Risinger M. Intracarotid sodium amobarbital
procedure. In Neuromethods (Vol. 17), edited by AA Boul-
that the noninvasive techniques have the ability to pre-
ton, GB Baker, and M Hiscock. Clifton, NJ: Humana, 1990,
dict postsurgical language and memory outcome at the p. 127.
individual level.57,102 The development of these nonin- 17. Klove H, Grabow JD, Trites RL. Evaluation of memory
vasive tests could be hastened by multicenter collabo- functions with intracarotid sodium amytal. Trans Am Neu-
rative research.57,94 Although its future is uncertain, it rol Assoc 1969;94:76-80.
is clear that for more than five decades the Wada test 18. Lansdell H. Laterality of verbal intelligence in the brain.
has made a unique and invaluable contribution to the Science 1962;135(3507):922.
CHAPTER 6 THE WADA TEST 89

19. Perria L. Determination of side of cerebral dominance 39. Masia SL, Perrine K, Westbrook L, et al. Emotional out-
with amobarbital. Arch Neurol 1961;4:173. bursts and post-traumatic stress disorder during intrac-
20. Serafetinides EA, Hoare RD, Driver MV. A modification arotid amobarbital procedure. Neurol 2000;54:1691.
of the intracarotid amylobarbitone test: findings about 40. Diaz-Arrastia R, Frol AB, Garcia MC, et al. Bilateral mem-
speech and consciousness. Lancet 1964;1:249. ory dysfunction in epilepsy surgery candidates detected
21. Serafetinides EA, Hoare RD, Driver MV. Intracarotid by the intracarotid amobarbital procedure (Wada memory
sodium amylobarbitone and cerebral dominance for test). Epilepsy Behav 2002;3:82.
speech and consciousness. Brain 1965;88:107. 41. Dodrill C. Preoperative criteria for identifying eloquent
22. Terzian H. Behavioral and EEG effects of intracarotid brain: intracarotid amytal for language and memory test-
sodium amytal injection. Acta Neurochir 1964;12:230. ing. Epilepsy Surg 1993;4:211.
23. Baxendale SA, Van Paesschen W, Thompson PJ, et al. The 42. Mateer CA, Dodrill C. Neuropsychological and linguis-
relation between quantitative MRI measures of hippocam- tic correlates of atypical language lateralization: evidence
pal structure and the intracarotid amobarbital test. Epilep- from sodium amytal studies. Human Neurobio 1983;2:135.
sia 1997;38:998. 43. Aasly J, Silfvenius H. Evaluation of early and late pre-
24. Rosenow F, Luders H. Presurgical evaluation of epilepsy. sented tasks in the intracarotid amytal test for epileptic
Brain 2001;124:1683. patients. Epilepsy Res 1990;7:155.
25. Baxendale S. Amnesia in temporal lobectomy patients: 44. Carpenter K, Oxbury JM, Oxbury S, et al. Memory for
historical perspective and review. Seizure 1998;7:15. objects presented early after intracarotid sodium amobar-
26. Milner B. Study of short-term memory after intracarotid bital: a sensitive clinical neuropsychological indicator of
injection of sodium amytal. Trans Am Neurol Assoc 1962; temporal lobe pathology. Seizure 1996;5:103.
87:224-226. 45. Fields JA, Troster AI. The sodium amytal (Wada) test: pro-
27. Milner B. Amobarbital memory testing: some personal re- cedural and interpretive considerations. In Handbook of
flections. Brain Cogn 1997;33:14. Neurolinguistics, edited by B Stemmer and HA Whitaker.
28. Jones-Gotman M, Barr WB, Dodrill CB, et al. Controver- New York: Elsevier, 1998.
sies concerning the use of intraarterial amobarbital proce- 46. Bengner T, Haettig H, Merschhemke M, et al. Memory as-
dures. In Surgical Treatment of the Epilepsies, edited by sessment during the intracarotid amobarbital procedure:
J Engel Jr., 2nd edition. Philadelphia: Lippincott-Raven, influence of injection order. Neurol 2003;61:1582.
1993, p. 445. 47. Grote CL, Wierenga C, Smith MC, et al. Wada difference
29. Simkins-Bullock J. Beyond speech lateralization: a review a day makes: interpretive cautions regarding same-day
of the variability, reliability, and validity of the intrac- injections. Neurol 1999;52:1577.
arotid amobarbital procedure and its nonlanguage uses in 48. Meador KJ, Loring DW. The Wada test: controversies, con-
epilepsy surgery candidates. Neuropsychol Rev 2000;0:41. cerns, and insights. Neurol 1999;52:1535.
30. Blume WT, Grabow JD, Darley FL, Aronson AE. Intrac- 49. Perrine K, Westerveld M, Sass KJ, et al. Wada memory dis-
arotid amobarbital test of language and memory be- parities predict seizure laterality and postoperative seizure
fore temporal lobectomy for seizure control. Neurol control. Epilepsia 1995;36:851.
1973;23:812. 50. Lesser RP, Dinner DS, Luders H, et al. Memory for ob-
31. Milner B. Clues to the cerebral organization of memory. jects presented soon after intracarotid amobarbital sodium
In Cerebral Correlates of Conscious Experience, INSERM injections in patients with medically intractable complex
Symposium No. 6, edited by PA Buser and A Rougeul- partial seizures. Neurol 1986;36:895.
Buser. Amsterdam, N. Holland: Elsevier, 1978, p. 139. 51. Jones-Gotman M. Intracarotid amobarbital testing in
32. Acharya JN, Dinner DS. Use of the intracarotid amobarbi- presurgical evaluation of patients with epilepsy. Rev Neu-
tal procedure in the evaluation of memory. J Clin Neuro- ropsychol 1997;7:171.
physiol 1997;14:311. 52. Loring DW, Meador KJ, Lee GP, et al. Stimulus timing ef-
33. Lansdell H, Purnell JK, Laskowski EJ. The relation of in- fects on Wada memory testing. Arch. Neurol 1994;51:806.
duced dysnomia to phoneme frequency. Lang Speech 53. Fedio P, Weinberg LK. Dysnomia and impairment of ver-
1963;6:88. bal memory following intracarotid injection of sodium
34. Milner B. Amnesia following operation on the temporal amytal. Brain Res 1971;31:159.
lobes. In Amnesia, edited by CWM Whitty and OL Zang- 54. O’Shea MF, Saling MM, Berkovic SF. Behavioural sta-
will. New York: Appleton-Century-Crofts, 1966. tus during the intracarotid amobarbital procedure (Wada
35. Rovit R, Gloor P, Rasmussen T. Intracarotid amobarbital in test): relevance for surgical management. J Neurol Neuro-
epileptic patients: a new diagnostic tool in clinical elec- surg Psychiatry 1999;67:549.
troencephalography. Arch Neurol 1961;5:606. 55. Ludemann W, Schneekloth C, Samii M, et al. Arterial sup-
36. Silfvenius H, Fagerlund M, Saisa J, et al. Carotid angiog- ply of the temporo-medial region of the brain: significance
raphy in conjunction with amytal testing of epilepsy pa- for preoperative vascular occlusion testing. Surg Radiol
tients. Brain Cogn 1997;33:33. Anat 2001;23:39.
37. Loring DW, Meador KJ, Lee, GP, et al. Amobarbital Effects 56. Hamberger MJ, Walczak TS. The Wada test: a critical re-
and Lateralized Brain Function: The Wada Test. New York: view. In Recent Advances in Epilepsy (Vol. 6), edited by
Springer, 1992. T Pedley and BS Meldrum. Edinburgh: Churchill Living-
38. de Paola L, Mader MJ, Germiniani FM, et al. Bizarre behav- stone, 1995, p. 57.
ior during intracarotid sodium amytal testing (Wada test). 57. Abou-Khalil B. An update on determination of language
Are they predictable? Arq Neurpsichiatr 2004;62:444. dominance in screening for epilepsy surgery: the Wada
90 SECTION I TECHNIQUES

test and newer noninvasive alternatives. Epilepsia 2007; 76. Andelman F, Kipervasser S, Neufeld MY, et al. Predictive
48:442. value of Wada memory scores on postoperative learning
58. Hietala SO, Silfvenius H, Aasly J, et al. Cerebral hemi- and memory abilities in patients with intractable epilepsy.
sphere perfusion with injection of Tc99m -HM-PAO in pa- J Neurosurg 2006;104:20.
tients with partial epilepsy during amobarbital testing. Eur 77. Davies KG, Hermann BP, Foley KT. Relation between
J Nucl Med 1990;16:683. intracarotid amobarbital memory asymmetry scores and
59. Baxendale SA, Thompson PJ, Savy L, et al. Dose ef- hippocampal sclerosis in patients undergoing anterior
fects on intracarotid amobarbital performance. J Epilepsy temporal lobe resections. Epilepsia 1996;37:522.
1996;9:135. 78. Hermann BP, Wyler AR, Sommes G, et al. Pathologi-
60. Izac S, Banoczi W. The Wada test. Am J Electroneurodi- cal status of the mesial temporal lobe predicts memory
agnostic Technol 1999;39:23. outcome from left anterior temporal lobectomy. Neuro-
61. Gotman J, Bouwer MS, Jones-Gotman M. Intracranial EEG surgery 1992;31:652.
study of brain structures affected by internal carotid injec- 79. Kneebone AC, Chelune GJ, Dinner DS, et al. Intrac-
tion of amobarbital. Neurol 1992;42:2136. arotid amobarbital procedure as a predictor of material-
62. Aasly J, Blom S, Silfvenius H, et al. Effects of amobarbital specific memory change after anterior temporal lobec-
and methohexital on epileptic activity in mesial temporal tomy. Epilepsia 1995;36:857.
lobe structures in epileptic patients: an EEG study with 80. Stroup E, Langfitt J, Berg M, et al. Predicting verbal mem-
depth electrodes. Acta Neurol Scand 1984;70:423. ory decline following anterior temporal lobectomy (ATL).
63. Benton AL, Hamsher K, des, Sivan AB. Multilingual Apha- Neurol 2003;60:1266.
sia Examination: Manual of Instructions, 3rd edition. Iowa 81. Loring DW, Meador KJ, Lee GP, et al. Wada memory
City, IA: AJA Assoc, 1994, p. 24. asymmetries predict verbal decline after anterior temporal
64. Rausch R, Fedio P, Ary CM, et al. Resumption of behavior lobectomy. Neurol 1995;45:1329.
following intracarotid sodium amobarbital injection. Ann 82. O’Rourke DM, Saykin AJ, Gilhool JJ, et al. Unilateral
Neurol 1984;15:31. hemispheric memory and hippocampal neuronal den-
65. Ravdin LD, Perrine K, Santschi Haywood C, et al. Serial sity in temporal lobe epilepsy. Neurosurgery 1993;32:
recovery of language during the intracarotid amobarbital 574.
procedure. Brain Cogn 1997;33:151. 83. Sass KJ, Lencz T, Westerveld M, et al. The neural substrate
66. Rausch R, Fedio P, Ary CM, et al. Resumption of behavior of memory impairment demonstrated by the intracarotid
following intracarotid sodium amobarbital injection. Ann amobarbital procedure. Arch Neurol 1991;48:48.
Neurol 1983;15:31. 84. Martin R, Sawrie S, Gilliam F, et al. Determining reliable
67. Meador KJ. Ambiguous language in Wada evaluations. cognitive change after epilepsy surgery: development
Epilepsia 2004;45(Suppl 4):24. of reliable change indices and standardized regression-
68. Loring DW, Meador KJ, Lee GP, et al. Cerebral language based change norms for the WMS-III and WAIS-III. Epilep-
lateralization: evidence from intracarotid amobarbital test- sia 2002;43:1551.
ing. Neuropsychologica 1990;28:831. 85. Helmstaedter C. Neuropsychological aspects of epilepsy
69. Bell BD, Davies KG, Haltiner AM, et al. Intracarotid amo- surgery. Epilepsy Behav 2004;5:S45.
barbital procedure and prediction of postoperative mem- 86. Spencer DC, Morrell MJ, Risinger MW. The role of the in-
ory in patients with left temporal lobe epilepsy and hip- tracarotid amobarbital procedure in evaluation of patients
pocampal sclerosis. Epilepsia 2000;41:992. for epilepsy surgery. Epilepsia 2000;41:320.
70. Chelune G. Hippocampal adequacy versus functional re- 87. Hamberger M, Walczak TS, Goodman RR. Intracarotid
serve: predicting memory functions following temporal amobarbital procedure memory performance and age at
lobectomy. Arch Clin Neuropsychol 1995;10:413. first risk for seizures distinguish between lateral neocorti-
71. Chiaravalloti ND, Glosser G. Material-specific memory cal and mesial temporal lobe epilepsy. Epilepsia 1996;37:
changes after anterior temporal lobectomy as predicted 1088.
by the intracarotid amobarbital test. Epilepsia 2001;42: 88. Kim H, Yi S. The effect of early versus late onset of tem-
902. poral lobe epilepsy on hemispheric memory laterality: an
72. Grunwald T, Lehnertz K, Helmstaedter C, et al. Limbic intracarotid amobarbital procedure study. J Korean Med
ERPs predict verbal memory after left-sided hippocam- Sci 1997;12:559.
pectomy. NeuroReport 1998;9:3375. 89. Loring DW, Murro AM, Meador KJ, et al. Wada mem-
73. Jokeit H, Ebner A, Holthausen H, et al. Individual predic- ory testing and hippocampal volume measurements in
tion of change in delayed recall of prose passages after the evaluation for temporal lobectomy. Neurol 1993;43:
left-sided anterior temporal lobectomy. Neurol 1997;49: 1789.
481. 90. Rausch R, Babb TL, Engel J Jr, et al. Memory following
74. Jokeit H, Ebner A, Arnold S, et al. Bilateral reduc- sodium amobarbital injection contralateral to hippocam-
tions of hippocampal volume, glucose metabolism, and pal damage. Arch Neurol 1989;46:783.
Wada hemispheric memory performance are related to 91. Roman DD, Beniak TE, Nugent S. Memory performance
the duration of mesial temporal lobe epilepsy. J Neurol on the intracarotid amobarbital procedure as a predictor
1999;246:926. of seizure focus. Epilepsy Res 1996;25:243.
75. van Emde Boas W, Juhn A. Wada and the sodium amy- 92. Sperling MR, Saykin AJ, Glosser G, et al. Predictors of out-
tal test: the first (and last?) 50 years. J Hist Neurosci come after anterior temporal lobectomy: the intracarotid
1999;8:286. amobarbital test. Neurol 1994;44:2325.
CHAPTER 6 THE WADA TEST 91

93. Wyllie E, Naugle R, Chelune G, et al. Intracarotid amo- 108. Morton N, Polkey C, Cox T, et al. Episodic memory dys-
barbital procedure: II. Lateralizing value in evaluation for function during sodium amytal testing of epileptic patients
temporal lobectomy. Epilepsia 1991;32:865. in relation to posterior cerebral artery perfusion. J Clin Exp
94. McGlone J, Black SE, Evans J, et al. Criterion-based va- Neuropsychol 1996;18:24.
lidity of an intracarotid amobarbital recognition-memory 109. Lassonde M, Sauerwein HC, Gallagher A, et al. Neuropsy-
protocol. Epilepsia 1999;40:430. chology: traditional and new methods of investigation.
95. Brahman J, Morris RG. Pre- and postoperative intracarotid Epilepsia 2006;47(Suppl 2):9.
amytal procedure: an assessment of validity. Epilepsy 110. Bookheimer S, Schrader LM, Rausch R, et al. Re-
Behav 2003;4:556. duced anesthetization during the intracarotid amobarbi-
96. Blum DE, Bortz JJ, Ehsan T. Factors affecting degree tal (Wada) test in patients taking carbonic anhydrase-
of sedation after intracarotid amytal injection. J Epilepsy inhibiting medications. Epilepsia 2005;46:236.
1997;10:42. 111. Grote C, Meador K. Has amobarbital expired? Considering
97. Miller DL, Fedio P. Whither the Wada? Radiology 1988; the future of the Wada. Neurol 2005;65:1692.
168:871. 112. Mikuni N, Takayama M, Satow T, et al. Evaluation of ad-
98. Weissenborn K, Ruckert N, Brassel F, et al. A proposed verse effects in intracarotid propofol injection for Wada
modification of the Wada test for presurgical assess- test. Neurol 2005;65:1813.
ment in temporal lobe epilepsy. Neuroradiology 1996;38: 113. Buchtel HA, Passaro EA, Selwa LM, et al. Sodium metho-
422. hexital (Brevital) as an anesthetic in the Wada test. Epilep-
99. Wieser H-G, Muller S, Schiess R, et al. The anterior sia 2002;43:1056.
and posterior selective temporal lobe amobarbital tests: 114. Andelman F, Kipervasser S, Reider-Groswasser II, et al.
angiographic, clinical, electroencephalographic, PET, Hippocampal memory function as reflected by the intrac-
SPECT findings, and memory performance. Brain Cogn arotid sodium methohexital Wada test. Epilepsy Behav
1997;33:71. 2006;9:579.
100. Dodrill C, Ojemann G. An exploratory comparison of 115. Davies KG, Bell BD, Bush AJ, et al. Prediction of verbal
three methods of memory assessment with the intrac- memory loss in individuals after anterior temporal lobec-
arotid amobarbital procedure. Brain Cogn 1997;33:210. tomy. Epilepsia 1998;39:820.
101. Kirsch HE, Walker JA, Winstanley FS, et al. Limitations of 116. Abou-Khalil B, Schlaggar BL. Is it time to replace the
Wada memory asymmetry as a predictor of outcomes after Wada? Neurol 2002;59:160.
temporal lobectomy. Neurol 2005;65:676. 117. Helmstaedter C, Kurthen M. Validity of the Wada test.
102. Meador KJ, Loring DW. Clinical significance of a “wrong- Epilepsy Behav 2002;3:558.
way” Wada: the Wada test for language and memory lat- 118. Diehl B, Luders HO. Temporal lobe epilepsy: when are in-
eralization. Neurol 2005;65:659. vasive recordings needed? Epilepsia 2000;41(Suppl 3):61.
103. Sabsevitz DS, Swanson SJ, Morris GL, et al. Memory out- 119. Baxendale S, Thompson P, Duncan J, et al. Is it time to
come after left anterior temporal lobectomy in patients replace the Wada? Neurol 2003;60:354.
with expected and reversed Wada memory asymmetry 120. Binder JR, Swanson SJ, Hammeke TA, et al. Determination
scores. Epilepsia 2001;42:1408. of language dominance using functional MRI: a compari-
104. McMackin D, Dubeau F, Jones-Gotman M, et al. Assess- son with the Wada test. Neurol 1996;46:978.
ment of the functional effect of the intracarotid sodium 121. Sabsevitz DS, Swanson SJ, Hammeke TA, et al. Use of
amobarbital procedure using co-registered MRI/HMPAO- preoperative functional neuroimaging to predict language
SPECT and SEEG. Brain Cogn 1997;33:50. deficits from epilepsy surgery. Neurol 2003;60:1788.
105. Urbach H, Kurthen M, Klemm E, et al. Amobarbital ef- 122. Rabin ML, Narayan VM, Kimberg DY, et al. Functional MRI
fects on the posterior hippocampus during intracarotid predicts post-surgical memory following temporal lobec-
amobarbital test. Neurol 1999;52:1596. tomy. Brain 2004;127:2286.
106. Perrine K, Devinsky O, Luciano DJ, et al. Correlates of 123. Richardson MP, Strange BA, Thompson PJ, et al. Pre-
arterial-filling patterns in the intracarotid amobarbital pro- operative verbal memory fMRI predicts post-operative
cedure. Arch Neurol 1995;52:712. memory decline after left temporal lobe resection. Brain
107. Smith IM, McGlone J, Fox AJ. Intracarotid amobarbital 2004;127:2419.
memory protocol: Muteness, dysphasia, and variations in 124. Swanson SJ, Sabsevitz DS, Hammeke TA, et al. Functional
arterial distributions of the drug do not affect recognition magnetic resonance imaging of language in epilepsy.
results. J Epilepsy 1993;6:75. Neuropsychol Rev 2007;17:491.
This page intentionally left blank
Chapter 7
Extraoperative Brain Mapping Using
Chronically Implanted Subdural
Electrodes
David E. Friedman1 and James J. Riviello, Jr.2
1
Department of Neurosciences, Winthrop-University Hospital,
Mineola, New York
2
Division of Pediatric Neurology, Department of Neurology and NYU Comprehensive Epilepsy Center,
New York University, New York, New York

䉴 INTRODUCTION cortex; in other words, what to resect and what not


to resect. Historically, the observations of clinical semi-
Epilepsy is the most common chronic neurological dis- ology associated with epileptic seizures first by John
order and affects more than two million people in the Hughlings Jackson7 then by Sir William Richard Gowers8
United States.1 Though newer medications for treat- provided the initial clues in functional localization. For
ment of epilepsy have been introduced over the past instance, the “Jacksonian march” is a classic term de-
two decades, many patients still do not attain seizure scribing the spread of involuntary muscle movements
control2 with medical therapy and other nonpharmaco- associated with progressive focal motor seizures, and in-
logical forms of treatment may be necessary, including dicates cortical involvement of the motor cortex.
resective surgery. In recent years, neuroimaging tech- Stimulation studies have provided much of our
niques have advanced, and many patients who undergo knowledge on brain functioning. Animal studies were
resective epilepsy surgery do not require invasive elec- first performed in the 19th century by Fritsch and Hitzig,9
troencephalographic (EEG) recording studies. However, in which frontal cortical stimulation produced contralat-
for some patients, particularly those without a lesion on eral forelimb movement and resection resulted in fore-
magnetic resonance imagining (MRI) (nonlesional cases) limb weakness in the dog. Roberts Bartholow’s 1874
or when the noninvasive diagnostic evaluation reveals study of the patient Mary Rafferty is widely cited as the
undefined or discordant results, monitoring with inva- first demonstration of the motor excitability of the human
sive EEG is necessary and potentially advantageous.3−6 cerebral cortex, although some question its priority.10
Invasive studies not only allow for a more detailed anal- Functional localization of human motor, sensory, and
ysis with intracranial EEG that is unfiltered by scalp and language-related cortex was initially determined from di-
skull, but also serve to permit functional localization, rect observations made in the operating room with elec-
or the mapping of cortical function, prior to cortical re- trical stimulation during the diagnosis and treatment of
sections. Chronically placed invasive electrodes offer the epilepsy.3 Intraoperative electrical stimulation eventually
possibility of extraoperative testing at the bedside, as op- led to the development of a topographic map of mo-
posed to intraoperative testing in which time limitations tor and somatosensory function of the respective pre-
and anesthesiology considerations are factors that may and postcentral gyri, better known as the motor and
limit the functional mapping. This chapter reviews the somatosensory homunculi.11 The homunculi were intro-
role of extraoperative cortical functional localization, its duced by Wilder Penfield and Theodore Rasmussen as
benefits, and limitations. a means of summarizing the functional organization of
the primary motor and sensory cortices.11 This allowed
the identification of eloquent cortex prior to surgery
䉴 HISTORICAL PERSPECTIVE for the treatment of epilepsy, and helped the neurosur-
geon avoid areas that could lead to functional neurolog-
Intracranial recording and adjunctive brain electrical ical deficits if resected.
stimulation offer information regarding epileptogenic Penfield described the first case of intracranial EEG
localization and preresection assessment of eloquent monitoring for surgical treatment of epilepsy in 1939.12

93
94 SECTION I TECHNIQUES

Over the last several decades, centers around the world


have utilized a technique of implanting various arrays of
electrodes in different portions of the subdural or epidu-
ral space in patients with medication-resistant epilepsy
for both epileptogenic and functional localization in
adults13−26 and children.27−29 These techniques help
clinicians pursue proper surgical management, while de-
creasing the risk of adverse affects of normally function-
ing cortical areas with resection.

䉴 METHODOLOGY

Chronic intracranial EEG recording is indicated for


selective patients with intractable, pharmacoresistant
epilepsy who are undergoing evaluation for epilepsy
surgery. Specific indications for indwelling EEG elec- Figure 7–1. Subdural electrode array placed over the
trodes must be met before pursuing this invasive proce- cortical surface in preparation for chronic extra-
dure. Localization of the epileptogenic zone starts with operative recording.
noninvasive methods including inpatient noninvasive
video-EEG monitoring, MRI, positron emission tomogra-
phy, and single photon emission computed tomography. Subdural strips are made of similar material and contain
If the area of interest is not localized after this evalua- similar electrodes with the electrode array in a single
tion, or if the results of the diagnostic assessment are row of varying number and length. The strips may be
discordant, intracranial EEG by way of subdural strips, inserted through burr holes or the edges of the cran-
grids, and depth electrodes may be employed to iden- iotomy and slipped into the subdural space providing
tify or further delineate epileptogenic regions.4,30 How- coverage beyond the “window” of the craniotomy. Strips
ever, placement of the electrodes should only be per- provide EEG coverage to areas that may not be easily
formed after the evaluation guides the epileptologist to accessed with a craniotomy, such as the orbitofrontal,
a reasonable hypothesis as to the location of the area of occipital, and basal surface of the temporal regions.19,34
epileptogenicity. The hypothesis should be based on not Once placed, it is imperative to test the electrodes in
only the results of the aforementioned studies, but also the operating room with electrocorticography to ex-
supplemented with the clinical history and neurologi- clude technical malfunction and ensure satisfactory ex-
cal examination. The strength of this hypothesis is pro- traoperative recording. It is suboptimal care to discover
portional to the likelihood of success with the invasive that the invasive electrodes, not tested in the operating
evaluation.31 room, are not working afterward when the patient is
in postoperative care or the epilepsy monitoring unit.
The craniotomy is then closed and the patient is eventu-
䉴 SUBJECT PREPARATION ally transferred to a video-EEG monitoring unit for con-
tinuous EEG to assist in determining the epileptogenic
Once a patient is considered for intracranial EEG mon- area(s).
itoring, that patient is taken to the operating room and In addition to localization of areas of epileptogenic-
electrodes are placed. Subdural strip and grid electrodes ity, the chronic indwelling intracranial electrodes allow
and depth electrodes are most commonly used.31−33 for functional localization studies that could be per-
Subdural electrodes are embedded in thin sheets or strips formed in the video-EEG monitoring unit at the bed-
of flexible biocompatible material, such as polyurethane, side. Functional localization techniques with subdural
and implanted in the subdural space (Fig. 7–1). Subdu- electrodes include cortical stimulation. Cortical stimu-
ral grids are typically placed over the cortical surface lation involves introducing small currents of electricity
through a craniotomy. Grid arrays contain disc elec- through individual electrodes with simultaneous obser-
trodes made of stainless steel or platinum alloy that vation for clinical symptoms or signs of either the acti-
measure 2–4 mm in diameter spaced at 1 cm fixed vation or interference with cortical functioning.35 For in-
intervals. Grids are commercially available in a vari- stance, applying electrical stimulation over the patient’s
ety of sizes and shapes, with varying number of elec- motor cortex would produce contralateral motor signs,
trodes in rows and columns, and may contain up to 64 manifesting with involuntary movements. This is referred
contacts points. Their placement requires a craniotomy. to as brain mapping.
CHAPTER 7 EXTRAOPERATIVE BRAIN MAPPING 95

䉴 BACKGROUND ELECTRONIC
PRINCIPLES
Ohm’s law states that voltage (V ) = current (I ) × re-
sistance (R). The current intensity, measured in mil-
liamperes (mA), is determined by the height of each
square wave pulse. The rate of stimulation is inversely
related to the pulse interval, which represents the in-
terval of time between the onset of each consecutive
pulse. The charge (coulombs, C) of each square wave
pulse is obtained by multiplying the current intensity by
the pulse duration. With biphasic stimulation, the charge
correlates with the area under each phase of the pulse.
The charge density represents the charge per unit cross-
sectional area stimulated.
The area of cortical tissue stimulated depends on
both the distance from the stimulating electrode to the
cortex and the amount of current applied. The area stim-
ulated decreases by the square of the distance from the
stimulating electrode. Therefore, only a relatively small
area is stimulated.36
Current density rapidly diminishes with increas-
ing distance from the tissue underlying the stimulat-
ing electrodes.37 Also, significant shunting could occur
through cerebrospinal fluid (CSF) so that the maximum
current density could be reduced as much as tenfold
when traveling from the CSF to the gray matter.38 Elec- Figure 7–2. Example of a constant current stimulator
trode shape also plays a role in the current distribution, device. (Nicolet Cortical Stimulator, CareFusion.)
as a smooth drop off occurs when currents move from
the center of a circular contact, and the drop off is not 1 cm center-to-center distances. Pulse width is set to ei-
smooth when current distributions were estimated at the ther 300 or 500 microsecond (␮s), and the pulse fre-
edges of the circular disk.37 These effects cause theoret- quency ranges from 20 to 50 Hz. An alternating current
ical issues when trying to estimate the amount of cortex is administered for anywhere between 3 and 10 sec-
affected by electrical stimulation. onds, with a subsequent stepwise advancement in cur-
rent. Initial current setting may be as low as 1 mA and
䉴 INSTRUMENTATION, RECORDING gradually increased to the maximum of 15–17.5 mA or
afterdischarge (AD) thresholds, whichever is lower. A
TECHNIQUES, AND DATA
summary of the typical parameters can be found in Ta-
ACQUISITION AND ANALYSIS ble 7–1. The duration of electrical stimulation (train dura-
tion) may vary based on the cortical region or the cortical
Electrical current is delivered to the intracranial elec- function being tested. For instance, when testing motor
trodes with a constant current stimulator device, such as
the Grass S12 stimulator (Grass Technologies, West War-
䉴 TABLE 7–1. TYPICAL STARTING
wick, RI, USA), Nicolet Cortical Stimulator (CareFusion
PARAMETERS FOR EXTRAOPERATIVE
Middleton, WI) (Fig. 7–2) or other commercially avail-
FUNCTIONAL CORTICAL MAPPING
able stimulators. Bipolar stimulation, defined by using
two adjacent electrodes rather than monopolar electrode Pulse Pulse Train
stimulation is typically employed. Monopolar stimula- Current Width Frequency Duration
tion was previously described,39 and involves testing a (mA) (␮s) (Hz) (seconds)
contact of interest by pairing the active electrode to a 1.0a 300 50 3b
distant reference electrode in a silent area. Bipolar stim-
a
ulation offers the advantage of theoretically producing Current is gradually increased to the maximum of 17.5 mA or
afterdischarge threshold, whichever is lower.
a higher current density in a more restricted area than b
Train duration can be administered between 3 and 10 seconds,
monopolar stimulation.39−41 Brief pulses of stimulation depending on the region stimulated, with longer periods of
are applied to indwelling electrodes that are spaced by stimulation typically required for language mapping.
96 SECTION I TECHNIQUES

functioning, relatively shorter periods of stimulation de- strated with cortical stimulation, and include frontal
livery (short train) may be necessary. Longer periods of eye fields,43 supplementary motor area,21,44 supplemen-
electrical stimulation (long train) may be required for tary negative motor areas,45 cingulate gyrus,46 Broca’s
more advanced testing, such as when mapping different aphasia,20,47 Wernicke’s aphasia,48 conduction aphasia,49
aspects of language function. Larger amounts of current, Gerstmann’s syndrome,50 basal temporal language,51
the energy, (an increase stimulation intensity, pulse du- basal temporal vision and alexia,25 auditory naming
ration, or both), may be needed to obtain responses in cortex,52 writing,53 and verbal memory.22
children as the amount of current needed is dependent During extraoperative functional mapping studies,
upon the degree of myelination.42 simultaneous video-EEG is recorded. The continuous
Symptoms during stimulation may include positive display of EEG allows the clinician to determine whether
motor phenomena (tonic or clonic contraction of a mus- stimulation induced an electrographic seizure causing
cle group), negative motor phenomena (inhibition of the patient’s symptoms or signs. It also permits the neu-
voluntary movements of the tongue, fingers, or toes), rologist to view for ADs.
somatosensory phenomena (tingling or numbness of One of the complications of direct cortical stimu-
a part of the body), or language impairment (speech lation is the occurrence of ADs, which are repetitive
hesitation or arrest, anomia, or difficulties with com- epileptiform potentials or rhythmic waveforms that fol-
prehension by either sentence completion or simple low the precipitating electrical stimulus (Fig. 7–3). The
commands).29,31 Language tasks may include having the presence of ADs, defined as spikes, sharp waves, or
patient recite a well-known phrase or poem, or read in rhythmic discharges following cortical stimulation or the
order to assess for speech hesitation or arrest. The pa- induction of an actual habitual seizure may help to local-
tient may also be asked to name an object or picture ize the area of epileptogenicity.54 In addition, the pres-
during stimulation to test for dysnomia. To assess recep- ence of ADs during cortical stimulation indicates the
tive language functioning, the patient could be asked local convulsive threshold.55 However, these findings
to complete simple phrases or follow commands during should be viewed with caution, as the induction of a
stimulation. Signs or symptoms during electrode stimu- habitual seizure following cortical stimulation does not
lation indicate that the cortex underlying the contacts is absolutely guarantee that the seizure did originate from
involved in that particular affected function. A summary that stimulated source (the actual electrodes stimulated),
of the possible responses can be found in Table 7–2. nor does it guarantee that the area stimulated repre-
Clinical syndromes and specific areas have been demon- sents the epileptogenic zone.56 In addition, there is not a

䉴 TABLE 7–2. RESPONSES INDUCED BY CORTICAL STIMULATION

Cortical Regions Responses Comments


Positive Responses:
Induce function (excitation)
Motor cortex Movements SMA: primarily proximal and
tonic contralateral upper
extremity movements
PMA: primarily distal and
clonic contralateral
extremity movements
Somatosensory cortex Dysesthesias
Auditory cortex Buzzing sensation
Visual cortex Phosphenes
Negative Responses:
Inhibit function (depolarization,
inhibition)
Primary negative motor area Inhibilion of voluntory
(lateral convexity) movements
Supplementary negative motor
area (mesial interhemispheric
surface)
Language areas (Broca’s, Speech hesitation, or
Wernicke’s) arrest, anomia,
alexia, difficulties with
comprehension

SMA, supplementary motor area; PMA, primary motor area.


CHAPTER 7 EXTRAOPERATIVE BRAIN MAPPING 97

Figure 7–3. Afterdischarges following direct cortical stimulation.

consistent relationship with the elicitation of ADs from last from one to several hours, and sessions may recur
a given site and the site of origin of habitual seizures.57 over several days.16 Furthermore, the testing involves
The AD threshold is determined in each stimulated intermittent brief pulses of stimulation, rather than
location, if possible. The AD threshold is obtained by continuous stimulation. Prior studies have shown that
sequentially increasing the amount of current applied intermittent stimulation is associated with less cortical
and monitoring the electrocorticogram for ADs. The AD damage than occurs with continuous stimulation, even
threshold varies in different cortical regions, and may when the intermittent periods of stimulation are far
not occur in every stimulated area.58 In adults, func- longer than what typically occurs during extraoperative
tional testing is usually done at subthreshold AD stimula- functional mapping.63,64 Gordon and colleagues evalu-
tion levels, as the clinical response stimulation intensity ated the possibility of stimulation-related tissue damage
threshold is lower than the AD stimulation intensity in three patients who had undergone subdural electrode
threshold level and ADs may interfere with function. stimulation. The histopathology on all three patients re-
However, the clinical response threshold is higher than vealed no evidence that electrical stimulation caused
the AD threshold level in children younger than 4–6 cortical changes.38 Girvin reported a patient who had
years.42 received occipital cortical stimulation via subdural elec-
trodes for a period of 10 years for the purpose of artifi-
cial vision production. No reports of pathologic changes
䉴 SAFTEY AND POTENTIAL were seen after removal of the electrodes.65
ADVERSE EFFECTS A more practical consideration regarding safety and
extraoperative cortical electrical stimulation is the possi-
Established safety parameters are necessary for cortical bility of infection. With indwelling subdural electrodes,
stimulation in humans. Animal studies have indicated the potential for infection exists, as the wires from the
that predictable damage to the cortex can occur when electrodes exit through the dura, skull, and scalp. Most
specific stimulation parameters are exceeded.59 Mecha- centers attempt to minimize the risk with strict anti-
nisms of stimulation-induced tissue injury include accu- septic preventative techniques both in and out of the
mulation of a negative charge at the cathode, production operating room. In addition, tunneling the wires for dis-
of metal ions from the anode, and tissue heat produced tances through the scalp, and vigorous use of prophylac-
by hydrolysis.60 However, most animal models typically tic antibiotics also help lower the potential for infection.
employ continuous stimulation over prolonged periods The incidence of infection was reported to be between
of time, for example, 0.5 hours,61 205 hours,62 and 9 5% and 15%66,67 over a decade ago, but has decreased
hours/day for 4 days.63 In contrast, cortical stimulation over the years to 1% to 5%, with the low end of that
in humans for functional mapping occurs over a brief range being applicable to most recent years.68−70 Infec-
period of time, typically lasting no more than 10 sec- tion may be less frequent with subdural strips than with
onds. Extraoperative individual sessions of testing may grids.42
98 SECTION I TECHNIQUES

Patients generally tolerate the actual stimulation 䉴 EXTRAOPERATIVE VERSUS


without difficulties. However, ipsilateral painful sensa- INTRAOPERATIVE FUNCTIONAL
tions can be induced with either epidural stimulation or
MAPPING
by stimulation of trigeminal nerve fibers accompanying
pia-arachnoid vessels.71 Epidural stimulation may oc-
One of the major advantages of mapping with extra-
cur with reoperation, when scar tissue may preclude
operative subdural arrays is the fact that functional lo-
only subdural positioning. Subthreshold AD stimulation
calization studies can be performed after the patient is
is also done to prevent the induction of a seizure during
transferred from the operating room eventually to the
cortical stimulation, since this often halts the stimulation
epilepsy monitoring unit. This allows for a less restricted
session due to lack of cooperation.
time limit, given that recording time of the intraoperative
Hemorrhage, namely subdural hematoma with sub-
technique is often limited to no more than a few hours.
dural electrodes, is one of the more feared complica-
Extraopertative testing may be repeated if information is
tions of indwelling electrodes. The incidence of life-
not obtained or if the patient has seizures that preclude
threatening hemorrhage has been reduced by using
further testing during a session. Also, the patient should
preoperative angiography to define vascular anatomy
be fully awake during extraoperative testing, allowing
and directly visualizing the cortex through use of a
the examiner to carefully gauge for clinical changes in a
craniectomy rather than a burr hole for electrode place-
relatively stress free environment.
ment. Preoperative evaluation of coagulation and clot-
The disadvantages of extraoperative placement in-
ting parameters is essential, as well as counseling pa-
clude the potential for a direct infection. One study
tients to avoid drugs that inhibit platelet functioning in
found an overall rate of infection of 7.9% and a 5.7% rate
the week before surgery.72 Caution should be used with
per craniotomy.75 Inflammatory responses have been
valproate, since it may interfere with hemostasis and pro-
seen on histopathology to the invasive electrodes. Multi-
mote bleeding.
focal septic meningitis and hypersensitivity meningoen-
Another major, yet rare, risk is edema underneath
cephalitis occurs, more with subdural electrodes rather
the grid electrodes. Herniation has been reported, usu-
than depth electrodes76 ; this may raise the question
ally in the first few days after electrode implantation,
of chronic encephalitis as the cause of the refractory
though the incidence is well less than 1% with modern
epilepsy. Intraoperative testing lengthens the operating
electrodes.72 Because of this potentially life-threatening
time, but otherwise imparts no added risk to the patient.
adverse effect, patients who undergo intracranial moni-
In addition, intraoperative testing allows better anatomic
toring require regular, careful neurological assessments,
resolution since there is direct observation of electrode
as emergent removal of the electrodes might be neces-
position and the recording electrodes can be moved, de-
sary. A recent epilepsy center reported complications
pending on the responses.77
during invasive video-EEG monitoring with subdural
An example of a map created from extraoperative
grid electrodes.73 Complications were encountered in
monitoring and cortical mapping in a patient with a prior
less than 20% of patients. Most were transient and did
resection is presented in Fig. 7–4. It is not possible to
not require treatment. A summary of these complications
obtain all this information from a shorter period of in-
can be found in Table 7–3. Complications were associ-
traoperative monitoring alone: ictal onset and spread of
ated with greater number of electrodes, longer duration
electrographic seizure activity, interictal activity, and cor-
of monitoring, older patient age, left-sided grid insertion,
tical mapping obtained from cortical stimulation (face
and burr holes in addition to the craniotomy. Prophylac-
motor, receptive language, Gertsmann’s syndrome, pic-
tic administration of steroids during monitoring is effica-
ture naming) and visual evoked potential data (courtesy
cious in reducing cerebral swelling and may reduce the
of Masanori Takeoka, Children’s Hospital, Boston). This
risk of potential complications.74
would require an awake craniotomy, which requires

䉴 TABLE 7–3. SUMMARY OF COMPLICATIONS DURING 198 EPISODES OF INVASIVE


VIDEO-ELECTROENCEPHALOGRAPHIC MONITORING WITH SUBDURAL GRID ELECTRODES∗

Complication Frequency n (%) Treatment Required n (%) Permanent Deficit n (%) Death n (%)

Neurological deficit 25 (12.6) 9 (4.6) 3 (1.5)


Infection 24 (12.1) 18 (9.1) 6 (3.0)
Bleeding 6 (3.0) 5 (2.5) 1 (0.5)
Decreased consciousness 5 (2.5) 1 (0.5)
Delirium 1 (0.5) 1 (0.5)

Values are expressed as n or (%).



Modified from Hamer HM, Morris HH, Mascha EJ, et al., (73).
CHAPTER 7 EXTRAOPERATIVE BRAIN MAPPING 99

41
42 33
43 34 PP
1 44 35
2 45 6
FP 3 6 73
4
5 56 474 83
6 6 8 3
55 4 4 0 39
Face motor
7 PO
8 63 4
Receptive language
546
53 2 32 31 30 29 28 27 26 25
61 24 23 22 21 20 19 18 17
5
AT 2 60
51
5
50 9 16 15 14 13
12 11 10 9
58 8 7 6 5
49 4 3 2 1
57
Gerstmann
syndrome TO

8
8

7
6
7
ST

5
Picture

4
naming

3
3

2
MT
2
1
1
VEP max

Figure 7–4. Example of a map of chronically implanted subdural electrodes for


extraoperative monitoring. AT, anterior temporal; MT, mesial temporal; FP, frontal
partial; PP, posterior partial; PO, partial occipital; TO, temporal occipital; ST, superior
temporal; VEP, visual evoked potential.

different anesthetic techniques and is also not suitable 2. Kwan P, Brodie MJ. Early identification of refractory
for many patients, especially younger children.78 epilepsy. N Engl J Med 2000;342:314-319.
3. Penfield W, Jasper H. Electrocorticography. In Epilepsy and
the Functional Anatomy of the Human Brain, edited by W
䉴 CONCLUSIONS Penfield and H Jasper. Boston: Little and Brown and Co,
1954, pp. 692-739.
Across the world, epilepsy surgery centers have been us- 4. Gloor P. Contributions of electroencephalography and
ing intracranial EEG monitoring for patients with phar- electrocorticography to the neurosurgical treatment of the
macoresistant epilepsy. Decisions to use this form of epilepsies. In Neurosurgical Management of the Epilepsies,
monitoring are not systematic, and are based on results edited by DP Purpura, JK Penry, RD Walter, et al. New York:
Raven Press, 1975, pp. 59-106.
of extensive diagnostic evaluation for the individual pa-
5. Engel J Jr. Intracerebral recordings: organization of the hu-
tient, which includes extracranial video-EEG monitoring, man epileptogenic region. J Clin Neurophysiol 1993;10:90-
structural and functional neuroimaging, and neuropsy- 98.
chological evaluation. Invasive monitoring allows for 6. Spencer SS, Sperling MR, Shewmon DA. Intracranial elec-
functional cortical mapping, which provides the treat- trodes. In Epilepsy, edited by J Engel Jr. and TA Pedley.
ing neurologist and neurosurgeon information regarding Philadelphia, New York: Lippincott, Williams and Wilkins,
eloquent cortex, which, in turn, may play a substantial 1998, pp. 1719-1749.
role in surgical planning. 7. Jackson H. A study of convulsions. Trans St Andrews Med
Grad Assoc 1870;3:162-204.
8. Gowers W. Epilepsy and Other Chronic Convulsive Dis-
REFERENCES eases: Their Causes, Symptoms and Treatment. London:
J&A Churchill, 1881.
1. Hauser WA, Annegers JF, Kurland LT. Incidence of epilepsy 9. Devinsky O. Electrical and magnetic stimulation of the cen-
and unprovoked seizures in Rochester, Minnesota: 1935– tral nervous system historical overview. In Advances in
1984. Epilepsia 1993;282:245-246. Neurology: Electrical and Magnetic Stimulation of the Brain
100 SECTION I TECHNIQUES

and Spinal Cord, edited by O Devinsky, A Beric, M Dogali. 28. Ojemann SG, Berger MS, Lettich E, et al. Localization of lan-
New York: Raven Press, 1993, pp. 1-16. guage function in children: results of electrical stimulation
10. Thomas RK, Young CD. A note on the early history of mapping. J Neurosurg 2003;98:465-470.
electrical stimulation of the human brain. J Gen Psychol 29. Schevon CA, Carlson C, Zaroff CM, et al. Pediatric language
1993;120:73-81. mapping: sensitivity of neurostimulation and Wada testing
11. Penfield W, Rasmussen T. The Cerebral Cortex of Man: in epilepsy surgery. Epilepsia 2007;48:539-545.
A Clinical Study of Localization of Function. New York: 30. Henry TR, Ross DA, Schuh LA, et al. Indications and out-
Macmillian, 1950. come of ictal recording with intracerebral and subdural
12. Penfield W. The epilepsies: with a note on radical therapy. electrodes in refractory complex partial seizures. J Clin
New Engl J Med 1939;221:209-218. Neurophysiol 1999;16:426-438.
13. Goldring S. A method for surgical management of focal 31. Benbadis SR, Wyllie E, Bingaman WE. Intracranial elec-
epilepsy, especially as it relates to children. J Neurosurg troencephalography and localization studies. In The Treat-
1978;49:344-356. ment of Epilepsy, edited by E Wyllie, A Gupta, DK Lachh-
14. Lesser RP, Hahn J, Leuders H, et al. The use of chronic sub- wani. Philadelphia: Lippincott, Williams, and Wilkins, 2006,
dural electrodes for cortical mapping of speech. Epilepsia p. 1059.
1981;22:240. 32. Adelson PD, Black PM, Madsen JR, et al. Use of subdu-
15. Lesser RP, Leuders H, Hahn J, et al. Localization of the ral grids and strip electrodes to identify a seizure focus in
speech area in candidates for temporal lobectomy: results children. Pediatr Neurosurg 1995;22:174-180.
of extraoperative studies. Neurology 1982;32:A91. 33. Wilson CL. Intracranial electrophysiological investigation of
16. Lesser RP, Leuders H, Dinner DS, et al. The localization the human brain in patients with epilepsy: contributions to
of speech and writing functions in the frontal language basic and clinical research. Exp Neurol 2004;187:240-245.
area: results of extraoperative cortical stimulation. Brain 34. Cohen-Gadol AA, Spencer DD. Use of anteromedial subdu-
1984;107:275-291. ral strip electrode in the evaluation of medial temporal lobe
17. Goldring S, Gregorie EM. Surgical management of epilepsy epilepsy. Technical note. J Neurosurg 2003;99:921-923.
using epidural recordings to localize the seizure focus. 35. Ojemann GA, Sutherling WW, Lesser RP, et al. Cortical stim-
J Neurosurg 1984;60:457-466. ulation. In Surgical Treatment of the Epilepsies, 2nd edi-
18. Laxer KD, Needleman R, Rosenbaum TJ, et al. Subdu- tion, edited by J Engel. New York: Raven Press, 1993, pp.
ral electrodes for seizure focus localization. Epilepsia 399-414.
1984;25:651. 36. Riviello JJ, Kull L, Troup C, Holmes GL. Cortical stimulation
19. Wyler AR, Ojemann GA, Lettick E, et al. Subdural strip in children: techniques and precautions. Tech Neurosurg
electrodes for localizing epileptogenic foci. J Neurosurg 2001;7:12-18.
1984;60:1195-1200. 37. Nathan SS, Sinha SR, Gordon B, et al. Determination
20. Lesser RP, Luders H, Klem G, et al. Extraoperative corti- of stimulation density distributions generated by electri-
cal functional localization in patients with epilepsy. J Clin cal stimulation of the human cerebral cortex. Electroen-
Neurophysiol 1987;4:27-53. cephalogr Clin Neurophysiol 1993;86:183-192.
21. Luders H, Lesser RP, Dinner DS, et al. Localization of cor- 38. Gordon B, Lesser RP, Rance NE, et al. Parameters for di-
tical function: new information from extraoperative mon- rect cortical electrical stimulation in the human: histopatho-
itoring of patients with epilepsy. Epilepsia 1988;29(Suppl logic confirmation. Electroencephalogr Clin Neurophysiol
2):S56-S65. 1990;75:371-377.
22. Lim SH, Dinner DS, Pillay PK, et al. Functional anatomy of 39. Nathan SS, Lesser RP, Gordon B, et al. Electrical stimula-
the human supplementary sensorimotor area: results of ex- tion of the human cerebral cortex: theoretical approach. In
traoperative electrical stimulation. Electroencephalogr Clin Advances in Neurology: Electrical and Magnetic Stimula-
Neurophysiol 1994;91:179-193. tion of the Brain and Spinal Cord, edited by O Devinsky, A
23. Perrine K, Devinsky O, Uysal S, et al. Left temporal Beric, M Dogali. New York: Raven Press, 1993, pp. 61-86.
neocortex mediation of verbal memory: evidence from 40. Jayakar P. Physiological principles of electrical stimulation.
functional mapping with cortical stimulation. Neurology In Advances in Neurology: Electrical and Magnetic Stimu-
1994;44:1845-1850. lation of the Brain and Spinal Cord, edited by O Devinsky,
24. Schaffler L, Luders HO, Beck GJ. Quantitative comparison A Beric, M Dogali. New York: Raven Press, 1993, pp. 17-28.
of language deficits produced by extraoperative electrical 41. Zangaladze A, Sharan A, Evans J, et al. The effectiveness
stimulation of Broca’s, Wernicke’s, and basal temporal lan- of low-frequency stimulation for mapping cortical function.
guage areas. Epilepsia 1996;37:463-475. Epilepsia 2008;49:481-487.
25. Lesser RP, Arroyo S, Crone N, Gordon B. Motor and sen- 42. Jayakar P, Alvarez LA, Duchowny MS, et al. A safe and
sory mapping of the frontal and occipital lobes. Epilepsia effective paradigm to functionally map the cortex in child-
1998;39(Suppl 4):S69-S80. hood. J Clin Neurophysiol 1992;9:288-293.
26. Mani J, Diehl B, Piao Z, et al. Evidence for a basal temporal 43. Godoy J, Luders H, Dinner DS, et al. Versive eye move-
visual language center: cortical stimulation producing pure ments elicited by cortical stimulation of the human brain.
alexia. Neurology 2008;71:1621-1627. Neurology 1990;40:296-299.
27. Chitoku S, Otsubo H, Harada Y, et al. Extraoperative corti- 44. Fried I, Katz A, McCarthy G, et al. Functional organization
cal stimulation of motor function in children. Pediatr Neu- of the human supplementary motor cortex studied by elec-
rol 2001;24:344-350. trical stimulation. J Neuroscience 1991;11:3656-3666.
CHAPTER 7 EXTRAOPERATIVE BRAIN MAPPING 101

45. Dinner DS, Luders H, Morris HH, et al. Human supple- of prolonged electrical stimulation of striate cortex in
mentary motor area (SMA) electrical stimulation. Epilepsia macaques. Brain Behav Evol 1977;14:46-66.
1987;28a:619. 64. Pudenz RH, Agnew WF, Bullara LA. Effects of electrical
46. Chassagnon S, Minotti L, Kremer S, et al. Somatosensory, stimulation of brain: light- and electron-microscope stud-
motor, and reaching/grasping responses to direct cortical ies. Brain Behav Evol 1977;14:103-125.
stimulation of the human cingulate motor areas. J Neuro- 65. Girven JP. Current status of artificial vision by electro-
surg 2008;109:593-604. cortical stimulation. Can J Neurol Sci 1988;15:58-62.
47. Luders H, Lesser RP, Dinner DS, et al. Comprehension 66. Wyllie E, Luders H, Morris HH, et al. Subdural electrodes in
deficits elicited by electrical stimulation of Broca’s area. the evaluation for epilepsy surgery in children and adults.
Epilepsia 1986;27:598-599. Neuropediatrics 1988;19:80-86.
48. Lesser RP, Luders H, Morris HH, et al. Electrical stimulation 67. Wyllie E, Luders H, Morris HH, et al. Clinical outcome
of Wernicke’s area interferes with comprehension. Neurol- after complete or partial cortical resection for intractable
ogy 1986;36:658-663. epilepsy. Neurology 1987;37:1634-1641.
49. Quigg M, Fountain NB. Conduction aphasia elicited by 68. Johnston JM Jr., Mangano FT, Ojemann JG, et al. Complica-
stimulation of the left posterior superior temporal gyrus. tions of invasive subdural electrode monitoring at St. Louis
J Neurol Neurosurg Psych 1999;96:393-396. Children’s Hospital, 1994-2005. J Neurosurg 2006;105:343-
50. Morris HH, Luders L, Lesser RP, et al. Transient neuropsy- 347.
chologic abnormalities (including Gerstmann’s syndrome) 69. Foutnas KN, Smith JR. Subdural electrode-associated com-
during cortical stimulation. Neurology 1984;34:877-883. plications: a 20-year experience. Stereotact Funct Neuro-
51. Luders H, Lesser RP, Hahn J, et al. Basal temporal lan- surg 2007;85:264-272.
guage area demonstrated by electrical stimulation. Neu- 70. Van Gompel JJ, Worrell GA, Bell ML, et al. Intracranial elec-
rology 1986;36:505-510. troencephalography with subdural electrodes: techniques,
52. Hamberger MJ, Seidel WT, Mckhann II GM, et al. Brain complications, and outcomes. Neurosurgery 2008;63:498-
stimulation reveals critical auditory naming cortex. Brain 505.
2005;128:2742-2749. 71. Lesser RP, Luders H, Klem G, et al. Ipsilateral trigeminal
53. Lubrano V, Roux F-E, Demonet J-F. Writing-specific sites sensory responses to cortical stimulation by subdural elec-
in frontal areas: a cortical stimulation study. J Neurosurg trodes. Neurology 1985;35:1760-1763.
2004;101:787-798. 72. Sperling MR. Intracranial electroencephalography. In Cur-
54. Bernier GP, Richer F, Giard N, et al. Electrical stimula- rent Practice of Clinical Electroencephalography, edited
tion of the human brain in epilepsy. Epilepsia 1990;31:513- by JS Ebersole and TA Pedley. Philadelphia: Lippincott,
520. Williams, and Wilkins, 2003, pp. 646-647.
55. Lesser RP, Gordon B, Fisher R, et al. Subdural grid elec- 73. Hamer HM, Morris HH, Mascha EJ, et al. Complications
trodes in surgery of epilepsy. In Epilepsy Surgery, edited of invasive video-EEG monitoring with subdural grid elec-
by H Luders. New York: Raven Press, 1992, pp. 399- trodes. Neurology 2002;58:97-103.
408. 74. Araki T, Otsubo H, Makino Y, et al. Efficacy of dexam-
56. Lesser RP, Luders H, Klem G, et al. Cortical afterdischarge ethasone on cerebral swelling and seizures during subdu-
and functional response thresholds: results of extraopera- ral grid EEG recording in children. Epilepsia 2006;47:176-
tive testing. Epilepsia 1984;25:615-621. 180.
57. Blume WT, Jones DC, Pathak P. Properties of after- 75. Wiggins GC, Elisevich K, Smith BJ. Morbidity and infec-
discharges from cortical stimulation in focal epilepsies. Clin tion in combined subdyral grid and strip electrode investi-
Neurophysiol 2004;115:982-989. gation for refractory epilepsy. Epilepsy Research 1999;37:
58. Agnew WF, McCreery DB. Considerations for safety in the 73-80.
use of extracranial stimulation for motor evoked potentials. 76. Stephan CL, Kepes JJ, SantaCruz K, et al. Spectrum
Neurosurgery 1987;20:143-147. of clinical and histopathologic responses to intracranial
59. Yeomans JS. Principles of Brain Stimulation. New York: electrodes: from multifocal aseptic meningitis to mul-
Oxford University Press, 1990. tifocal hypersensitivity-type meningovasculitis. Epilepsia
60. Mortimer JT, Shealy CN, Wheeler C. Experimental nonde- 2001;42:895-901.
structive electrical stimulation of the brain and spinal cord. 77. Ojemann GA, Bookheimer SY. Intraoperative functional
J Neurosurg 1970;32:553-559. mapping. In Epilepsy: A Comprehensive Text Book, 2nd
61. Babb TL, Soper HV, Lieb JP, et al. Long-term surface edition, edited by Engel J Jr. and Pedley TA. Philadelphia:
stimulation of the cerebellum in monkeys. J Neurosurg Walters Kluwer/Lippincott Williams & Wilkins, 2008, pp.
1977;47:353-365. 1843-1849.
62. Pudenz RH, Bullara LA, Jaques S, et al. Electrical stimulation 78. Soriano SG, Eldredge EA, Wank FK, et al. The effect of
of the brain III. The neural damage model. Surg Neurosurg propofol on intraoperative electrocorticography and cor-
1975;4:389-400. tical stimulation during awake craniotomies in children.
63. Bartlett JR, Doty RW Sr, Less BB, et al. Deleterious effects Paed Anesth 2000;10:29-34.
This page intentionally left blank
Chapter 8
Brain Mapping in the Operating Room
Sepehr Sani 1 , Edward F. Chang 2 , and Nicholas M. Barbaro 3
1
Department of Neurosurgery, Rush University Medical Center, Chicago, Illinois
2
Department of Neurological Surgery, University of California, San Francisco, California
3
Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana

䉴 INTRODUCTION AND HISTORY extensive mapping time are often time-consuming and
uncomfortable for the patient. More recently, it has been
Classic anatomic descriptions of cortical language and shown by Sanai et al.10 that a tailored craniotomy includ-
motor locations are not sufficient for localizing elo- ing the cortical margins of lesion with negative mapping
quent cortices. Language organization in the dominant of language areas can be used as effectively in tumor
hemisphere is highly variable in the setting of normal resections with morbidity comparable to more extensive
anatomy.1–5 Furthermore, reorganization and plasticity craniotomies and positive mapping.
of language and primary motor cortices due to tumors, Cortical electrical stimulation involves both excita-
epilepsy, or vascular lesions is common.6–8 Although tion and inhibition of input/output fibers, local interneu-
uses of neuronavigation and functional neuroimaging rons, and passing fibers affecting distant sites.11 With mo-
have improved localization, these techniques only reveal tor cortex stimulation, a “positive response” is elicited:
areas that are involved in language or motor function, movement of a body region detected either visually or
but not critical to it. Intraoperative cortical mapping is a with electromyography electrodes. Conversely, language
method to identify essential cortical areas for language mapping involves a “negative response” with cessation
or motor functions, which provides the neurosurgeon of language or induction of errors in naming. Motor map-
with a more precise intraoperative understanding of ac- ping appears to cause direct excitatory depolarization
ceptable resection borders. of the corticospinal tract, whereas speech mapping ap-
Originally described by Penfield and Boldrey9 for pears to result from local interference in the language
identification of the central sulcus, the technique of di- network. Electrical spread of current across the cortex is
rect cortical stimulation mapping involves 50- or 60-Hz dependent on duration and amplitude of stimulation but
frequency intermittent stimulation of small cortical is typically 1 cm or less.
patches while observing motor or language responses
in the patient. Stimulation mapping of eloquent cor-
tex continues to remain the gold standard for providing
in vivo representation of motor and language areas. 䉴 METHODS

The anesthetic technique varies greatly depending on


which cortical functions are to be mapped and whether
䉴 UNDERLYING PRINCIPLES intraoperative electroencephalography (EEG) will be uti-
lized. If language mapping is desired, awake craniotomy
Intraoperative mapping of a hemisphere for localizing with local anesthesia and sedation is required. For motor
language function is performed after lateralization has mapping alone, the operation may be performed under
been determined preoperatively by the amobarbital test. general anesthesia. If EEG will be used as in determin-
Traditionally, in order to ensure the lowest possible risk ing a seizure focus, then preanesthetic sedative medi-
of postoperative neurological deficit, the planned cran- cations and intraoperative anesthetic agents should be
iotomy included the area of proposed lesion resection as avoided. In fact, even brief use of certain inhalational
well as the central sulcus, pre- and postcentral gyri, an- agents can interfere with EEG for several hours. Resec-
gular gyrus, and the anterior superior temporal/inferior tions involving the premotor region and/or primary sen-
prefrontal Broca’s areas. This technique relies on positive sory cortex should be performed in the awake setting.
identification of cortical areas responsible for language Baseline dysphasia or aphasia, significant language bar-
or motor function, as well as negative areas that can be rier, confusion, or decreased level of consciousness are
safely resected. However, the large craniotomy size and contraindications to awake craniotomy. Development of

103
104 SECTION I TECHNIQUES

patient rapport and review of steps of operations be- is asked to hyperventilate before dural opening. Once
forehand are very helpful in reducing anxiety in awake mapping is complete, sedatives are restarted.
patients. For picture naming, it is important to have Intraoperative seizures due to cortical stimulation
the person who will perform the intraoperative testing, have been reported in up to 24% of cases.12,13 Epilepsy
review the material with the patient the day prior to be patients are at particularly increased risk of intraop-
sure that instructions are understood and that the pa- erative seizures due to decreased anticonvulsant lev-
tient can name all objects to be used in the operating els. These seizures, whether focal or general, are usu-
room. ally transient and can be suppressed by application of
local ice-cold Ringer’s solution and a bolus of intra-
venous propofol (1 mg/kg).14 If seizures are prolonged,
they should be controlled with intravenous methohexi-
POSITIONING
tal, 0.5–1 mg/kg. If airway control is of concern, tracheal
intubation may be necessary.
The patient is positioned on the operating table accord-
ing to the usual routine and monitors are attached, an
indwelling urinary catheter is inserted, and oxygen deliv-
ery via nasal cannula is provided. Usually, the semilateral
CRANIOTOMY
position is preferred using a padded roll for back sup-
port. Pressure points are padded. Medications such as
The draping is performed accordingly to allow the anes-
mannitol and dexamethasone, as well as antibiotics are
thesiologist and examiner the full view of the patients
given prior to making the incision based on individual
face as well as contralateral arm and leg for intraoper-
surgeon preference. If frameless stereotactic techniques
ative monitoring. A frontotemporal craniotomy is per-
are needed, the patient will typically be placed in pins.
formed using standard neurosurgical technique with the
Otherwise, a foam headrest is sufficient and avoids po-
assistance of neuronavigation. A tailored craniotomy is
tential injury if the patient has a seizure during the pro-
preferred, including the underlying lesion or seizure
cedure.
focus, along with exposure of surrounding posterior
frontal and superior temporal lobes. After the durotomy,
neuronavigation is used to identify the cortical margins
ANESTHESIA of the lesion. Expected locations of central sulcus and
sylvian fissure are also identified. All margins and land-
For general anesthesia, routine cranial neuroanesthe- marks are labeled on the cortical surface.
sia protocol is followed. Pharmacological paralysis and
high concentrations of inhalation anesthetic agents are
avoided. In awake craniotomy cases, the following reg-
imen is employed: SOMATOSENSORY EVOKED
Before incision, midazolam (2 mg; avoided if EEG POTENTIAL RECORDING
to be recorded) and fentanyl (50–100 ␮g) are adminis-
tered. During surgery, either propofol (50–100 ␮g/kg of Evoked potential recording is used to localize the central
body weight per minute; avoided if EEG to be recorded) sulcus.15,16 This is particularly helpful in cases in which
or dexmedetomidine (0.7–2 ␮g/kg/h) and remifentanil underlying lesions have distorted normal anatomy. The
(0.05–0.2 ␮g/kg/min) are given. Local anesthesia is given technique is performed by placing an electrode strip per-
along the Mayfield pin sites as well as a circumferential pendicular across the proposed location of central sul-
scalp field block. A mixture of 0.5% lidocaine, 0.25% cus. A contralateral suprathreshold median nerve stimu-
Marcaine (bupivacaine hydrochloride), and epinephrine lation is made, an N20 wave is recorded over the hand
(1/200,000) are used. Once the craniotomy is performed, somatosensory cortex, and a phase reversal is observed
intradural injections along either side of the middle across the fissure. This process is performed several
meningeal artery are given. Anesthetic agents are discon- times as the strip is moved along the sensory-motor strip.
tinued at this time until mapping is completed. Painful Recording is usually begun 3 cm above sylvian fissure
portions of the operation, such as early portions of the and repeated a few times as the strip is moved more
exposure and dural opening are managed by patient medial. Two or three recordings are usually needed to
reassurance and transient increase in propofol infusion localize the central sulcus. The hand region is the most
rate. In cases of insufficient sedation, supplementary bo- readily identifiable area and is generally localized to
luses of propofol (0.5 mg/kg) are given and the infu- 4–6 cm above the sylvian fissure. Once identified, the
sion rate may be increased to 125 ␮g/kg/min. Nausea or sulcus is labeled accordingly and the strip is removed.
vomiting can be controlled with intravenous droperidol Evoked potential recording can be performed in the
(1.2–2.5 mg) or metoclopramide (5–10 mg). The patient awake patient or under general anesthesia.
CHAPTER 8 BRAIN MAPPING IN THE OPERATING ROOM 105

ELECTROCORTICOGRAPHY cases.12,13 They are usually transient. Application of ice-


cold Ringer’s solution for 5–10 seconds is often effective
In patients with intractable epilepsy, electrocorticogra- at breaking the seizures. A bolus of intravenous propo-
phy may be used in order to identify the abnormal in- fol (1 mg/kg)14 can also be given as adjunct to stop
terictal spikes. The technique requires the presence of a the seizure. If a prolonged refractory seizure occurs,
neurophysiologists or epileptologist with neurophysiol- it should be controlled with intravenous methohexital,
ogy expertise in the operating room for recording inter- 0.5–1 mg/kg and intubation may be considered if air-
pretation. An electrode grid is laid over the cortical area way is of concern.
of interest and several minutes of electrocortical activity
are recorded. Areas with abnormal interictal spikes are
marked.17,18 Surgical resection of these areas depends
on their functional importance based on motor or lan- LANGUAGE MAPPING
guage mapping. Electrocorticography is also employed
in monitoring of afterdischarges during sensorimotor Language mapping is done in the awake patient when
or language mapping as described in the succeeding the lesion involves the dominant perisylvian frontotem-
section. poral region. Preoperatively, the patient is extensively
counseled on the nature of intraoperative testing and
undergoes a baseline language evaluation as follows:
The patient is asked to count numbers from 1 to 50,
SENSORIMOTOR STIMULATION name objects seen on a computer-generated slide show,
read single words projected on a computer screen se-
The localization of pre- and postcentral gyri is con- quentially, repeat complex sentences, and write words
firmed by direct cortical stimulation after identification and sentences on paper. Language deficits are classified
with evoked potentials. Detailed somatotopic mapping as anomia when the patient is unable to name an ob-
is also possible with cortical stimulation. This technique ject but able to repeat sentences and has fluent speech.
is used when the lesion or seizure focus is close to or Alexia is defined as the retention of the ability to write
involves sensorimotor locations. and spell, but with reading errors. Aphasias may be
Stimulation is performed using an Ojemann Cortical expressive, receptive, or mixed. Mild language errors
Stimulator (Radionics Corp., Burlington, MA). Primary such as paraphasic errors are not considered in resec-
sensory cortex is mapped with the patient awake. Stim- tion planning. Intraoperative language mapping is con-
ulation parameters of 1–2 mA total, with a frequency traindicated in patients with significant language deficits.
of 60 Hz and a pulse duration of 1 ms are used. Corti- Intraoperatively, Broca’s area is identified by
cal patches of 1 cc are stimulated sequentially with rest stimulation-induced speech arrest. Mapping is initiated
periods between stimulations. The probe is applied to at stimulation parameters of 1.5–6 mA, frequency of
the cortex for short 1–2 seconds duration and the pa- 60 Hz, and a pulse duration of 1 ms. Once identified,
tient is asked to report the onset and location of any cortical patches of 5 mm are stimulated sequentially with
perceived paresthesias. Stimulation starts in the supra- rest periods between stimulations. The probe is applied
sylvian portion of the sensory gyrus and advanced su- to the cortex for 1–2 seconds and the patient is moni-
periorly, thereby sequentially identifying the tongue, lip, tored. Each cortical patch is tested three times.
and hand sensory areas. Electrocorticography is monitored to determine the
If the operation is being performed under general threshold for afterdischarges. It is important to keep
anesthesia, only motor mapping is possible.17 Stimula- all stimulation below this threshold and meticulously
tion parameters remain the same, but usually a higher monitor for afterdischarges as they can produce false
threshold of initial stimulation is used (3 mA). Stimula- localizing results during mapping. For each site, the
tion intensity increases until contralateral movement is patient is tested for counting errors, object naming er-
observed by the anesthesiologist or the examiner. Am- rors, and word reading errors as they are presented
plitudes greater than 18 mA are not recommended in on a computer-generated slide show. A cortical patch
motor cortex stimulation. Congruent with sensory map- is considered positive for language function if the pa-
ping, stimulation is initiated in the suprasylvian region in tient is unable to count, name objects, or read words
1 cc patches and moves superiorly along the gyrus un- two out of three stimulation times.8 Positive sites are la-
til somatotopic mapping of tongue, lips, thumb, hand, beled by sterile numbers on the cortex and marked us-
and arm are obtained sequentially. If mapping of motor ing neuronavigation. Usually, no more than 25–30 sites
leg region is needed, stimulation is given through a strip are tested around the intended resection site to delin-
electrode that is inserted in the interhemispheric fissure. eate the positive language areas. All positive language
Seizures, focal and general, can occur during mo- areas must be preserved during resection with a margin
tor mapping and have been reported in up to 24% of of 1 cm. In anterior temporal resections of the dominant
106 SECTION I TECHNIQUES

hemisphere, resections within 2 cm of a positive lan-


guage area, particularly stimulation-induced anomia, will
produce a mild but identifiable general language deficit
observed on an aphasia battery administered 1 month
after the operation.19
If all tested areas are negative for language errors,
wider cortical exposure is not necessary and the resec-
tion can be carried out based on delineated margins of
the lesion or seizure focus. The cortical incision is made
in a “silent” area first and resection is carried out. Subcor-
tical stimulation may be performed to preserve essential
fibers. Once mapping is completed, additional anesthetic
agent may be given for patient comfort.
It may be difficult to distinguish the mechanism
of speech arrest during stimulation of the inferior pre-
frontal cortex as this area is intimately involved in both Figure 8–1. Speech and motor mapping during
language and motor function. Speech arrest can be awake craniotomy for epilepsy surgery. Left lateral
hemisphere with labels (1, chin tingling; 2, lip tingling;
attributed to a stimulus disturbance of language func-
3, lower lip movement; 4, lower lip and jaw
tion or arrest of motor activity.20 Indeed, a combined movement; 5 and 6, tongue tingling; 7, speech
language and motor function in these cortical areas has arrest). Syl = sylvian fissure.
been suggested.21–23 These areas, when encountered,
should be marked as eloquent cortex and preserved,
as their resection will lead to postoperative language 䉴 EXAMPLE IN CLINICAL PRACTICE
deficits.
A 54-year-old woman presented with a long history
of intractable complex partial seizures. Over the last 2
years, her seizures increased in frequency and resulted
䉴 POSTOPERATIVE OUTCOME in word finding difficulties, anomia, and alexia dur-
ing a prolonged postictal period resulting in significant
After the operation, the patient is maintained on high functional disability. Her MRI revealed atrophy of the
normal levels of anticonvulsants. Steroids are used in left mesial temporal lobe structures, and positron emis-
cases of tumor resections for control of vasogenic sion tomography imaging demonstrated left temporal
edema. In cases involving resection around the language hypometabolism. Her intracarotid injection of amobarbi-
areas, approximately 25%24 of patients exhibit temporary tal test (Wada examination) showed left language domi-
worsening of speech function at 1 week after surgery. nance. She was therefore a good candidate for left tem-
This is particularly seen in anterior temporal lobe resec- poral lobectomy. In order to protect language function,
tion cases where seizure focus or lesion resections are an awake craniotomy was performed with speech and
carried up to 1 cm away from the positively identified motor mapping with electrical cortical stimulation. Fig-
language areas. In longer term follow-ups, however, in ure 8–1 shows the left lateral exposure with frontal,
review of 250 patients with language area glioma resec- temporal, and parietal exposure. Numbers 1–6 were
tion at 6 months, only 1.6% of surviving patients exhib- placed over the sensorimotor strip. Speech mapping re-
ited a persistent language deficit.10,24 vealed a speech arrest site in the posterior frontal op-
Resection of lesions involving the supplementary erculum. No anomia sites were found in the temporal
motor cortex results in a contralateral hemiparesis in lobe cortex. Intraoperative electrocorticography showed
80–85% of cases in the immediate postoperative period. epileptiform activity in the amygdala and hippocampal
If the primary motor area and underlying corticospinal depth electrodes, with spread to the lateral cortical sur-
white matter tracts are preserved, however, the hemi- face. Therefore, a standard anatomic resection was car-
paresis recovers fully in 2–4 weeks.17 Resections involv- ried out. Postoperatively, she had completely preserved
ing the premotor upper or lower extremity areas should language function and had excellent long-term seizure
be performed with the patient awake. In our experi- control.
ence, with repeated contralateral motor examinations,
premotor region resections can be carried out until the
motor strength is diminished to no less than three-fifths REFERENCES
in the contralateral extremity. This paresis resolves in
several weeks to months with aggressive postoperative 1. Mogilner A, Grossman JA, Ribary U, et al. Somatosen-
therapy. sory cortical plasticity in adult humans revealed by
CHAPTER 8 BRAIN MAPPING IN THE OPERATING ROOM 107

magnetoencephalography. Proc Natl Acad Sci USA 1993;90: multichannel electromyographic recording. J Neurosurg
3593-3597. 1999;91:922-927.
2. Buonomano DV, Merzenich MM. Cortical plasticity: from 14. Sartorius CJ, Berger MS. Rapid termination of intraopera-
synapses to maps. Annu Rev Neurosci 1998;21:149- tive stimulation-evoked seizures with application of cold
186. Ringer’s lactate to the cortex: technical note. J Neurosurg
3. Sanes JN, Donoghue JP. Plasticity and primary motor cor- 1998;88:349-351.
tex. Annu Rev Neurosci 2000;23:393-415. 15. Lueders H, Lesser RP, Hahn J, et al. Cortical somatosen-
4. Feldman DE, Brecht M. Map plasticity in somatosensory sory evoked potentials in response to hand stimulation. J
cortex. Science 2005;310:810-815. Neurosurg 1983;58:885-894.
5. Lee HW, Shin JS, Webber WRS, et al. Reorganisation of 16. Wood CC, Spencer DD, Alison T. Localization of human
cortical motor and language distribution in human brain. J sensorimotor cortex during surgery by cortical surface
Neurol Neurosurg Psychiatry 2009;80:285-290. recording of somatosensory evoked potentials. J Neuro-
6. Wunderlich G, Knorr U, Herzog H, et al. Precentral glioma surg 1988;68:99-111.
location determines the displacement of cortical hand rep- 17. Berger MS. Lesions in functional cortex and subcortical
resentation. Neurosurgery 1998;42:18-26. white matter. Clin Neurosurg 1994;41:444-463.
7. Duffau H, Denvil D, Capelle L. Long term reshaping of lan- 18. Plicher WH, Silbergel DL, Berger MS, et al. Intraopera-
guage, sensory, and motor maps after glioma resection: a tive electrocorticography during tumor resection: impact
new parameter to integrate in the surgical strategy. J Neurol on seizure outcome in patients with gangliogliomas. J Neu-
Neurosurg Psychiatry 2002;72:511-516. rosurg 1993;78:891-902.
8. Ojemann G, Ojemann J, Lettich E, et al. Cortical language 19. Ojemann GA, Dodrill CB. Verbal memory deficits after left
localization in left dominant hemisphere. An electrical stim- temporal lobectomy for epilepsy. Mechanism and intraop-
ulation mapping investigation in 117 patients. J Neurosurg erative prediction. J Neurosurg 1985;62:101-107.
1989;71:316-326. 20. Lueders H, Lesser RP, Dinner DS, et al. Inhibition of mo-
9. Penfield W, Roberts L. Speech and Brain Mechanisms. tor activity by elicited electrical stimulation of the human
Princeton, NJ: Princeton University Press, 1959. cortex. Epilepsia 1983;24:519.
10. Sanai N, Mirzadeh Z, Berger MS. Functional outcome af- 21. Kimura D. Left-hemisphere control of oral and brachial
ter language mapping for glioma resection. N Engl J Med movements and their relation to communication. Philos
2008;358:18-27. Trans R Soc Lond (Biol) 1982;298:135-149.
11. Ranck JB. Which elements are excited in electrical stimula- 22. Liberman AM, Cooper FS, Shankweiler DP, et al. Perception
tion of mammalian central nervous system: a review. Brain of the speech code. Psychol Rev 1967;74:431-461.
Res 1975;98:417-440. 23. Ojemann GA. Brain organization for language from the per-
12. Sartorius CJ, Wright G. Intraoperative brain mapping in a spective of electrical stimulation mapping. Behav Brain Sci
community setting: technical considerations. Surg Neurol 1983;6:189-206.
1997;47:380-388. 24. Haglund MM, Berger MS, Shamseldin M, et al. Cortical lo-
13. Yingling CD, Ojemann S, Dodson B, et al. Identification calization of temporal lobe language sites in patients with
of motor pathways during tumor surgery facilitated by gliomas. Neurosurgery 1994;34:567-576.
This page intentionally left blank
Chapter 9
Anesthesia for Brain
Mapping Surgery
Nicholas P. Carling1 , Chris D. Glover 1 , Daryn H. Moller 2 , and Ira J. Rampil 2
1
Department of Pediatrics (Anesthesiology), Texas Children’s Hospital, and Baylor College of Medicine,
Houston, Texas
2
Department of Anesthesiology, University at Stony Brook, Stony Brook, New York

䉴 INTRODUCTION stimulation threshold for somatic motor mapping. Suc-


cessful mapping requires the anesthesiologist be aware
Brain mapping techniques are utilized during neurosur- of the aforementioned intraoperative plan, and cog-
gical procedures to precisely identify pathological tissue nizant of the electrophysiological and behavioral ef-
while trying to preserve viable functional cortex. Proce- fects of anesthetic agents in the context of neurosurgical
dures include surgical resections of lesions lying close to mapping. Epilepsy surgery on the dominant hemisphere
or within eloquent tissue such as motor, sensory, or vi- using intraoperative mapping to identify seizure foci
sual cortex. They also include resection of epileptogenic and speech areas has the most stringent requirements
foci and placement of electrodes to treat dyskinesias and for anesthetic management, whereas motor area map-
Parkinsonism via deep brain stimulation. Currently no ping has the least. The evolution of techniques such as
prospective randomized trials have determined an op- magnetoencephalography and functional magnetic res-
timal anesthetic for brain mapping procedures, and the onance imaging for preoperative evaluation and local-
ever-burgeoning retrospective analyses have created in- ization of seizure foci and eloquent cortex has, at some
stitutionally developed protocols without consensus. In institutions, reduced the reliance on pristine neural ac-
fact, the anesthetic maintenance choice has little bear- tivity and allow a wider range of anesthetic agents in the
ing on neurosurgical outcomes when given properly.1 management of these patients. This chapter will discuss
While each has advantages and drawbacks, the one cho- the pharmacology of commonly used anesthetic agents,
sen depends on a variety of factors including familiarity their effects on neural electrophysiological monitoring,
of the anesthesia provider with a particular technique, and their administration during anesthetic protocols that
individual patient characteristics such as difficulty of air- may be utilized when intraoperative brain mapping or
way management or ability to tolerate an awake proce- neurocognitive testing are performed.
dure, the location and type of lesion to be resected, and
the type of electrophysiological monitoring to be used
䉴 EFFECT OF ANESTHETICS AND
intraoperatively.
Communication is paramount and a comprehensive SEDATIVES ON NEURAL
plan should be developed prior to the day of surgery EXCITABILITY: GENERAL
if possible. For instance, if speech or sensory mapping PRINCIPLES
is required, the expectation is that the patient will be
awake during electrocorticography (ECoG) and exci- Most anesthetic and sedative agents produce a dose-
sion. A comprehensive preoperative visit with the pa- related synchronization and suppression of cortical ac-
tient to discuss the anesthetic plan and its implications tivity as illustrated in Fig. 9–1. Small doses may produce
will allay many of the fears associated with undergoing a disinhibition and clinical excitement phase and large
an awake procedure. Surveys of these patients postoper- doses may lead to burst suppression or even electrocor-
atively demonstrate that they tolerate awake craniotomy tical silence (flat electroencephalography (EEG)).4 Also,
well both physically and psychologically.2,3 it is important to note that the GABAA -mimetic effect of
There are two electrophysiological issues with re- many anesthetics places them squarely in the antiepilep-
gard to anesthesia (1) alteration of the seizure threshold tic family of drug actions. Thiopental,5,6 propofol,7,8 and
and loss or significant change in the character and lo- potent inhaled anesthetics9,10 have all been used to
cation of interictal spiking (IIS), and (2) increase in the treat status epilepticus. By definition, these anesthetics

109
110 SECTION I TECHNIQUES

Fast Halothane

ICV
EEG Therapeutic
Activity General anesthetic effect
drugs Desflurane

Propofol
N2O/Opioid
Slow Opioids
St Hy In M M
er pe du an ain
Flat oi rv ct ni t.
ds -P to An
en ro l
t po es
Awake Excitement Sedation Surgical Anesth Deep th
fo
Behavioral State l

Figure 9–1. Dose response of electroencephalo- Figure 9–2. Innervation of the scalp and face.
graphy (EEG) to general anesthetics. True anesthetic (Adapted from Lalwani AK. Current Diagnosis &
agents will, at sufficient dose create a state of Treatment in Otolarnyngology-Head and Neck
surgical immobility and amnesia, and at high doses Surgery, 2nd edition. http://www.access
lead to burst suppression and electrocortical silence. medicine.com.)
The effect of opioids plateau at delta range slows
wave activity. (Modified from Whittle IR, Midgley S, of pin holder sites and the surgical incision site should
Georges H, Pringle AM, Taylor R. Patient perceptions also be performed to ensure adequate analgesia.
of “awake” brain tumour surgery. Acta Neurochir
To provide for adequate duration of analgesia,
(Wien) 2005;147(3):275–277; discussion 277.)
longer acting anesthetic agents such as ropivacaine,
levobupivacaine, and bupivacaine should be used. Al-
increase the seizure threshold and negatively modulate though local anesthetic toxicity is possible with any
the IIS activity, which defines a seizure focus. agent, ropivacaine and levobupivacaine have a better
safety profile than bupivacaine with regards to car-
diotoxicity and neurotoxicity.14 Epinephrine 1:200,000
䉴 LOCAL ANESTHETICS (5 ␮g/cc) is often added to the local anesthetic solu-
tion to provide both hemostasis as well as reduce the
Local anesthetics were first used by Harvey Cushing for systemic uptake of local anesthetic. Doses of up to 4.5
brain tumor excision during the 1920s. Their use persists mg/kg ropivacaine and 2.5 mg/kg levobupivacaine have
today. Local anesthetics play a vital role in awake pro- been proven safe.15,16
cedures or in those that require minimal sedation. Ade-
quate scalp analgesia decreases dependence on sedative
and narcotic agents that may interfere with neurophysi- 䉴 BENZODIAZEPINES
ological monitoring and patient compliance. In addition,
local infiltration can decrease the tachycardia and hyper- Benzodiazepines are very popular anesthetic adjuvants.
tension during head pinning as well as postoperatively Their mechanism of action appears to include binding
due to improved pain control.11,12 A poorly performed to and activation of the native ligand site on the GABAA
block may lead to intravascular injection, respiratory de- family of ionophores.17 These drugs reduce anxiety and
pression, nausea, and disinhibition. sympathetic tone, reduce the likelihood of patient move-
A commonly employed technique for providing ment, and also reduce the probability of disturbing recall
scalp analgesia involves placing a field block with a of intraoperative events. If intraoperative localization of
long lasting local anesthetic agent. However, circumfer- an epileptic focus is part of the operative plan, midazo-
ential infiltration requires a large volume of local anes- lam should not be given because in clinical doses it may
thetic and increases the likelihood of local anesthetic suppress IIS for many hours despite its short duration of
toxicity. A more precise technique involves infiltration action. The routine use of benzodiazepines is not war-
around the sensory branches of the trigeminal nerve ranted in cases in which mapping is indicated or when
including the supraorbital, supratrochlear, auriculotem- resection is planned given its potential interference with
poral, zygomaticotemporal, lesser occipital, and greater ECoG.
occipital nerves (see Fig. 9–2). Blockade at each of these
nerves requires less total anesthetic than a field block 䉴 BARBITURATES
and has been shown to provide effective scalp analge-
sia with better hemodynamic control when compared to Barbiturates have been used for decades as both intra-
the field block technique.13 Local anesthetic infiltration venous general anesthetics and as antiepileptic drugs.
CHAPTER 9 ANESTHESIA FOR BRAIN MAPPING SURGERY 111

Pentobarbital and phenobarbital are barbiturates that are 䉴 OPIATES


entirely antiepileptic in their effect. There are, however,
other members in this subgroup whose effects are pro- Opiates are a mainstay of anesthesia for all forms of brain
convulsant. Methohexital and thiopental are both ultra- mapping surgery. Currently available agents are highly
short acting induction agents that have the expected selective agonists of the ␮-class of endogenous opiate
barbiturate depressive effects (progression to isoelectric receptors. While poor sedatives themselves, the analge-
EEG). In small doses, however, excitement and an in- sia they provide allows the patient to remain relatively
crease in IIS activity may be seen. This observation has comfortable during surgery. Within the usual range of
been used clinically to enhance IIS when needed during dosing, opiates do not usually alter seizure threshold or
epilepsy surgery.18–21 In a mouse model of seizures IIS activity. Moderate doses may result in opioid-induced
induced by pentylenetetrazol, methohexital, and muscle rigidity without EEG spiking. At extremely high
thiopental exhibited only an antiepileptic action on doses, many opiates will induce seizures.29–31 Interest-
(presumably) normal tissue.22 However, some studies ingly, in patients with partial complex epilepsy, mod-
fail to confirm that the enhanced IIS activity comes erate doses of opioids like fentanyl may have induced
solely from the targeted seizure focus23,24 or that the increases in IIS, and in these cases IIS was not con-
drugs reliably increase IIS.25,26 In somewhat larger fined to the previously identified seizure foci.32 Alfen-
doses, these barbiturates will stop the generalization of tanil, an opioid half-life than fentanyl, has been used
focal seizures that are occasionally induced by the cor- in boluses similar to methohexital to increase IIS activ-
tical electrical stimulation used in motor mapping. The ity for mapping foci.33 Remifentanil has a similar effect
rapid redistribution of methohexital or thiopental from on IIS but its duration of action is much shorter mak-
the brain to other tissue allows this antiepileptic effect to ing it a safer choice during awake craniotomy when
disappear quickly and allow further mapping after a few ventilation cannot be mechanically controlled.34 Patients
minutes. While thiopental is ultra-short acting for the who receive remifentanil generally require supplemen-
first few hundred milligrams delivered, it is only slowly tal postoperative analgesia. This has been posited as
metabolized and excreted, so redistribution will slow creation of an acute tolerance state of the ␮-receptors
with continued dosing. Methohexital is metabolized by remifentanil.35,36 Opiate overdose will, via depressed
and cleared much faster than thiopental, so it retains ventilatory drive, result in chest wall rigidity or soft tissue
its ultra-short kinetic profile despite repetitive dosing. airway obstruction. This can lead to hypercapnia lead-
Methohexital has been used to elicit or activate seizure ing in turn to cerebral vasodilation and swelling, or even
foci when quiescence is encountered intraoperatively.27 herniation. Even brief periods of hypercapnia can result
With the popularity of propofol and the increasing in contusion of the cerebral cortex by the bony edges of
use of dexmedetomidine, the use of barbiturates has the craniotomy. Adjuvant agents of the sedative classes
declined substantially in neuroanesthesia. listed later are usually given during the case to reduce
the amount of opiate required and the impact of hyper-
capnic brain swelling.

䉴 KETAMINE

Ketamine is another parenteral anesthetic agent whose 䉴 ETOMIDATE


use in neurosurgical cases is quite controversial and still
evolving. While considered a general anesthetic in doses Etomidate, a carboxylated imidazole derivative, is a
ranging from 1–4 mg/kg intravenous bolus followed by GABAA -ergic agent that acts at a different site than the
0.1–0.5 mg/min infusion, this drug produces an anal- benzodiazepines. This drug is frequently used to induce
gesic state in which the patient is dissociated from their anesthesia in hypovolemic patients, or those with se-
surroundings. EEG activity increases, particularly in the vere heart failure since it does not significantly depress
theta band,28 as does cerebral blood flow (CBF) and myocardial function. Etomidate also leads to less res-
cerebral metabolic rate. Ketamine, at significantly lower piratory depression than thiopental, but is associated
doses, has been used as an analgesic adjuvant and can with significant pain on injection and with myoclonic
be used in combination with low-dose opioids to allow movements.37 The use of etomidate is controversial in
patients to tolerate painful parts of the procedures. Data neurosurgery since there is conflicting data on its bal-
supporting the use of ketamine in traumatic brain in- ance of cerebroprotective38,39 versus neutral or even
jury may soon be available; however, its use in epilepsy harmful cerebral effects.40–42 It is clear, however, that
surgery is relatively contraindicated due to dissociated etomidate should be avoided in patients undergoing
sedation precluding cooperation, activation of the EEG, surgery for epilepsy since it may activate the EEG of
and general lack of a need for postoperative analgesia epileptic patients43 and may confound seizure focus
following craniotomy. mapping.44
112 SECTION I TECHNIQUES

䉴 PROPOFOL airway obstruction or hypoventilation in the sedated pa-


tient. Typical dexmedetomidine doses include a loading
Propofol is an intravenous anesthetic with a strong dose of 0.7–1 ␮g/kg over 10 minutes followed by infu-
GABAA -ergic effect. It is a very versatile agent, easily sion rates of 0.2–0.7 ␮g/kg/h. Doses as low as 0.2–0.3
titratable to behavioral endpoints ranging from light se- ␮g/kg/h have been used throughout the “asleep-awake-
dation to coma. It has relatively minimal effects on the asleep” (AAA) technique.
cardiovascular or respiratory system when given as an In addition to its minimal effects on ventilation,
infusion, but can cause significant hypotension with bo- dexmedetomidine also provides hemodynamic stability
lus or induction dosing. Signs of upper airway obstruc- during neurosurgical procedures.54 Via alpha-2 medi-
tion should be sought and treated if needed with an ated adrenoreceptor activity, we see modest reduction
oral or nasal airway. In low-dose regimens, propofol in blood pressure and heart rate. We also see reduction
causes activation of EEG activity,45,46 occasionally to the of circulating catecholamines effectively resulting in a
point of creating background activity, which may resem- decreased incidence of tachycardia and/or hypertension
ble epileptiform spiking.47 Larger doses lead to slowing during the perioperative period.55
and synchronization, followed by burst suppression and Transient hypertension caused by peripheral vaso-
isoelectricity. constriction of vascular smooth muscle can occur initially
The use of propofol for both awake and anes- during bolus infusion of dexmedetomidine.56 However,
thetized mapping procedures has become common- through alpha-2 mediated sympatholysis, dexmedeto-
place. Propofol does, however, have an impact on midine infusion will increase vagal activity result-
seizure foci and IIS activity, probably due to action ing in a corresponding bradycardia and hypotension.
as a GABA A agonist.48 Propofol shortens seizure dura- Dexmedetomidine has not been shown to increase in-
tion following electroconvulsant therapy compared to tracranial pressure (ICP). CBF is usually decreased while
methohexital.49,50 Herrick found that the EEG effects autoregulation and CMRO2 are preserved.
of propofol sedation on ECoG could be minimized by Perhaps most important in its role in brain map-
allowing 15 minutes recovery after terminating propo- ping, dexmedetomidine has been shown to have
fol infusion before recording.47 Soriano further demon- minimal effects on intraoperative neurophysiological
strated this in children with ECoG readings, obtained monitoring techniques. Animal and human studies have
when propofol was discontinued 30 minutes prior to shown dexmedetomidine preserves cortical somatosen-
ECoG.51 It is therefore imperative that communication sory evoked potentials and does not decrease cortical
and vigilance be maintained throughout to ensure that responses.57 Multiple studies show dexmedetomidine
optimal conditions are present for ECoG monitoring and use in epilepsy surgery results in minimal to no sup-
recording. pression of epileptiform activity and one patient had a
subclinical seizure detected while receiving dexmedeto-
midine infusion.58,59
Dexmedetomidine has minimal effects on intraoper-
䉴 DEXMEDETOMIDINE ative ECoG. Dexmedetomidine infusion can be used not
only for the asleep portion of an AAA anesthetic tech-
Dexmedetomidine is a relativity new anesthetic agent nique, but also during the awake period of language
that has seen increasing popularity in the field of neu- mapping and ECoG.58
roanesthesia. This highly specific alpha-2 receptor ag- Dexmedetomidine has many properties, which
onist provides anxiolysis, analgesia without depression make it an ideal anesthetic agent for awake craniotomy
of respiratory drive, and what has been termed “co- and intraoperative brain mapping. Its provides a sta-
operative sedation,” making it an ideal agent for use ble hemodynamic profile, no respiratory depression, and
during awake craniotomy procedures.52 Dexmedetomi- a cooperative patient who can be promptly awakened
dine has been shown to have minimal effect on neu- from anesthesia for neurocognitive testing, while also
rophysiological monitoring. The anxiolytic and sedative having a minimal effect on neurophysiological parame-
properties of dexmedetomidine are from activation of ters monitored during brain mapping.
alpha-2 receptors located in the locus ceruleus. With
the lack of respiratory depression, dexmedetomidine
has a favorable neurophysiological profile as it does 䉴 NITROUS OXIDE
not cause carbon dioxide retention or hypoxemia,
which can lead to cerebral edema during neurosurgical Nitrous oxide (N2 O) is an odorless, colorless gas with
procedures.53 low solubility and potency. It does not affect interic-
Patients can be adequately sedated and will toler- tal spikes and can be used in neurosurgery. N2 O has
ate an oral or nasopharyngeal airway if upper airway been noted to attenuate spike frequency on EcoG, but
obstruction occurs during infusion. This helps to avoid did not seem to affect the extent of the areas affected.60
CHAPTER 9 ANESTHESIA FOR BRAIN MAPPING SURGERY 113

There are certain drawbacks to its use however. N2 O for maintenance during craniotomy and mapping proce-
is a cerebral vasodilator, which can increase CBF and dures. Usually, levels below 0.5 MAC have had minimal
subsequently increase ICP. N2 O has the potential to in- effect on ECoG and EEG recordings.61,62
crease closed air spaces in noncompliant areas and can
rapidly increase pressure and volume in compliant ones,
which can lead to potentially devastating complications 䉴 ANESTHETIC TECHNIQUE FOR
like tension pneumocephalus and air embolus. Postop- BRAIN MAPPING WITH AWAKE
erative nausea and vomiting has not been consistently CRANIOTOMY
linked to this agent but is still of concern in this pa-
tient population. With the advent of newer intravenous Anesthesia for brain mapping procedures and awake
agents, we suggest avoiding N2 O during brain mapping craniotomy presents many clinical challenges to the
and seizure surgery. anesthesiologist. The patient’s specific pathology often
dictates the nature of the intraoperative neuromonitor-
ing and therefore the most appropriate anesthetic to be
䉴 VOLATILE ANESTHETIC used (see Table 9–2). Goals during an awake craniotomy
NEUROPHARMACOLOGY include maintaining patient comfort and safety, ensuring
patient cooperation while keeping the patient immobile,
The neurophysiological effects of the commonly used and ensuring adequate ventilation and circulatory sup-
volatile anesthetics are summarized in Table 9–1. Of the port. Anesthetic agents commonly used by anesthesia
commonly used volatile anesthetics, isoflurane, sevoflu- providers work well, but usually confound electrophysi-
rane, and desflurane all increase CBF via a direct va- ological monitoring and neurocognitive testing for brain
sodilatory effect. This effect on the CBF is dose-related mapping. Multiple techniques have been developed to
and biphasic. At 0.5 MAC, the vasodilatation that occurs accomplish these tasks ranging from local anesthesia as
is largely compensated by suppression in the CMR such the sole adjunct to general anesthesia with or without
that the changes in CBF are minimal. At increased doses airway instrumentation.63 Palese64 reported that roughly
(greater than 1 MAC), vasodilation occurs on a larger half of patients described craniotomy as the worst ex-
scale compared to CMR depression, resulting in uncou- perience encountered during surgery. This finding, cou-
pling of the two, and an overall increase in CBF. This pled with the deleterious effects of anxiolytics on elec-
can lead to substantial increases in ICP as 60–70% of the trophysiological monitoring, has led to the widespread
cerebral blood volume resides in the sinuses and veins. practice of performing AAA techniques when an awake
All decrease cerebral vascular resistance with suppres- patient is needed intraoperatively. Here we will discuss
sion of the CMR. This suppression is not uniform, as the various techniques for performing an AAA anesthetic;
neocortex seems more vulnerable to suppression than however, certain topics such as adequate analgesia from
deep structures like the hippocampus. local anesthetic infiltration (see Section on Local Anes-
The volatile anesthetics have similar effects on EEG thetics), preoperative patient planning, and preparation
monitoring. At doses greater than 1.5 MAC, burst sup- for the possibility of intraoperative seizure activity also
pression occurs on the EEG. Differences have been go hand in hand with any anesthetic technique used for
noted in their propensity to elicit epileptiform activ- these procedures.
ity. Epileptiform activity (spike activity) with sevoflu-
rane during single breath induction techniques or with
high-concentration exposure have been reported. This PREOPERATIVE CARE
is similar to what was seen historically with enflurance
and occurs as we proceed from burst suppression to For epilepsy surgery, antiepileptic drugs should be re-
isoelectricity. This epileptogenic potential is completely duced or discontinued for 12–24 hours prior to surgery
abolished when N2 O is administered. The volatile agents for maximal IIS. Some centers trade-off accuracy of focus
isoflurane or desflurane seem to not have this epilep- mapping for increased safety of the patient related to
togenic potential. The volatile anesthetics can be used the lower risk of seizure in the operating room (OR) by

䉴 TABLE 9–1. NEUROPHARMACOLOGY OF VOLATILE ANESTHETICS

Drug Effect on CBF Effect on EEG Epileptogenicity ECoG

Isoflurane ↔at 0.5 MAC,↑ >1 MAC ↓, burst suppression >1.5 MAC None ↔<0.5 MAC, ↓>0.5 MAC
Sevoflurane ↔at 0.5 MAC,↑ >1 MAC ↓, burst suppression >1.5 MAC Yes ↔<0.5 MAC, ↓>0.5 MAC
Desflurane ↔at 0.5 MAC,↑ >1 MAC ↓, burst suppression >1.5 MAC None ↔<0.5 MAC, ↓>0.5 MAC

CBF, cerebral blood flow; EEG, electroencephalography; ECoG, electrocorticography.


114 SECTION I TECHNIQUES

䉴 TABLE 9–2. NEUROSURGICAL PROCEDURES AND ANESTHETIC CONSIDERATIONS

Surgical Procedure Anesthetic Technique Agents Used Considerations

Corpus callosotomy Any tAny t


Hemorrhage as this is near
sagittal sinus
t
Lethargy postoperation
Intracranial grid placement Asleep t
Low dose volatile + opioid t
Avoid N2 O until dura open
t
TIVA + dexmedetomidine t
VAE risk
(0.2–0.7 ␮g/kg/h) t
Testing of grids requires
minimal anesthetics
t
Propofol off 30 min prior to
EEG/ECoG
Tumors near eloquent areas Awake for language mapping t
TIVA + dexmedetomidine t
Proper patient selection If
(speech, language) or asleep-awake-asleep infusion 0.3–0.7 ␮g/kg/h cortical stimulation—avoid
t
Local and MAC NMBs
t
gropofol off 30 min prior to
EEG/ECoG
t t
Deep brain stimulation Awake Local and MAC Avoidance of GABA
t
Dexmedetomidine agonists
0.3–0.7 ␮g/kg/h

EEG, electroencephalography; ECoG, electrocorticography, VAE, venous air embolism; TIVA, total intravenous anesthesia; NMB,
neuromuscular blockade.

continuing antiepileptic drugs until surgery. No anxi- quired. If opiates or sedatives are given, a sidestream
olytic premedications should be given; the practitioner sample of exhaled gas should be sampled for end-tidal
should remember that the best anxiolytic is a good pre- CO2 assessment. A Foley catheter may be inserted into
operative visit. An antisialagogue such as glycopyrrolate the bladder in awake or sedated cases if the case is pro-
(0.1–0.2 mg) may be useful in patients who smoke. As jected to last beyond 4–5 hours (which is typical), or if
prophylaxis against nausea and vomiting, antiemetic mannitol is being used (not typical). Crystalloid infusion
agents such as ondansetron and dexamethasone should should be minimal in these cases to reduce postsurgi-
be considered preoperatively. This is especially impor- cal edema. Surgical blood loss rarely exceeds 500 mL
tant if the patient is to be awake for any part of the and if volume replacement is deemed necessary either
procedure. blood or albumin are the recommended choices. Inva-
Preoperative teaching is usually performed by the sive monitoring is not generally indicated in these cases
team neurologist or psychologist regarding mapping except possibly for the management of serious concur-
procedures and what to expect in terms of the OR ex- rent cardiopulmonary disease.
perience and should be verified in the chart or with the
patient. It is wise to counsel the patient regarding the
rare possibility of postoperative recall following a gen-
eral anesthetic planned for a mapping case. Emphasis on ANESTHETIC MANAGEMENT
aggressive pain control and minimal discomfort should
be the goal. AAA technique has been described both with and with-
On call to the OR the patient should void. A pe- out airway instrumentation. Before continuing, it is im-
ripheral IV greater than 18 gauge should be placed. On portant to reiterate the crucial role of adequate analge-
the basis of the possibility of focal ischemic injury during sia from a well-performed scalp block to help maximize
brain retraction, dextrose-containing solutions should be patient comfort. If airway instrumentation is to be used,
avoided. Finally, consider a three-way stopcock at the taping the endotracheal tube or supraglottic airway de-
hub of IV for continuous drug infusion while allowing vice requires considering security and the need to vi-
maintenance fluids to infuse at a minimal rate. sualize muscles of facial expression during mapping. A
soft bite block must be used in light of the potential for
seizure activity during surgery. Obviously, during motor
mapping neuromuscular blockade must be avoided.
INTRAOPERATIVE MONITORING During an AAA technique, the patient is placed un-
der general anesthesia for the rigid fixation if used, fol-
Standard ASA monitors are appropriate for mapping lowed by scalp incision, dissection of temporalis muscle,
craniotomies, that is, oscillometric (noninvasive) blood drilling and sawing (not so much painful as disturbingly
pressure, electrocardiogram, and pulse oximeter. If the loud), and dural opening. One of the primary con-
patient is to remain awake no temperature probe is re- cerns for this technique involves airway management.
CHAPTER 9 ANESTHESIA FOR BRAIN MAPPING SURGERY 115

Induction of general anesthesia and endotracheal 䉴 TABLE 9–3. ANESTHETIC AGENTS USED TO
intubation mandates airway topicalization with local ELICIT SPIKE WAVE ACTIVITY
anesthetics to minimize straining or coughing upon ex- Agents and Dosages
tubation as well as facilitate re-intubation after mapping
is complete. Lidocaine 5% in water-soluble ointment has Methohexital 0.3–0.5 mg/kg
been quite effective in this role. Modifications of the AAA Etomidate 0.1–0.2 mg/kg
technique involve propofol as the sole sedative agent for Alfentanil 50 ␮g/kg
Remifentanil 2.5 ␮g/kg
sedation with spontaneous respiration with or without
a laryngeal mask airway (LMA). The propofol infusion
(typically 75–250 ␮g/kg/min) is discontinued approxi-
continued mapping and prevented the conversion to
mately 15–30 minutes prior to ECoG. As noted previ-
general anesthesia.72 Doses in this latter report were
ously, propofol has a significant effect on IIS48 that may
even lower infusion rates (0.15 ␮g/kg/hr) yet still pro-
be mitigated by allowing sufficient time to pass between
viding adequate patient comfort and cooperation and
stopping the infusion and beginning ECoG.47 The transi-
good cortical mapping conditions.72 If no airway instru-
tion from asleep to awake is the most difficult aspect of
mentation is desired, dexmedetomidine is an ideal agent
this anesthetic plan secondary to issues related to airway
because, as described previously, it has minimal effect
complications with the brain exposed. Nausea and vom-
on ventilatory drive, systemic or cerebral hemodynam-
iting during this period may lead to aspiration as well as
ics, and provides for a comfortable, cooperative patient.
dramatic increases in ICP. The complication rate during
There is limited experience with dexmedetomidine in
this period is difficult to discern and variable given there
children and adolescents but several case reports have
are no prospective trials, but retrospective analysis by
demonstrated its use.73 Dexmedetomidine is frequently
Skucas and Artu showed the airway complication rate to
used as an anesthetic adjuvant during the “asleep” com-
be 2% in a review of 332 cases. Other studies point to
ponent of a craniotomy and in many hands does not
an average complication rate of 9.5%.65 The highest in-
appear to interfere significantly with neurocognitive test-
cidence of complications during this technique is based
ing. Its infusion is typically terminated 20 minutes prior
on airway management with 17% of patients experienc-
to testing and the patient allowed to be awaken to a Ram-
ing moderate desaturation (91–95%)66 and respiratory
sey Sedation score of 2 to 3.74 Obviously its use should
depression (respiratory rate <8 breaths per minute),67
be discussed preoperatively with the neurologist or psy-
which was commonly managed with LMA placement.
chologist who will be doing the intraoperative testing.
Once ECoG has finished, propofol may be reinstituted
Despite the use of anesthetic agents with minimal
for the resection and closure.68
effect on electrophysiological monitoring, sometimes
Additional modifications of the AAA technique have
there is still a lack of spike wave activity. Intravenous
involved infusion or bolus administration of short acting
medications that can be used to induce spike wave activ-
narcotics.69 At present, the most commonly used nar-
ity include methohexital, etomidate, and alfentanil (see
cotic appears to be remifentanil, most likely because
Table 9–3).
of its short half-life and titratability. Remifentanil use
The AAA technique is well tolerated by patients
has been advocated to ensure adequate analgesia dur-
and generally reported to have high satisfaction rates.
ing bone flap removal. Using the propofol–remifentanil
Approximately one-third of patients’ post-procedure re-
technique infusion rates are 100–150 ␮g/kg/min for
ported complaints about rigid head fixation being too
propofol and 0.02–0.09 ␮g/kg/min for remifentanil. With
tight and painful scalp incision.2,3 Additional complaints
this technique the time from discontinuation of infusion
were noted regarding bladder distension and pressure
to the time of “emergence,” defined as opening eyes and
on the dependent limb during lateral position. Of note,
following commands, ranged between 6 and 13 min-
no patients had recall of pain upon dural incision.3
utes following an average asleep craniotomy time of 48
minutes.70 This technique can be used either with or
without airway instrumentation, as it provides adequate
sedation for patients to tolerate either an LMA or endo- 䉴 ANESTHETIC TECHNIQUE FOR
tracheal tube. If no airway is used, the rapid titratability BRAIN MAPPING WITH GENERAL
of remifentanil allows for return of ventilatory drive if ANESTHESIA
respiratory depression were to occur.
Dexmedetomidine has become a popular adjuvant Not all patients are suitable candidates for an awake
to sedation or analgesia for the AAA technique based craniotomy. This may be because of lack of patient co-
upon its lack of respiratory depression. At low infu- operation such as occurs in the pediatric population or
sion rates (0.2 ␮g/kg/hr) there appears to be little ef- because of other factors such as a difficult airway, severe
fect on epileptiform activity and satisfactory spike and obstructive sleep apnea, or coexisting cardiopulmonary
wave discharges.71 Moore, et al. reported the use of disease. These factors may necessitate general anesthe-
dexmedetomidine as a rescue medication to allow for sia throughout the entire procedure. However, despite
116 SECTION I TECHNIQUES

presenting different challenges, general anesthesia is still 4. Faulconer A, Bickford RG. Electroencephalography in
compatible with intraoperative motor mapping as long Anesthesiology. Springfield, IL: Charles C. Thomas, 1960.
as attention is paid to how anesthetic agents can affect 5. Brown AS, Horton JM. Status epilepticus treated by in-
the various electrophysiological techniques being used travenous infusions of thiopentone sodium. Br Med J
during the procedure. 1967;1(5531):27-28.
6. Dundee JW, Gray RC. Thiopentone in status epilepticus. Br
As discussed previously, all volatile anesthetics,
Med J 1967;1(5536):362.
benzodiazepines, and hypnotic agents can suppress 7. Wood PR, Browne GP, Pugh S. Propofol infusion for the
spike wave activity. When volatile anesthetic agents are treatment of status epilepticus. Lancet 1988;1(8583):480-
being used, they should be discontinued and eliminated 481.
prior to performing ECoG. This can be accomplished 8. Yanny HF, Christmas D. Propofol infusions for status
by having a balanced anesthetic consisting of low-dose epilepticus. Anaesthesia 1988;43(6):514.
volatile agent combined with N2 O and opioid infusion. 9. Kofke WA, Snider MT, Young RS, Ramer JC. Prolonged low
When the volatile agent has been eliminated, N2 O and flow isoflurane anesthesia for status epilepticus. Anesthe-
opioid infusion can be maintained to ensure an adequate siology 1985;62(5):653-656.
anesthetic depth. 10. Ropper AH, Kofke WA, Bromfield EB, Kennedy SK. Com-
Since N2 O is not an ideal agent for craniotomies parison of isoflurane, halothane, and nitrous oxide in status
epilepticus. Ann Neurol 1986;19(1):98-99.
owing to its increase in CBF and the inherent risk of
11. Shiau JM, Chen TY, Tseng CC, et al. Combination of bupi-
venous air embolism during craniotomy, dexmedetomi- vacaine scalp circuit infiltration with general anesthesia to
dine may be substituted either for maintenance through- control the hemodynamic response in craniotomy patients.
out the anesthetic or instituted during the time that brain Acta Anaesthesiol Sin 1998;36:215-220.
mapping is being performed. If used, dexmedetomidine 12. Batoz H, Verdonck O, Pellerin C, et al. The analgesic prop-
should be run at low infusion rates of 0.2–0.7 ␮g/kg/h to erties of scalp infiltrations with ropivacaine after intracra-
ensure there is no interference with brain mapping. All nial tumoral resection. Anesth Analg 2009;109:240-244.
narcotic agents have been used successfully during brain 13. Geze S, Yilmaz AA, Tuzuner F. The effect of scalp block and
mapping, and while studies have shown various effects local infiltration on the haemodynamic and stress response
on spike wave activity, any may be used as long as they to skull-pin placement for craniotomy. Eur J Anaesthesiol
are kept at normal clinical dosages. While it is tempt- 2009;26(4):298-303.
14. Scott DB, Lee A, Fagan D, et al. Acute toxicity of ropi-
ing to administer benzodiazepines during this period
vacaine compared with that of bupivacaine. Anesth Analg
without volatile anesthetic in the effort to prevent pa- 1989;69:563-569.
tient recall or awareness, one must remember that these 15. Costello T, Cormack JR, Hoy C, et al. Plasma ropivacaine
agents have been shown to suppress spike wave activ- levels following scalp block for awake craniotomy. J Neu-
ity and should be avoided while brain mapping is be- rosurg Anesthesiol 2004;16:147-150
ing performed. Despite these maneuvers, the period of 16. Costello TG, Cormack JR, Mather LE, et al. Plasma lev-
brain mapping and lack of volatile anesthetic represents obupivacaine concentrations following scalp block in
a time when the patient is at the highest risk for lightened patients undergoing awake craniotomy. Br J Anaesth
anesthesia and therefore bucking or movement. Increas- 2005;94:848-851.
ing doses of remifentanil or fentanyl should decrease the 17. Doble A. New insights into the mechanism of action of
risk for this potential complication. If no motor mapping hypnotics. J Psychopharmacol 1999;13(4 Suppl 1):S11-S20.
18. Wyler AR, Richey ET, Atkinson RA, Hermann BP. Metho-
is being performed, adequate paralysis should be main-
hexital activation of epileptogenic foci during acute elec-
tained. Once the brain mapping has been completed, trocorticography. Epilepsia 1987;28(5):490-494.
volatile anesthetic agents may be restarted for the re- 19. Hardiman O, Coughlan A, O’Moore B, Phillips J, Staunton
mainder of the procedure. H. Interictal spike localisation with methohexitone: pre-
operative activation and surgical follow-up. Epilepsia
1987;28(4):335-339.
20. Ford EW, Morrell F, Whisler WW. Methohexital anesthesia
REFERENCES in the surgical treatment of uncontrollable epilepsy. Anesth
Analg 1982;61(12):997-1001.
1. Todd MM, Warner DS, Sokoll MD, et al. A prospective, com- 21. Harris R, Paul R. The use of methohexitone in elec-
parative trial of three anesthetics for elective supratentorial trocorticography. Electroencephalogr Clin Neurophysiol
craniotomy. Anesthesiology 1993;78:1005-1020. 1969;27(3):333-334.
2. Danks RA, Rogers M, Aglio LS, Gugino LD, Black PM. Pa- 22. Lowson S, Gent JP, Goodchild CS. Convulsive thresholds in
tient tolerance of craniotomy performed with the patient mice during the recovery phase from anaesthesia induced
under local anesthesia and monitored conscious sedation. by propofol, thiopentone, methohexitone and etomidate.
Neurosurgery 1998;42(1):28-34; discussion 34-36. Br J Pharmacol 1991;102(4):879-882.
3. Whittle IR, Midgley S, Georges H, Pringle AM, Taylor R. 23. Fiol ME, Torres F, Gates JR, Maxwell R. Methohexital
Patient perceptions of “awake” brain tumour surgery. Acta (Brevital) effect on electrocorticogram may be misleading.
Neurochir (Wien) 2005;147(3):275-277; discussion 277. Epilepsia 1990;31(5):524-528.
CHAPTER 9 ANESTHESIA FOR BRAIN MAPPING SURGERY 117

24. Cascino GD. Pharmacological activation. Electroen- 41. Hoffman WE, Charbel FT, Edelman G, Misra M, Ausman
cephalogr Clin Neurophysiol 1998;48(Suppl):70-76. JI. Comparison of the effect of etomidate and desflurane
25. Aung MH. Electroencephalographic activation with metho- on brain tissue gases and pH during prolonged middle
hexital in an epileptic population. Dis Nerv Syst cerebral artery occlusion. Anesthesiology 1998;88(5):1188-
1974;35(5):246-248. 1194.
26. Harel D, Sharf B, Bental E. Methohexital as an activator in 42. Amadeu ME, Abramowicz AE, Chambers G, Cottrell JE,
epileptic patients with normal electroencephalograms. Isr Kass IS. Etomidate does not alter recovery after anoxia of
J Med Sci 1975;11(10):986-990. evoked population spikes recorded from the CA1 region
27. Paul R, Harris R. A comparison of methohexitone and of rat hippocampal slices. Anesthesiology 1998;88(5):1274-
thiopentone in electrocorticography. Neurol Neurosurg 1280.
Psychiatry 1970;33:100-104. 43. Gancher S, Laxer KD, Krieger W. Activation of epilep-
28. Celesia GG, Bamforth BJ, Chen RC. Letter: effects of ke- togenic activity by etomidate. Anesthesiology 1984;61(5):
tamine on the EEG in normals and epileptics. Anesth Analg 616-618.
1976;55(3):445-451. 44. Ebrahim ZY, DeBoer GE, Luders H, Hahn JF, Lesser RP. Ef-
29. de Castro J, Van de Water A, et al. Comparative study fect of etomidate on the electroencephalogram of patients
of cardiovascular, neurological and metabolic side effects with epilepsy. Anesth Analg 1986;65(10):1004-1006.
of 8 narcotics in dogs. Pethidine, piritramide, morphine, 45. Seifert HA, Blouin RT, Conard PF, Gross JB. Sedative doses
phenoperidine, fentanyl, R 39 209, sufentanil, R 34 995. of propofol increase beta activity of the processed elec-
II. Comparative study on the epileptoid activity of the troencephalogram. Anesth Analg 1993;76(5):976-978.
narcotics used in high and massive doses in curarised 46. Veselis RA, Reinsel RA, Wronski M, Marino P, Tong WP,
and mechanically ventilated dogs. Acta Anaesthesiol Belg Bedford RF. EEG and memory effects of low-dose infusions
1979;30(1):55-69. of propofol. Br J Anaesth 1992;69(3):246-254.
30. Scott JC, Sarnquist FH. Seizure-like movements during a 47. Herrick IA, Craen RA, Gelb AW, et al. Propofol se-
fentanyl infusion with absence of seizure activity in a simul- dation during awake craniotomy for seizures: electro-
taneous EEG recording. Anesthesiology 1985;62(6):812- corticographic and epileptogenic effects. Anesth Analg
814. 1997;84(6):1280-1284.
31. Benthuysen JL, Smith NT, Sanford TJ, Head N, Dec-Silver 48. Rampil IJ, Lopez CE, Laxer KD, Barbaro NM. Propofol se-
H. Physiology of alfentanil-induced rigidity. Anesthesiol- dation may disrupt interictal epileptiform activity from a
ogy 1986;64(4):440-446. seizure focus. Anesth Analg 1993;77(5):1071-1073.
32. Tempelhoff R, Modica PA, Bernardo KL, Edwards I. 49. Simpson KH, Halsall PJ, Carr CM, Stewart KG. Propofol
Fentanyl-induced electrocorticographic seizures in patients reduces seizure duration in patients having anaesthesia
with complex partial epilepsy. J Neurosurg 1992;77(2):201- for electroconvulsive therapy. Br J Anaesth 1988;61(3):343-
208. 344.
33. Cascino GD, So EL, Sharbrough FW, et al. Alfentanil- 50. Rampton AJ, Griffin RM, Stuart CS, Durcan JJ, Huddy
induced epileptiform activity in patients with partial NC, Abbott MA. Comparison of methohexital and propo-
epilepsy. J Clin Neurophysiol 1993;10(4):520-525. fol for electroconvulsive therapy: effects on hemody-
34. Wass CT, Grady RE, Fessler AJ, et al. The effects of remifen- namic responses and seizure duration. Anesthesiology
tanil on epileptiform discharges during intraoperative elec- 1989;70(3):412-417.
trocorticography in patients undergoing epilepsy surgery. 51. Soriano SG, Eldredge EA, Wang FK, et al. The effect
Epilepsia 2001;42(10):1340-1344. of propofol on intraoperative electrocorticography and
35. Vinik HR, Kissin I. Rapid development of tolerance to anal- cortical stimulation during awake craniotomies in children.
gesia during remifentanil infusion in humans. Anesth Analg Paediatr Anaesth 2000;10(1):29-34.
1998;86(6):1307-1311. 52. Frost EA, Booij LH. Anesthesia in the patient for awake
36. Stricker PA, Kraemer FW, Ganesh A. Severe remifentanil- craniotomy. Curr Opin Anaesthesiol 2007;20:331-335.
induced acute opioid tolerance following awake cran- 53. Ramsey MA, Luterman DL. Dexmedetomidine as a total in-
iotomy in an adolescent. J Clin Anesth 2009;21(2): travenous anesthetic agent. Anesthesiology 2004;101:787-
124-126. 781.
37. Ghoneim MM, Yamada T. Etomidate: a clinical and elec- 54. Bekker A, Sturaitis MK. Dexmedetomidine for neurological
troencephalographic comparison with thiopental. Anesth surgery. Neurosurgery 2005;57:1-10.
Analg 1977;56(4):479-485. 55. Talke P, Chen R, Thomas B, et al. The hemodynamic and
38. Ashton D, van Reempts J, Wauquier A. Behavioural, elec- adrenergic effects of perioperative dexmedetomidine in-
troencephalographic and histological study of the protec- fusion after vascular surgery. Anesth Analg 2000;90:834-
tive effect of etomidate against histotoxic dysoxia produced 839.
by cyanide. Arch Int Pharmacodyn Ther 1981;254(2):196- 56. Bloor BC, Ward DS, Belleville JP, et al. Effects
213. of intravenous dexmedetomidine in humans. Part II-
39. Milde LN, Milde JH. Preservation of cerebral metabolites Hemodynamic changes. Anesthesiology 1992;77:1134-
by etomidate during incomplete cerebral ischemia in dogs. 1142.
Anesthesiology 1986;65(3):272-277. 57. Bai-Han Li, Lohmann JS, Schuler H, et al. Preserva-
40. Guo J, White JA, Batjer HH. Limited protective effects of tion of the cortical somatosensory-evoked potential dur-
etomidate during brainstem ischemia in dogs. J Neurosurg ing dexmedetomidine infusion in rats Anesth Analg
1995;82(2):278-283. 2003;96:1155-1160.
118 SECTION I TECHNIQUES

58. Souter MJ, Rozet I, Ojemann J, et al. Dexmedetomidine 67. Herrick IA, Craen RA, Gelb AW, et al. Propofol sedation
during awake craniotomy for seizure resection: effects on during awake craniotomy for seizures: patient-controlled
electrocorticography. J Neurosurg Anesthesiol 2007;19:38- administration versus neurolept analgesia. Anesth Analg
44. 1997;84(6):1285-1291.
59. Thornton C, Lucas MA, Newton DE, Dore CJ, Jones RM. 68. Silbergeld DL, Mueller WM, Colley PS, Ojemann GA, Let-
Effects of dexmedetomidine on isoflurane requirements tich E. Use of propofol (Diprivan) for awake craniotomies:
in healthy volunteers. Part 2-Auditory and somatosensory technical note. Surg Neurol 1992;38(4):271-272.
evoked responses. Br J Anaesth 1999;83:381-386. 69. Gignac E, Manninen PH, Gelb AW. Comparison of fen-
60. Kurita N, Kawaguchi M, Hoshida T, et al. Effects of ni- tanyl, sufentanil and alfentanil during awake craniotomy
trous oxide on spike activity on electrocorticogram under for epilepsy. Can J Anaesth 1993;40(5 Pt 1):421-424.
sevoflurane anesthesia in epileptic patients. J Neurosurg 70. Keifer JC, Dentchev D, Little K, Warner DS, Friedman
Anesthesiol 2005;17(4):199-202. AH, Borel CO. A retrospective analysis of a remifen-
61. Soriano S. Anesthesia for epilepsy surgery in children. tanil/propofol general anesthetic for craniotomy before
Childs Nerv Syst 2006;22:834-843. awake functional brain mapping. Anesth Analg 2005;
62. IlijimaT, Nakamura Z, Iwao Y, et al. The epileptogenic 101(2):502-508.
properties of the volatile anesthetics sevoflurane and isoflu- 71. Souter MJ, Rozet I, Ojemann JG, et al. Dexmedetomidine
rane with epilepsy. Anesth Analg 2000;91(4):989-995. sedation during awake craniotomy for seizure resection:
63. Dinsmore J. Anesthesia for elective neurosurgery. Br effects on electrocorticography. J Neurosurg Anesthesiol
JAnaesth 2007;99:68-74. 2007;19(1):38-44.
64. Palese A, Skrap M, Fachin M, et al. The experience of pa- 72. Moore TA 2nd, Markert JM, Knowlton RC. Dexmedeto-
tients undergoing awake craniotomy: in the patients’ own midine as rescue drug during awake craniotomy for cor-
words. A qualitative study. Cancer Nurse 2008;31(2):166- tical motor mapping and tumor resection. Anesth Analg
172. 2006;102(5):1556-1558.
65. Szelenyi A, Joksimovic B, Seifert V. Intraoperative risk of 73. Ard J, Doyle W, Bekker A. Awake craniotomy with
seizures associated with transient direct cortical stimulation dexmedetomidine in pediatric patients. J Neurosurg Anes-
in patients with symptomatic epilepsy. J Clin Neurophysiol thesiol 2003;15(3):263-266.
2007;24:39-43. 74. Mack PF, Perrine K, Kobylarz E, Schwartz TH, Lien
66. Skucas AP, Artru AA. Anesthetic complications of CA. Dexmedetomidine and neurocognitive testing in
awake craniotomies for epilepsy surgery. Anesth Analg awake craniotomy. J Neurosurg Anesthesiol 2004;16(1):
2006;102(3):882-887. 20-25.
Chapter 10
Clinical Applications of
Magnetoencephalography in
Neurology and Neurosurgery
Panagiotis G. Simos 1 , Eduardo M. Castillo2 , and Andrew C. Papanicolaou2
1
Department of Psychology, University of Crete, Rethymno, Greece
2
Department of Pediatrics, Center for Clinical Neurosciences; Departments of Neurosurgery and Neurology.
University of Texas – Health Science Center at Houston, Houston, Texas

䉴 INTRODUCTION TO rents that take place within populations of pyramidal


MAGNETOENCEPHALOGRAPHY cells in neo- and archeocortical brain structures. While
TECHNIQUES electroencephalography (EEG) primarily measures the
secondary currents, which spread through the extracel-
Magnetoencephalography (MEG) is a noninvasive brain lular space and structures, reaching the head surface
imaging method that detects minute changes in the dis- with significant distortion, the magnetic flux produced
tribution of magnetic flux at the surface of the head. by primary currents penetrates the brain and other tis-
Recordings are performed either at rest (targeting spon- sues that surround the populations of active neurons
taneous abnormal activity associated with brain dam- without significant distortion.
age, e.g., in epilepsy) or during the performance of The instrument used to measure these signals is
specific motor, language, or memory tasks. In the lat- the neuromagnetometer that is equipped with an ar-
ter case recordings and subsequent analyses of the ray of sensors (either magnetometers or gradiometers)
magnetic signals aim to reveal the anatomic outline each coupled to a special, low noise amplifier. State-
of the brain mechanisms responsible for the motor, of-the-art systems feature are equipped with 150–250 or
cognitive, or linguistic function(s) engaged during task more sensors that cover the entire head. Recording takes
performance. While the anatomical resolution of MEG place inside a magnetically shielded room designed to
techniques appears to be adequate for most clinical ap- reduce extraneous magnetic fields. Special postacquisi-
plications, a conclusion supported by several normative tion signal-processing software is required in order to
and clinical studies investigating reproducibility and con- distinguish the minute magnetic signals produced by the
current validity, it is the temporal resolution of MEG brain from extraneous magnetic fields (mechanical and
that clearly distinguishes this method among noninva- biological noise).
sive brain imaging modalities that rely on hemodynamic The instantaneous surface distribution of the mag-
measures such as positron emission tomography and netic flux measured by the entire array of sensors is
functional magnetic resonance imaging (fMRI). The tem- represented by a contour map. Special algorithms are
poral and spatial resolution of MEG, as well as its demon- applied to this distribution, which is typically registered
strated reliability and external validity, have led to its at every 1–4 millisecond during the recording session, in
routine use in many neurosurgery centers around the order to estimate the position and strength of neurophys-
world. In this chapter we review the primary clinical ap- iological events (sources) that produced the recorded
plications of MEG, namely the identification of epilep- magnetic activity. The most commonly used source esti-
togenic foci in candidates for epilepsy surgery and the mation algorithm in clinical MEG applications considers
mapping of function-specific cortex prior to resection, the intracranial activity sources as equivalent to electri-
following a brief description of MEG methodology. cal current dipoles. Once estimates of the coordinates
MEG involves the measurement of neuromagnetic of such sources are made, in the MEG coordinate sys-
signals emanating from the brain (for detailed discussion tem, they are co-registered with a set of structural im-
see Papanicolaou).1 These signals reflect instantaneous ages of the patient’s brain (MRI), in order to determine
changes in intracellular dendritic (primary) electrical cur- the anatomical location of each source.

119
120 SECTION I TECHNIQUES

The capacity to identify relatively small cortical magnetic spikes some clinicians have sought to evoke
patches that show transient increases in neurophysio- epileptiform activity with the use of proconvulsant drugs
logical activity (i.e, signaling between neuronal popula- such as methohexital or clonidine.2,5 The effectiveness
tions) renders MEG suitable for a variety of clinical appli- of such techniques however, to increase the diagnos-
cations, which can be classified in two broad categories: tic yield of MEG is still uncertain. Moreover, while the
those that involve recording and subsequent localiza- preponderance of MEG applications in epilepsy have re-
tion of the intracranial sources of spontaneous, abnormal lied on spike-like activity, the localization of generators
neurophysiological activity, and those that target stim- of focal slow activity, in the delta and theta frequency
ulus evoked activity, either during passive stimulation range, has also been found to be useful for localization
conditions or while the patient is performing a cognitive of epileptic areas.6–10
or linguistic task. The third prerequisite for the clinical utility of MEG
has been addressed in two ways. First, in studies that
record and attempt to localize the intracranial origin of
䉴 SPONTANEOUS MAGNETIC ictal magnetic activity.11–16 These studies can be per-
ACTIVITY formed during nonconvulsive seizures, because mag-
netic artifacts associated with electrical muscle activity
Among the clinical applications of MEG that target spon- and movement render abnormal brain activity unusable.
taneous, abnormal brain activity, the localization of the Alternatively, brief samples of magnetic activity at at
sources of interictal transients (spikes) is the most es- the very onset of a convulsive seizure can be fortu-
tablished one world-wide. At different epilepsy centers itously obtained. In those studies where magnetic ac-
MEG is part of either the Phase I or Phase II evalu- tivity of adequate quality was obtained (for instance at
ation procedure, as an adjunct to noninvasive (video- the very onset of a seizure) epileptogenic zone local-
EEG monitoring (VEEG)) or invasive (intracranial VEEG) izations were in good agreement with subsequent in-
techniques, respectively. In this context, MEG studies vasive recordings14,17,18 or localizations that appear to
may contribute to the consensus decision of whether a be more accurate than those based surface VEEG (see
particular patient is a surgical candidate and help guide Fig. 10–1).19
subdural grid placement. The clinical utility of MEG data A fruitful approach to determine the importance of
has a number of prerequisite conditions. First, that the interictal source localizations obtained by MEG in rela-
MEG procedures and algorithms used for the localization tion to the epileptogenic zone(s) is to conduct studies
of abnormal magnetic activity are sufficiently accurate that directly compare the results of interictal MEG stud-
in identifying the location and extent of the cortical ies with those of VEEG and invasive VEEG having, to the
patches that give rise to transient increases in mag- degree possible, ensured that the former two prerequi-
netic flux recorded at the surface of the head (magnetic sites are met (localization accuracy and yield). Examples
spikes). Second, that a sufficient quantity of spikes is de- of two such studies are described in more detail later.
tected during the MEG recording session (which rarely In the first study MEG recordings were compared
exceeds 1 or 2 hours in duration). And third, that the with both ictal and interictal noninvasive video EEG
cortical patches where generators of magnetic spikes are (VEEG or Phase I EEG) in order to assess the rela-
localized coincide with the epileptogenic zone(s). It is tive sensitivity and selectivity of each method (Pataraia
essential that the MEG study identifies at least a por- et al.).3 The data set included 82 successive patients with
tion of the epileptogenic zone (sensitivity) and that the medically refractory epilepsy who underwent surgery.
abnormally active cortical patch(es) identified by MEG The epileptogenic region identified by interictal and ictal
cause seizures (selectivity). The degree to which MEG VEEG and MEG was defined in relation to the resected
procedures meet these requirements depend faces both area as either (1) perfectly overlapping with the resected
practical and physiological limitations. With respect to area, (2) partially overlapping, or (3) nonoverlapping.
the first requirement, several studies concur that the The sensitivity of the 30-minute interictal MEG record-
equivalent current model is sufficient in order to ac- ing session for detecting clinically significant epilepti-
curately localize the origin of interictal spikes.2–4 As- form activity was 79.2%, while in the remaining patients
sessment of the reliability of source localization across no interictal MEG events were detected. The proportion
several spikes and, potentially, obtaining a second of extra-temporal lobe epilepsy patients for whom in-
recording session with the same patient may help im- terictal events were registered was slightly higher than
prove the accuracyof identification of abnormally active the proportion of patients with temporal lobe epilepsy
cortical patches. (86.7% vs. 75%, respectively).
A second practical problem faced by clinicians us- On the basis of the MEG data alone, the correct lo-
ing MEG for presurgical epilepsy planning is the rarity of calization of the resected region would have been possi-
usable, abnormal epileptiform activity during the, typi- ble in 72.3% of the patients for whom at least five inter-
cally brief, recording session. To optimize the yield of ictal spikes of similar morphology were registered. This
CHAPTER 10 CLINICAL APPLICATIONS OF MEG IN NEUROLOGY AND NEUROSURGERY 121

Figure 10–1. Localization of epileptiform activity using magnetoencephalography


(MEG). Ictal electroencephalography/MEG tracing (left) recorded during right body
(distal arm) epilepsia partialis continua with time-locked discharges with each twitch.
The topographic distribution of the magnetic fields (center) associated with each
epileptiform discharge was used to select the MEG sensors used for source
modeling. After co-registration with the patient’s MRI (right), MEG revealed overlap of
ictal sources (triangles) with the sensorimotor areas (green square representing S1).
(Image reproduced with permission from Wheless JW, Castillo EM, Maggio V, et al.
Magnetoencephalography (MEG) and Magnetic Source Imaging (MSI). Neurologist
2004;10(3):1–16.)

figure was significantly higher than the proportion of epileptogenic area overlapped with the area resected
correct localizations made on the basis of the surface and the patient was seizure-free postoperatively, (2) in-
VEEG results alone (40%). In addition, MEG contributed correct if the identified area was the same as resected
to the localization of the resected region in 58.8% of the area, but the patient was not seizure-free postopera-
patients in whom VEEG recordings did not provide lo- tively, or if the localized area they identified was different
calization information and in 72.8% of the patients for from the resected area but the patient was seizure-free
whom VEEG identified only part of the resected zone. postoperatively, and (3) indeterminate if the identified
Overall, MEG and VEEG results were equivalent in 32.3% zone differed from the resected one and the patient was
of the cases, and additional localization information was not seizure-free postoperatively. With respect to post-
obtained using MEG in 40% of the patients. These results operative outcome, MEG localization was correct in 23
clearly suggest that MEG can be useful for presurgical of the 41 patients and invasive VEEG localization was
planning in patients who have either partially localizing correct in 22 of the 41 patients. The MEG predictions
or nonlocalizing VEEG results. Findings from this study were incorrect for 12 patients and the invasive VEEG
concur with subsequent reports that resecting the region predictions in 16 patients. Finally, there were six and
demonstrating dense sources of interictal spikes would three indeterminate cases for Invasive VEEG and MEG,
lead to a favorable surgical outcome (see Table 10–1).21 respectively. No significant differences between the two
For a subset of 41 patients (29 with temporal and 12 methods were found in terms of their ability to pre-
with extra-temporal epilepsy) it was possible to assess dict the localization of the epileptic zone and, conse-
the relative efficacy of interictal MEG studies and inva- quently, seizure outcome in individual patients. There-
sive electrophysiological procedures (both ictal and in- fore, at present, both methods can be considered to have
terictal, invasive VEEG) in predicting seizure outcome.22 the same degree of accuracy for identifying the epilep-
All patients had positive interictal findings in MEG. The togenic zones.
epileptogenic zone was defined separately on the basis These results are in agreement with other reports
of MEG and invasive VEEG, each in relation to post- demonstrating good agreement of interictal MEG local-
operative seizure outcome. The results of each method izations with that of invasive recordings.2,4,14,18,19,23–28,30
were separately classified as (1) correct, if the identified For instance, Knowlton and colleagues (2006)31 reached
122 SECTION I TECHNIQUES

䉴 TABLE 10–1. CONTRIBUTION OF MEG DATA TO THE IDENTIFICATION OF THE RESECTED REGION

MEG

Noninvasive VEEG (interictal and ictal) Improvement No Change Misidentification Total

Perfect overlap – 21 (80.8) 5 (19.2) 26 (40)


Partial overlapa 16 (72.8) 3 (13.6) 3 (13.6) 22 (34)
Nonlocalizable 10 (58.8) 7 (41.2) 17 (26)
Total 26 (40.0) 31 (47.7) 8 (12.3) 65 (100)

Source: With permission, Pataraia et al., 2004.


EEG, electroencephalography; MEG, magnetoencephalography.
a
p <0.01.
Values are n (%).
Note: Out of 22 patients for whom the scalp video-EEG (VEEG) ictal and interictal data would have resulted in the partial delineation of
the resected area, the MEG data would have contributed to a more accurate determination of that zone in 16 patients (72.8%). In this
series, MEG data would have made a significant contribution to the delineation of the resected region in 58.8% of the patients for
whom the VEEG results failed to provide any localization information.

a similar conclusion in their study of 49 patients who differ from those that aim at the identification of epilep-
underwent both MEG and invasive VEEG. togenic zones in that the phenomenon that exemplifies
Efficiency of MEG has been demonstrated for both the function under investigation is repeated several times
temporal3,7,32,33 and nontemporal lobe epilepsy,34–37 al- while the magnetic flux around the head is sampled at
though in general, interictal magnetic spikes can be regular intervals. An external stimulus is invariably pre-
recorded more readily in neocortical epilepsy than in sented at each instance in order either to induce the phe-
mesial temporal lobe epilepsy.2,38 nomenon (in the case of somatosensory and receptive
MEG studies have been successfully employed in language functions) or to act as a time cue (in the case of
helping to determine or confirm targets for surgical re- expressive language and motor functions). Each segment
section in a variety of epileptic syndromes, such as focal of recorded activity, beginning a few milliseconds before
cortical dysplasia,39–47 tuberous sclerosis complex,48–52 and extending up to approximately 1000 milliseconds (in
pediatric epilepsy,23,27,35,53–58 and conditions that of- the case of language-specific cortex mapping) and up to
ten cause seizures such as cavernomas,59 and mass 200 milliseconds (in the case of visual, auditory, and
lesions.23,60,61 For surgical treatment, MEG localizations somatosensory cortex mapping) after each repetition of
have been successfully combined with neuronaviga- the stimulus, is stored separately as a MEG epoch. The
tional systems.62–64 resulting averaged event-related field (ERF) consists of
Finally, exploratory MEG studies have been con- early components (50–200 milliseconds poststimulus on-
ducted in search of biological markers of focal brain set) that correspond to activation of the sensory cortex
damage associated with traumatic brain injury,65 brain specific to the stimulus modality, and late components
lesions,66 cerebrovascular disease6,67–69 and tumors,70 (200 – ∼800 milliseconds poststimulus onset), reflecting
as well as progressive degenerative disease. For in- activation of the association cortex or higher functions.
stance, enhanced power of slow-wave activity (in the The intracranial sources that give rise to the recorded
theta and delta bands) has been reported in patients ERFs at each point in time can be estimated and super-
with Alzheimer’s disease.71,72 More recent results pro- imposed on an MRI of the patient’s brain by applying
vided promising results highlighting the potential utility the same algorithms and procedures used for localizing
of MEG, in conjunction with magnetic resonance spec- the sources of abnormal spontaneous activity, described
troscopic as an adjunct in the evaluation for dementia.73 previously.

䉴 EVOKED MAGNETIC ACTIVITY: 䉴 SOMATOSENSORY EVOKED


PRESURGICAL MAPPING OF MAGNETIC FIELDS (SEFs)
ELOQUENT CORTEX
The spatial resolution of MEG is adequate to reveal
MEG recordings performed for the purpose of determin- the somatotopic organization of primary somatosensory
ing the location and extent of cortex mediating visual, cortex and the technique is used clinically for pre-
auditory, somatosensory, motor, and language functions surgical planning.74–78 The sources of the early and
CHAPTER 10 CLINICAL APPLICATIONS OF MEG IN NEUROLOGY AND NEUROSURGERY 123

middle-latency components of magnetic responses (oc- sources that give rise to the early component of the
curring <60 milliseconds after stimulus onset) obtained magnetic waveform evoked by rapidly changing pat-
through separate mechanical stimulation of each finger, terned stimulation are used as markers of the primary
toe, and of the perioral area (usually the corner of the visual area.97–102 Hemifield or single quadrant stimula-
lower lip) and modeled as successive, single equivalent tion has been successfully used to localize visual cortex
current dipoles (ECDs), originate from the contralateral in patients with organic brain diseases before surgical
primary sensory cortex within the central sulcus (mostly interventions such as craniotomy,74,103 and stereotactic
area 3b). Independent confirmation of the accuracy of procedures.104
localization of the primary somatosensory area (and con-
sequently of the central sulcus) has been obtained in
several studies through direct comparisons with the re- 䉴 AUDITORY EVOKED MAGNETIC
sults of intra- or extraoperative corticography.77,78,80–82 FIELDS (AEFs)
Moreover, MEG has proven sensitive to alterations of
the typical anatomical organization of the somatosen- Determining the precise location of the primary auditory
sory cortex in patients with mass lesions in the vicinity cortex may be useful for planning resections of mass le-
of the central sulcus.77,83–88 sions that encroach on the supratemporal plane.105,106 In
many cases application of AEFs can be evoked by bin-
aural stimulation with meaningful stimuli in the context
䉴 MOVEMENT-RELATED of language mapping.
MAGNETIC FIELDS (MRFs)
Activation protocols permitting identification of motor 䉴 LANGUAGE-RELATED BRAIN
cortex have also been developed.89–90 Movement-related MAGNETIC FIELDS (LRFs)
magnetic fields (MRFs) preceding voluntary movement
are generated by neurons oriented tangential to the scalp Recording of language-related brain magnetic fields
in motor cortex contra lateral to the movement, and are has two main clinical indications: (1) to establish the
recorded much more clearly than EEG.91–96 Typically, profile of hemispheric involvement in the mechanism
somatosensory and motor mapping are performed in that supports basic language functions (hemispheric
separate sessions although, recently, a procedure for dominance107–109 ) and (2) to identify the location and
mapping both somatosensory and motor functions in extent of functionally intact cortex involved in language
the same session has been developed.76 According to functions in relation to the area to be resected (epilep-
this protocol mechanical tactile stimulation is applied to togenic zone or mass lesion103,110–112 ).
the subject’s index finger and serves as a cue for the
individual to perform a full wrist extension. Tactile stim-
ulation activates initially the contralateral primary so-
ESTABLISHING HEMISPHERIC
matosensory cortex followed by activation of the sec-
DOMINANCE FOR LANGUAGE
ondary somatosensory cortex, lasting for approximately
150 milliseconds. Activity in the contralateral precentral
With the advent of functional imaging methods, there is
gyrus is seen next immediately preceding the onset of
great potential for replacing the Wada with a less inva-
electromyographic activity marking the onset of finger
sive procedure.113–116 Validation studies of the accuracy
movement. Finally, somatosensory activity is again ob-
of fMRI, which is currently the most commonly used
served during the movement presumably the result of
hemodynamic imaging technique for mapping eloquent
proprioceptive input. The location estimates of the mo-
cortex, have shown good concordance with the Wada
tor cortex were subsequently verified in six patients with
procedure and with intraoperative electrocortical stimu-
perirolandic lesions through intraoperative electrocorti-
lation mapping (ioESM), under particular conditions and
cal stimulation (see Fig. 10–2).
with some exceptions (for recent reviews see Billingsley
and Simos,117 Papanicolaou,108 Bookheimer,118 Pelletier
et al.119 ).
䉴 VISUAL EVOKED MAGNETIC Using MEG as a method of functional imaging, oth-
FIELDS (VEFs) ers and we have verified that MEG assessments of hemi-
spheric dominance for language are concordant with
Mapping the precise location of the primary visual cor- those based on the Wada procedure.107,109,120 Szyman-
tex is often required prior to resecting mass lesions ski and colleagues109 showed that the duration of late
in the vicinity of the calcarine fissure. The intracranial components of event-related magnetic fields elicited by
124 SECTION I TECHNIQUES

A B

CS
LH

M
S

anterior

Les-

posterior
C D

Figure 10–2. Intraoperative verification of magnetoencephalography (MEG)-derived


estimates for primary somatosensory (S) and motor (M) cortex. (A) Axial view of
patient’s magnetic resonance imaging (MRI) were the MEG-derived estimates for
sensory (black dot) and motor (black square) are projected in proximity to the
patient’s lesion (cavernous angioma). (B) Intraoperative photograph showing the
placement of a strip of electrodes for sensory and motor mapping. (C) Schematic
diagram representing the spatial relation between the lesion (Les.), the estimated
dipoles, and the positive areas for sensory (S) and motor (M) function as were derived
from the invasive recordings. (D) Photograph taken after ressecting the cavernous
angioma showing the identified sensory (S) and motor (M) areas. CS, central sulcus;
LH, left hemisphere. (Image reproduced with permission from Castillo EM, Simos PG,
Wheless JW, et al. Integrating sensory and motor mapping in a comprehensive MEG
protocol: clinical validity and replicability. Neuroimage 2004;21(3):973–983).

vowel stimuli demonstrated hemispheric asymmetries Most recently the sensitivity and selectivity of our
concordant with hemispheric dominance patterns de- noninvasive language mapping procedure for determin-
rived from the Wada procedure. Studies using a 148- ing hemispheric dominance was assessed in a series of
channel whole-head MEG system and a continuous 100 surgical candidates aged 8–56 years with intractable
word recognition task to elicit brain activity, have also epilepsy.108 The degree of language-specific activity was
found excellent concordance between the MEG lateral- indexed by the number of consecutive sources (modeled
ity estimates and the results of the Wada procedure in as single, ECDs, or ECDs) in perisylvian brain areas, as
children, adolescents, and adults.107 Using identical stim- in the aforementioned studies. Only late-component ac-
ulation and analysis procedures, Maestú et al.120 repli- tivity sources that were observed with a high degree
cated these results independently for Spanish-speaking of spatial and temporal overlap in two “split-half” data
patients. sets were used to compute the MEG laterality index.
CHAPTER 10 CLINICAL APPLICATIONS OF MEG IN NEUROLOGY AND NEUROSURGERY 125

Independently, all patients had Wada testing performed a particular sound (phonemic cue condition). Although
for the determination of hemispheric language domi- this study provided new insights into the timing of corti-
nance. There was a high degree of concordance (87%) cal interactions during expressive speech tasks, the num-
between independent clinical judgments based on MEG ber of activity sources in Broca’s area was not as abun-
and Wada data. MEG laterality judgments had an overall dant as we have previously found in posterior speech
sensitivity of .98 and a lower selectivity of .83, due to the regions during the performance of receptive language
fact that MEG detected more activity in the nondominant tasks, thereby rendering this task, if used alone, insuffi-
hemisphere than would be predicted on the basis of the cient for routine presurgical patient mapping. We plan
results of the Wada procedure. The same activation pro- to determine whether combining the verbal fluency and
tocol yields reliable estimates of the location of receptive the naming tasks is sufficient to fulfill that stringent re-
language-specific cortex (Wernicke’s area), and the ac- quirement.
curacy of these estimates have been verified against the Kober et al.121 have also reported success in elic-
results of invasive intro- or extraoperative electrocortical iting inferior frontal activity using a picture naming
stimulation mapping.111,112 Kober et al.121 and Bowyer task, using spatial filtering rather than the single ECD
et al.122 have also reported success in obtaining estimates method.107,111,112,123 Kober et al.’s spatial localization and
of hemispheric laterality during performance of expres- time course results were consistent with the Wernicke-
sive language tasks, which will be discussed in more Geschwind model of language organization. Moreover,
detail later. they showed a high degree of concordance between
results from spatial filtering and single ECD modeling,
which has been externally validated.107,112 As currently
FUNCTIONAL MAPPING OF THE utilized, the single ECD approach permits localization of
EXPRESSIVE LANGUAGE-SPECIFIC up to three simultaneous activity sources in each hemi-
CORTEX sphere, provided that they are located at least 3–4 cm
apart and the orientation of each dipolar source results
Although mapping of receptive language cortex was in visually distinguishable surface isofield maps.
found to be sufficient for the purpose of assessing hemi- More recently, Bowyer et al.122 using yet another
spheric dominance for language, it is often necessary mathematical procedure for estimating the underlying
to determine the location of expressive language cor- cortical sources of magnetic fields, namely multires-
tex in relation to diseased cortex. Several groups have olution focal underdetermined system solution (MR-
thus far reported success in eliciting prefrontal magnetic FOCUSS), showed in a group of 24 epileptic patients and
activity in the vicinity of Broca’s area, associated with 18 control subjects that this current density imaging tech-
the performance of a variety of expressive language nique can provide reasonable localization of language
tasks. cortex. Using this technique, all participants showed ac-
Castillo et al.123 used a picture naming task and tivation of the left superior temporal gyrus during a word
MEG analysis procedures identical to those employed generation task and the left inferior frontal gyrus during
earlier in the context of receptive language mapping to a picture naming task. Although the spatial filtering and
derive maps of activation related to expressive language current source density methods have not been externally
function. The results for seven normal volunteers and validated for language mapping against the Wada or
nine patients with epilepsy can be summarized as fol- electrocortical stimulation mapping, they hold promise
lows: (1) The task resulted in activation of the expressive for detecting multiple, simultaneous sources contribut-
language-specific cortex (Broca’s area) in only a fraction ing to late components of ERFs during expressive and
of the cases (in 3 of the 9 patients and 3 of the 7 neuro- receptive. Promising results in the same direction have
logically intact participants). Consequently, it cannot be also been reported by Hirata et al.125 using a different
used alone for routine identification of Broca’s area. (2) algorithm for source estimation. Oscillatory activity as-
When prefrontal activity sources were identified, their sociated with reading was recorded in 20 patients ob-
location matched very closely with estimates of the loca- taining good agreement with the results of the Wada
tion of Broca’s area based on electrocortical stimulation test.
mapping.123 Clearly, future developments will decide on the
Using a different task (word generation to phone- most efficient method for assessing laterality as well as
mic or semantic cues) and identical analysis procedures, precise location of the cortical circuitry subserving ex-
Billingsley et al.124 reported consistent activity in the pressive and receptive language as well as other cog-
vicinity of Broca’s area in neurologically intact partic- nitive functions. At present, however, the ECD model
ipants. The duration of this activity was consistently appears to be the safest to use for all types of presur-
greater in the left as compared to the right hemisphere gical evaluation, from determination of epileptogenic
and this hemispheric asymmetry was larger when partic- zones to localization of the sensory, motor and language
ipants were asked to generate a word that started with cortex.
126 SECTION I TECHNIQUES

䉴 ACKNOWLEDGMENT cinations of status epilepticus. J Neurol Neurosurg Psychi-


atry 2003;74(4):525-527.
The research presented in this chapter was partly 14. Stefan H, Schneider S, Feistel H, et al. Ictal and in-
supported by NIH Grant RO1 NS46588 to the last terictal activity in partial epilepsy recorded with multi-
channel magnetoelectroencephalography: correlation of
author.
electroencephalography/electrocorticography, magnetic
resonance imaging, single photon emission computed to-
mography, and positron emission tomography. Epilepsia
REFERENCES 1992;33(5):874-887.
15. Tilz C, Hummel C, Kettenmann B, Stefan H. Ictal onset
1. Papanicolaou AC. Fundamentals of Functional Brain localization of epileptic seizures by magnetoencephalog-
Imaging: A Guide to the Methods and their Applications to raphy. Acta Neurol Scand 2002;106(4):190-195.
Psychology and Behavioral Neurosciences. Netherlands: 16. Yoshinaga H, Ohtsuka Y, Watanabe Y, et al. Ictal MEG in
Swets & Zeitlinger, 1998. two children with partial seizures. Brain Dev 2004;26(6):
2. Knowlton RC, Laxer KD, Aminoff MJ, Roberts TP, Wong 403-408.
ST, Rowley HA. Magnetoencephalography in partial 17. Ishibashi H, Morioka T, Shigeto H, Nishio S, Yamamoto T,
epilepsy: clinical yield and localization accuracy. Ann Fukui M. Three-dimensional localization of subclini-
Neurol 1997;42(4):622-631. cal ictal activity by magnetoencephalography: correla-
3. Pataraia E, Simos PG, Billingsley RL, Sarkari S, Castillo EM, tion with invasive monitoring. Surg Neurol 1998;50(2):
Swank PR, Breier JI, Maggio V, Wheless JM, Papanico- 157-163.
laou AC. Does MEG improve standard noninvasive elec- 18. Sutherling WW, Crandall PH, Engel J Jr, Darcey TM, Ca-
trophysiological methods in presurgical epilepsy evalua- han LD, Barth DS. The magnetic field of complex partial
tion? Neurology 2004;62,943-948. seizures agrees with intracranial localizations. Ann Neurol
4. Ebersole JS. Classification of MEG spikes in temporal lobe 1987;21(6):548-558.
epilepsy. In Recent Advances in Biomagnetism, edited by 19. Ko DY, Kufta C, Scaffidi D, Sato S. Source localization
Yoshimoto T, Kotani M, Kuriki S, Karibe H, Nakasoto N. determined by magnetoencephalography and electroen-
Sendai: Tohoku University Press, 1999, pp. 758-761. cephalography in temporal lobe epilepsy: comparison
5. Brockhaus A, Lehnertz K, Wienbruch C, et al. Possi- with electrocorticography: technical case report. Neuro-
bilities and limitations of magnetic source imaging of surgery 1998;42(2):414-421.
methohexital-induced epileptiform patterns in temporal 20. Wheless JW, Castillo EM, Maggio V, et al. Magnetoen-
lobe epilepsy patients. Electroencephalogr Clin Neuro- cephalography (MEG) and Magnetic Source Imaging
physiol 1997;102:423-436. (MSI). Neurologist 2004;10(3):1-16.
6. Butz M, Gross J, Timmermann L, et al. Perilesional patho- 21. Fischer MJ, Scheler G, Stefan H. Utilization of magnetoen-
logical oscillatory activity in the magnetoencephalogram cephalography results to obtain favourable outcomes in
of patients with cortical brain lesions. Neurosci Lett 2004; epilepsy surgery. Brain 2005;128(1):153-157.
355(1):93-96. 22. Papanicolaou AC, Pataraia E, Billingsley-Marshall R, et al.
7. Fernandez A, de Sola RG, Amo C, et al. Dipole den- Toward the substitution of invasive electroencephalogra-
sity of low-frequency and spike magnetic activity: a reli- phy in epilepsy surgery. J Clin Neurophysiol 2005;22(4):
able procedure in presurgical evaluation of temporal lobe 231-237.
epilepsy. J Clin Neurophysiol 2004;21(4):254-266. 23. Morioka T, Nishio S, Hisada K, et al. Neuromagnetic as-
8. Gallen CC, Tecoma E, Iragui V, Sobel DF, Schwartz BJ, sessment of epileptogenicity in cerebral arteriovenous
Bloom FE. Magnetic source imaging of abnormal low- malformation. Neurosurg Rev 2000;23(4):206-212.
frequency magnetic activity in presurgical evaluations of 24. Nakasato N, Levesque MF, Barth DS, Baumgartner C,
epilepsy. Epilepsia 1997;38(4):452-460. Rogers RL, Sutherling WW. Comparisons of MEG, EEG,
9. Ishibashi H, Simos PG, Castillo EM, et al. Detection and and ECoG source localization in neocortical partial
significance of focal, interictal, slow-wave activity visual- epilepsy in humans. Electroencephalogr Clin Neurophys-
ized by magnetoencephalography for localization of a pri- iol 1994;91(3):171-178.
mary epileptogenic region. J Neurosurg 2002a;96(4):724- 25. Sutherling WW, Crandall PH, Cahan LD, Barth DS. The
730. magnetic field of epileptic spikes agrees with intracra-
10. Kamada K, Saguer M, Moller M, et al. Functional and nial localizations in complex partial epilepsy. Neurology
metabolic analysis of cerebral ischemia using magne- 1988;38(5):778-786.
toencephalography and proton magnetic resonance spec- 26. Stefan H, Hummel C, Scheler G, et al. Magnetic brain
troscopy. Ann Neurol 1997;42(4):554-563. source imaging of focal epileptic activity: a synopsis of
11. Assaf BA, Karkar KM, Laxer KD, et al. Ictal magne- 455 cases. Brain 2003;126(11):2396-2405.
toencephalography in temporal and extratemporal lobe 27. Paetau R, Hamalainen M, Hari R, et al. Magnetoencephalo-
epilepsy. Epilepsia 2003;44(10):1321-1328. graphic evaluation of children and adolescents with in-
12. Eliashiv DS, Elsas SM, Squires K, Fried I, Engel J Jr. Ictal tractable epilepsy. Epilepsia 1994;35(2):275-284.
magnetic source imaging as a localizing tool in partial 28. Smith JR, Schwartz BJ, Gallen C, et al. Multichannel mag-
epilepsy. Neurology 2002;59(10):1600-1610. netoencephalography in ablative seizure surgery outside
13. Oishi M, Otsubo H, Kameyama S, et al. Ictal magnetoen- the anteromesial temporal lobe. Stereotact Funct Neuro-
cephalographic discharges from elementary visual hallu- surg 1995;65(1-4):81-85.
CHAPTER 10 CLINICAL APPLICATIONS OF MEG IN NEUROLOGY AND NEUROSURGERY 127

29. Minassian BA, Otsubo H, Weiss S, Elliott I, Rutka JT, Snead cephalography, electrocorticography, and magnetoen-
OC 3rd. Magnetoencephalographic localization in pedi- cephalography. J Child Neurol 2005;20(4):357-363.
atric epilepsy surgery: comparison with invasive intracra- 45. Taniguchi M, Yoshimine T, Kato A, et al. Dysembryoplastic
nial electroencephalography. Ann Neurol 1999;46(4):627- neuroepithelial tumor in the insular cortex. Three dimen-
633. sional magnetoencephalographic localization of epileptic
30. Otsubo H, Sharma R, Elliott I, Holowka S, Rutka JT, Snead discharges. Neurol Res 1998b;20(5):433-438.
OC 3rd. Confirmation of two magnetoencephalographic 46. Toulouse P, Agulhon CK, Taussig D, et al. Magnetoen-
epileptic foci by invasive monitoring from subdural elec- cephalographic studies of two cases of diffuse subcortical
trodes in an adolescent with right frontocentral epilepsy. laminar heterotopia or so-called double cortex. Neuroim-
Epilepsia 1999;40(5):608-613. age 2003;19(4):1251-1259.
31. Knowlton RC, Elgavish R, Howell J, et al. Magnetic 47. Zhang W, Simos PG, Ishibashi H, et al. Multimodality neu-
source imaging versus intracranial electroencephalogram roimaging evaluation improves the detection of subtle
in epilepsy surgery: a prospective study. Ann Neurol 2006; cortical dysplasia in seizure patients. Neurol Res 2003;
59(5):835-842. 25(1):53-57.
32. Iwasaki M, Nakasato N, Shamoto H, et al. Surgical impli- 48. Iida K, Otsubo H, Mohamed I, et al. Characterizing
cations of neuromagnetic spike localization in temporal magnetoencephalographic spike sources in children with
lobe epilepsy. Epilepsia 2002;43(4):415-424. tuberous sclerosis complex. Epilepsia 2005a;46(9):1510-
33. Stefan H, Schuler P, Abraham-Fuchs K, et al. Magnetic 1517.
source localization and morphological changes in tempo- 49. Jansen FE, Huiskamp G, van Huffelen AC, et al. Identifica-
ral lobe epilepsy: comparison of MEG/EEG, ECoG and tion of the epileptogenic tuber in patients with tuberous
volumetric MRI in presurgical evaluation of operated pa- sclerosis: a comparison of high-resolution EEG and MEG.
tients. Acta Neurol Scand Suppl 1994;152:83-88. Epilepsia 2006;47(1):108-114.
34. Mamelak AN, Lopez N, Akhtari M, Sutherling WW. 50. Kamimura T, Tohyama J, Oishi M, et al. Magnetoen-
Magnetoencephalography-directed surgery in patients cephalography in patients with tuberous sclerosis and
with neocortical epilepsy. J Neurosurg 2002;97(4):865- localization-related epilepsy. Epilepsia 2006;47(6):991-
873. 997.
35. Otsubo H, Ochi A, Elliott I, et al. MEG predicts epileptic 51. Wu JY, Sutherling WW, Koh S, et al. Magnetic source imag-
zone in lesional extrahippocampal epilepsy: 12 pediatric ing localizes epileptogenic zone in children with tuberous
surgery cases. Epilepsia 2001;42(12):1523-1530. sclerosis complex. Neurology 2006;66(8):1270-1272.
36. Shiraishi H, Watanabe Y, Watanabe M, Inoue Y, Fujiwara 52. Xiao Z, Xiang J, Holowka S, et al. Volumetric localization
T, Yagi K. Interictal and ictal magnetoencephalographic of epileptic activities in tuberous sclerosis using synthetic
study in patients with medial frontal lobe epilepsy. Epilep- aperture magnetometry. Pediatr Radiol 2006;36(1):16-21.
sia 2001;42(7):875-882. 53. Hattori H, Yamano T, Tsutada T, Tsuyuguchi N, Kawawaki
37. Stefan H, Hummel C, Hopfengartner R, et al. Magnetoen- H, Shimogawara M. Magnetoencephalography in the de-
cephalography in extratemporal epilepsy. J Clin Neuro- tection of focal lesions in West syndrome. Brain Dev 2001;
physiol 2000;17(2):190-200. 23(7):528-532.
38. Baumgartner C, Pataraia E, Lindinger G, Deecke L. Neu- 54. Ishibashi H, Simos PG, Wheless JW, et al. Multimodal-
romagnetic recordings in temporal lobe epilepsy. J Clin ity functional imaging evaluation in a patient with Ras-
Neurophysiol 2000;17:177-189. mussen’s encephalitis. Brain Dev 2002b;24(4):239-244.
39. Bast T, Oezkan O, Rona S, et al. EEG and MEG source 55. Otsubo H, Snead OC 3rd. Magnetoencephalography and
analysis of single and averaged interictal spikes reveals in- magnetic source imaging in children. J Child Neurol 2001;
trinsic epileptogenicity in focal cortical dysplasia. Epilep- 16(4):227-235.
sia 2004;45(6):621-631. 56. Paetau R, Granstrom ML, Blomstedt G, Jousmaki V,
40. Burneo JG, Bebin M, Kuzniecky RI, Knowlton RC. Electro- Korkman M, Liukkonen E. Magnetoencephalography in
clinical and magnetoencephalographic studies in epilepsy presurgical evaluation of children with the Landau-
patients with polymicrogyria. Epilepsy Res 2004a;62 Kleffner syndrome. Epilepsia 1999;40(3):326-335.
(2–3):125-133. 57. Verrotti A, Pizzella V, Trotta D, Madonna L, Chiarelli F,
41. Ishibashi H, Simos PG, Wheless JE, et al. Somatosensory Romani GL. Magnetoencephalography in pediatric neu-
evoked magnetic fields in hemimegalencephaly. Neurol rology and in epileptic syndromes. Pediatr Neurol 2003b;
Res 2002c;24(5):459-462. 28(4):253-261.
42. Minami T, Tasaki K, Yamamoto T, Gondo K, Yanai S, 58. Yanai S, Minami T, Yamamoto T, et al. Magnetoencephalo-
Ueda K. Magneto-encephalographic analysis of focal cor- graphic analysis of hypsarrhythmia in West syndrome.
tical heterotopia. Dev Med Child Neurol 1996;38(10):945- J Epilepsy 1997;10:131-138.
949. 59. Stefan H, Scheler G, Hummel C, et al. Magnetoen-
43. Morioka T, Nishio S, Ishibashi H, et al. Intrinsic epilep- cephalography (MEG) predicts focal epileptogenicity in
togenicity of focal cortical dysplasia as revealed by mag- cavernomas. J Neurol Neurosurg Psychiatry 2004;75(9):
netoencephalography and electrocorticography. Epilepsy 1309-1313.
Res 1999;33(2-3):177-187. 60. Amo C, Saldana C, Hidalgo MG, et al. Magnetoencephalo-
44. Otsubo H, Iida K, Oishi M, et al. Neurophysiologic graphic localization of peritumoral temporal epileptic
findings of neuronal migration disorders: intrinsic epilep- focus previous surgical resection. Seizure 2003;12(1):
togenicity of focal cortical dysplasia on electroen- 19-22.
128 SECTION I TECHNIQUES

61. Patt S, Steenbeck J, Hochstetter A, et al. Source localiza- 76. Jannin P, Morandi X, Fleig OJ, et al. Integration of sulcal
tion and possible causes of interictal epileptic activity in and functional information for multimodal neuronaviga-
tumor-associated epilepsy. Neurobiol Dis 2000;7(4):260- tion J Neurosurg 2002;96(4):713-723.
269. 77. Roberts TP, Zusman E, McDermott M, Barbaro N, Rowley
62. Gross RE, Dean A, Lewine J, et al. The relationship of HA. Correlation of functional magnetic source imaging
magnetic source imaging to ictal electrocorticography in a with intraoperative cortical stimulation in neurosurgical
neuronavigational workspace. Stereotact Funct Neurosurg patients. J Image Guid Surg 1995;1(6):339-347.
1999;73(4):109-114. 78. Roberts TP, Ferrari P, Perry D, Rowley HA, Berger MS.
63. Duffner F, Freudenstein D, Schiffbauer H, et al. Com- Presurgical mapping with magnetic source imaging: com-
bining MEG and MRI with neuronavigation for treat- parisons with intraoperative findings. Brain Tumor Pathol
ment of an epileptiform spike focus in the precentral 2000;17(2):57-64.
region: a technical case report. Surg Neurol 2003;59(1): 79. Castillo EM, Simos PG, Wheless JW, et al. Integrating sen-
40-45. sory and motor mapping in a comprehensive MEG pro-
64. Iida K, Otsubo H, Matsumoto Y, et al. Characterizing mag- tocol: clinical validity and replicability. Neuroimage 2004;
netic spike sources by using magnetoencephalography- 21(3):973-983.
guided neuronavigation in epilepsy surgery in pediatric 80. Gallen CC, Schwartz BJ, Bucholz RD, Malik G, Barkley
patients. J Neurosurg 2005;102(2):187-196. GL, Smith J, Tung H, Copeland B, Bruno L, Assam S, et al.
65. Lewine JD, Davis JT, Sloan JH, Kodituwakku PW, Orrison Presurgical localization of functional cortex using mag-
WW Jr. Neuromagnetic assessment of pathophysiologic netic source imaging. J Neurosurg. 1995;82(6):988-994.
brain activity induced by minor head trauma. AJNR Am J 81. Ishibashi H, Morioka T, Nishio S, Shigeto H, Yamamoto T,
Neuroradiol 1999;20(5):857-866. Fukui M. Magnetoencephalographic investigation of so-
66. Vieth JB, Kober H, Grummich P. Sources of spontaneous matosensory homunculus in patients with peri-rolandic
slow waves associated with brain lesions, localized by tumors. Neurol Res 2001;23(1):29-38.
using the MEG. Brain Topogr 1996a;8(3):215-221. 82. Morioka T, Yamamoto T, Katsuta T, Fujii K, Fukui M.
67. Seki S, Nakasato N, Ohtomo S, Kanno A, Shimizu H, Presurgical three-dimensional magnetic source imaging
Tominaga T. Neuromagnetic measurement of unilateral of the somatosensory cortex in a patient with a peri-
temporo-parietal theta rhythm in patients with internal Rolandic lesion: technical note. Neurosurgery 1994;34(5):
carotid artery occlusive disease. Neuroimage 2005;25(2): 930-933.
502-510. 83. Schiffbauer H, Berger MS, Ferrari P, Freudenstein D, Row-
68. Stippich C, Kassubek J, Kober H, Soros P, Vieth JB. ley HA, Roberts TP. Preoperative magnetic source imaging
Time course of focal slow wave activity in transient is- for brain tumor surgery: a quantitative comparison with
chemic attacks and transient global amnesia as measured intraoperative sensory and motor mapping. J Neurosurg
by magnetoencephalography. Neuroreport 2000;11(15): 2002;97(6):1333-1342.
3309-3313. 84. Breier JI, Simos PG, Zouridakis G, et al. A MEG study of
69. Vieth J, Kober H, Weise E, et al. Functional 3D localiza- cortical plasticity. Neurocase 1999b;5:277-284.
tion of cerebrovascular accidents by magnetoencephalog- 85. Burneo JG, Kuzniecky RI, Bebin M, Knowlton RC. Cor-
raphy (MEG). Neurol Res 1992;14(Suppl 2):132-134. tical reorganization in malformations of cortical devel-
70. de Jongh A, Baayen JC, de Munck JC, Heethaar RM, Van- opment: a magnetoencephalographic study. Neurology
dertop WP, Stam CJ. The influence of brain tumor treat- 2004b;63(10):1818-1824.
ment on pathological delta activity in MEG. Neuroimage 86. Oishi M, Fukuda M, Kameyama S, Kawaguchi T, Masuda
2003;20(4):2291-2301. H, Tanaka R. Magnetoencephalographic representation of
71. Fernandez A, Maestu F, Amo C, et al. Focal temporopari- the sensorimotor hand area in cases of intracerebral tu-
etal slow activity in Alzheimer’s disease revealed by mag- mour. J Neurol Neurosurg Psychiatry 2003b;74(12):1649-
netoencephalography. Biol Psychiatry 2002;52(7):764- 1654.
770. 87. Papanicolaou AC, Simos PG, Breier JI, et al. Brain plastic-
72. Franciotti R, Iacono D, Della Penna S, et al. Corti- ity for sensory and linguistic functions: a functional imag-
cal rhythms reactivity in AD, LBD and normal subjects: ing study using magnetoencephalography with children
a quantitative MEG study. Neurobiol Aging 2006;27(8): and young adults. J Child Neurol 2001;16(4):241-252.
1100-1109. 88. Vates GE, Lawton MT, Wilson CB, et al. Magnetic source
73. Fernandez A, Rodriguez-Palancas A, Lopez-Ibor M, et al. imaging demonstrates altered cortical distribution of func-
Increased occipital delta dipole density in major depres- tion in patients with arteriovenous malformations. Neuro-
sive disorder determined by magnetoencephalography. surgery 2002;51(3):614-627.
J Psychiatry Neurosci 2005;30(1):17-23. 89. Kassubeck J, Stippich C, Soros P. A motor field local-
74. Alberstone CD, Skirboll SL, Benzel EC, et al. Magnetic ization protocol using magnetoencephalography. Biomed
source imaging and brain surgery: presurgical and intra- Eng 1996;41:334-335.
operative planning in 26 patients. J Neurosurg 2000;92(1): 90. Ganslandt O, Fahlbusch R, Nimsky C, et al. Func-
79-90. tional neuronavigation with magnetoencephalography:
75. Hund M, Rezai AR, Kronberg E, et al. Magnetoencephalo- outcome in 50 patients with lesions around the motor
graphic mapping: basic of a new functional risk profile in cortex. J Neurosurg 1999;91(1):73-79.
the selection of patients with cortical brain lesions. Neu- 91. Gerloff C, Uenishi N, Nagamine T, Kunieda T, Hallett
rosurgery 1997;40(5):936-942. M, Shibasaki H. Cortical activation during fast repetitive
CHAPTER 10 CLINICAL APPLICATIONS OF MEG IN NEUROLOGY AND NEUROSURGERY 129

finger movements in humans: steady-state movement- 105. Nakasato N, Fujita S, Seki K, et al. Functional localization
related magnetic fields and their cortical generators. Elec- of bilateral auditory cortices using an MRI-linked whole
troencephalogr Clin Neurophysiol 1998;109(5):444-453. head magnetoencephalography (MEG) system. Electroen-
92. Hashimoto I, Mashiko T, Odaka K, Imada T, Mizuta T, cephalogr Clin Neurophysiol 1995;94(3):183-190.
Tomarikawa K. Bilateral activation of the human motor 106. Suzuki K, Okuda J, Nakasato N, et al. Auditory evoked
cortex preceding unilateral voluntary finger extension as magnetic fields in patients with right hemisphere language
evidenced by magnetic measurements. In Biomagnetism: dominance. Neuroreport 1997;8(15):3363-3366.
Fundamental Research and Clinical Applications, edited 107. Breier JI, Simos PG, Zouridakis G, et al. Language dom-
by Baumgartner C, Deecke L, Stroink G, Williamson SJ. inance determined by magnetic source imaging: a com-
Amsterdam: Elsevier, 1995, pp. 131-135. parison with the Wada procedure. Neurology 1999a;53(5):
93. Hoshiyama M, Kakigi R, Berg P, et al. Identification of 938-945.
motor and sensory brain activities during unilateral finger 108. Papanicolaou AC, Simos PG, Castillo EM, et al. Magne-
movement: spatio-temporal source analysis of movement toencephalography: a noninvasive alternative to the Wada
associated magnetic fields. Exp Brain Res 1997;115(1): procedure. J Neurosurg 2004;100(5):867-876.
6-14. 109. Szymanski MD, Perry DW, Gage NM, et al. Magnetic
94. Nagamine T, Toro C, Balish M, et al. Cortical magnetic source imaging of late evoked field responses to vow-
and electric fields associated with voluntary finger move- els: toward an assessment of hemispheric dominance for
ments. Brain Topogr 1994;6(3):175-183. language. J Neurosurg 2001;94(3):445-453.
95. Nakasato N, Itoh H, Hatanaka K, Nakahara H, Kanno A, 110. Grummich P, Nimsky C, Pauli E, Buchfelder M, Ganslandt
Yoshimoto T. Movement-related magnetic fields to tongue O. Combining fMRI and MEG increases the reliability of
protrusion. Neuroimage 2001;14(4):924-935. presurgical language localization: a clinical study on the
96. Taniguchi M, Yoshimine T, Cheyne D, et al. Neuromag- difference between and congruence of both modalities.
netic fields preceding unilateral movements in dextrals Neuroimage 2006;32(4):1793-1803.
and sinistrals. Neuroreport 1998a;9(7):1497-1502. 111. Simos PG, Breier JI, Maggio WW, et al. Atypical tempo-
97. Barnikol UB, Amunts K, Dammers J, et al. Pattern rever- ral lobe language representation: MEG and intraopera-
sal visual evoked responses of V1/V2 and V5/MT as re- tive stimulation mapping correlation. Neuroreport 1999a;
vealed by MEG combined with probabilistic cytoarchitec- 10(1):139-142.
tonic maps. Neuroimage 2006;31(1):86-108. 112. Simos PG, Papanicolaou AC, Breier JI, et al. Localiza-
98. Hashimoto T, Kashii S, Kikuchi M, Honda Y, Nagamine T, tion of language-specific cortex by using magnetic source
Shibasaki H. Temporal profile of visual evoked responses imaging and electrical stimulation mapping. J Neurosurg
to pattern-reversal stimulation analyzed with a whole- 1999b;91(5):787-796.
head magnetometer. Exp Brain Res 1999;125(3):375- 113. Anderson DP, Harvey AS, Saling MM, et al. FMRI lateral-
382. ization of expressive language in children with cerebral
99. Hatanaka K, Nakasato N, Seki K, Mizoi K, Yoshimoto T. lesions. Epilepsia 2006;47:998-1008.
Striate cortical generators of the N75, P100 and N145 com- 114. Binder JR, Swanson SJ, Hammeke TA, et al. Determination
ponents localized by pattern reversal visual evoked mag- of language dominance using functional MRI, A compar-
netic fields. Tohoku J Exp Med 1997;182:9-14. ison with the Wada test. Neurology 1996;46:978-984.
100. Nakamura M, Kakigi R, Okusa T, Hoshiyama M, Watanabe 115. Cuenod CA, Brookheimer SY, Hertz-Pannier L, Zeffiro TA,
K. Effects of check size on pattern reversal visual evoked Theodore WH, Le Bihan D. Functional MRI during word
magnetic field and potential. Brain Res 2000;872(2): generation using conventional equipment: a potential tool
77-86. for language localization in the clinical environment. Neu-
101. Seki K, Nakasato N, Fujita S, et al. Neuromagnetic evi- rology 1995;45:1821-1827.
dence that the P100 component of pattern reversal visual 116. Stippich C, Rapps N, Dreyhaupt J, et al. Localizing
evoked response originates in the bottom of calcarine fis- and lateralizing language in patients with brain tumors:
sure. Electroencephalogr Clin Neurophysiol 1996;100(5): feasibility of routine preoperative functional MR imag-
436-442. ing in 81 consecutive patients. Radiology 2007;243:828-
102. Shigeto H, Tobimatsu S, Yamamoto T, Kobayashi T, Kato 836.
M. Visual evoked cortical magnetic responses to checker- 117. Billingsley RL, Simos PG, Castillo EM, et al. Spatio-
board pattern reversal stimulation: a study on the neu- temporal cortical dynamics of phonemic and semantic flu-
ral generators of N75, P100 and N145. J Neurol Sci 1998; ency. J Clin Exp Neuropsychol 2004;26:1031-1043.
156(2):186-194. 118. Bookheimer S. Pre-surgical language mapping with func-
103. Ganslandt O, Buchfelder M, Hastreiter P, Grummich P, tional magnetic resonance imaging. Neuropsychol Rev
Fahlbusch R, Nimsky C. Magnetic source imaging sup- 2007;17:145-155.
ports clinical decision making in glioma patients. Clin 119. Pelletier I, Sauerwein HC, Lepore F, Saint-Amour D, Las-
Neurol Neurosurg 2004;1007(1):20-26. sonde M. Non-invasive alternatives to the Wada test in
104. Inoue T, Fujimura M, Kumabe T, Nakasato N, Higano the presurgical evaluation of language and memory func-
S, Tominaga T. Combined three-dimensional anisotropy tions in epilepsy patients. Epileptic Disord 2007;9:111-
contrast imaging and magnetoencephalography guidance 126.
to preserve visual function in a patient with an occipi- 120. Maestu F, Ortiz T, Fernandez A, et al. Spanish language
tal lobe tumor. Minim Invasive Neurosurg 2004;47(4):249- mapping using MEG: a validation study. Neuroimage
252. 2002;17(3):1579-1586.
130 SECTION I TECHNIQUES

121. Kober H, Moller M, Nimsky C, Vieth J, Fahlbusch R, guage cortex using magnetic source imaging. Neurocase
Ganslandt O. New approach to localize speech relevant 2001;7(5):419-422.
brain areas and hemispheric dominance using spatially fil- 124. Billingsley RL, Simos PG, Papanicolaou AC. Reliability and
tered magnetoencephalography. Hum Brain Mapp 2001; validity of functional neuroimaging techniques for iden-
14(4):236-250. tifying language-critical areas in children and adults. Dev
122. Bowyer SM, Moran JE, Mason KM, et al. MEG localization Neuropsychol 2004;26:541-563.
of language-specific cortex utilizing MR-FOCUSS. Neurol- 125. Hirata M, Kato A, Taniguchi M, et al. Determination of lan-
ogy 2004;62:2247-2255. guage dominance with synthetic aperture magnetometry:
123. Castillo EM, Simos PG, Venkataraman V, Breier JI, Whe- comparison with the Wada test. Neuroimage 2004;23(1):
less JW, Papanicolaou AC. Mapping of expressive lan- 46-53.
Chapter 11
Optical Spectroscopic Imaging of the
Human Brain—Clinical Applications
Hongtao Ma, Minah Suh, Mingrui Zhao, Challon Perry, Andrew Geneslaw,
and Theodore H. Schwartz
Department of Neurosurgery, Weill Medical College of Cornell University, New York, New York

Clinical neuroscience relies on a variety of human brain the degree to which incident light is reflected by the
mapping techniques to aid in the diagnosis and manage- active cortex. These are: (1) blood volume changes;
ment of patients with neurological illnesses. Anatomic (2) changes in hemoglobin oxygenation; and (3) light
imaging techniques such as computed tomography (CT), scattering. Each optical imaging method measures these
ultrasound, and magnetic resonance imaging (MRI) pro- hemodynamic changes with different wavelengths. Optical
vide static renderings of brain parenchyma and vascular recording of intrinsic signal (ORIS) works on the visi-
anatomy at a precise moment in time. These techniques ble wavelength (450–700 nm), while near infrared spec-
can also be adapted to provide physiological information troscopy (NIRS) and diffuse optical tomography (DOT)
about cerebral blood flow (CBF) (xenon CT, Doppler use higher wavelengths (700–1100 nm). Depending on
and perfusion MRI, respectively). Functional mapping the optical window (i.e., intact skull vs. exposed cortex),
techniques, on the other hand, provide information these techniques can measure perfusion, oxygenation,
about the brain’s electrical activity, whether it be normal and light scatter with a spatial resolution of approxi-
cortical processing or abnormal activity such as epilepsy. mately 200 ␮m and a temporal resolution that is limited
These can be divided into techniques that map neuronal by the onset of the signal (tens to hundreds of mil-
activity directly and those that map it indirectly. Elec- liseconds). By measuring light at multiple wavelengths
troencephalography (EEG) and magnetoencephalogra- (i.e., spectroscopy), these techniques can quantify
phy measure the electrical or magnetic fields resulting hemoglobin in its oxygenated and deoxygenated states.
from the synchronized population activity of neuronal
tissue. While the temporal resolution of these techniques
is on the order of milliseconds, spatial resolution is of- 䉴 NEUROVASCULAR COUPLING
ten measured in centimeters and sampling is limited to
cortex adjacent to the electrodes. Indirect brain mapping The basis of functional imaging using intrinsic signals
techniques, on the other hand, such as functional MRI is the coupling between neuronal activity, metabolism,
(fMRI), positron emission tomography (PET) and single- oxygen utilization and perfusion. The idea that neu-
photon emission tomography (SPECT) measure nonelec- ronal activity, which causes an increase in local cerebral
trical physiological events that correlate with neuronal metabolic rate (CMRO2 ), would lead to a focal in-
activity such as metabolism, perfusion, and oxygenation. crease in perfusion is based on the work of Roy and
Although these techniques can sample the brain widely, Sherrington.2 They hypothesized that techniques which
both on the surface and at a depth, they have limited measure perfusion could be used to map neuronal ac-
spatial and temporal resolution, on the order of millime- tivity. More recently, Fox and Raichle demonstrated that
ters and seconds. In addition, correct interpretation of increases in CBF, which occur 1–2 seconds after the
these methods requires a comprehensive understanding neurons become active, were far greater than increases
of neurovascular coupling during both normal and ab- CMRO2 .3 This mismatch, or uncoupling of metabolism
normal brain activity, which is dynamic and complex. and blood flow, leads to a local increase in oxygenated
Optical spectroscopic imaging is based on measur- hemoglobin (HbO2 ), which also forms a significant com-
ing changes in the absorption of light in the brain and ponent of the BOLD signal used for brain mapping with
its blood vessels associated with neuronal activity. The fMRI, since differences in the paramagnetic properties
sources of this intrinsic signal include several optically of oxygenated and deoxygenated hemoglobin can be
active events,1 which correlate indirectly with neuronal measured with high magnetic fields.4 In more recent
activity. Characteristic physiological parameters affect years, it has become clear that before the increase in

131
132 SECTION I TECHNIQUES

CBF occurs, the focal increase in CMRO2 elicits an early evolution before it overperfuses the population of active
decrease in hemoglobin oxygenation (or increase in de- neurons,6,26,27 especially if one can separate the signal
oxygenated hemoglobin (Hbr)) called the “initial dip,” arising from the microvasculature from that arising from
which is spatially and temporally more focally related to the macrovasculature.28
the populations of active neurons than the later increase
in CBF and cerebral blood volume (CBV). This early dip
can be measured with ORIS,5,6 imaging spectroscopy,7,8 䉴 OPTICAL IMAGING TECHNIQUES
oxygen-dependent phosphorescence quenching,9 and
even high-tesla fMRI.10,11 However, the initial dip can OPTICAL RECORDING OF
be very difficult to measure in rodents during normal INTRINSIC SIGNAL
sensory processing and is quite sensitive to anesthe-
sia, systemic oxygenation, and blood pressure.12−14 Di- ORIS measures small changes in light absorption (or re-
rect measurements of tissue oxygenation with oxygen- flection) associated with neuronal activity. The brain is
sensitive electrodes, however, have shown that the dip exposed and either illuminated with visible light (400–
clearly exists.15−18 During epileptiform events, on the 650 nm) or light at a specific wavelength. If illuminated
other hand, in which a large population of neurons are with visible light, then emission filters are needed to se-
synchronously active and metabolism is dramatically in- lect the wavelength of interest for recording. The im-
creased, the dip in hemoglobin oxygenation is longer ages are then acquired with a CCD camera while the
and more profound.19−25 brain is activated, either by normal activity or patholog-
The existence of this “initial dip” in hemoglobin ical events. While a variety of intrinsic chromophores
oxygenation is of critical importance. As a mapping sig- exist in normal brain tissue, the majority of the signal in
nal, the early CMRO2 -related increase in Hbr appears the visible range is absorbed by hemoglobin. At higher
to have more precise spatial correlation with the popu- wavelengths, light scattering becomes a larger percent-
lation of active neurons than the later CBF-related in- age of the signal. Because of the different absorption rate
crease in HbO2 that forms a large part of the BOLD of Hbr and HbO2 (Fig. 11–1), one can use the intrinsic
mapping signal.5,7,9 Hence, fMRI studies may need to fo- signal to quantitate hemoglobin oxygenation. At isos-
cus more attention on the earlier inverted BOLD signal bestic wavelengths of hemoglobin where Hbr and HbO2
to increase its localization potential. In addition, using reflect light equally well (525 nm, 545 nm, 570.5 nm,
ORIS, the CBV signal has also been shown to provide and 583 nm; corrected for path length),6 ORIS measures
a high-resolution mapping signal, if imaged early in its total hemoglobin (Hbt), which is directly proportional

Hbt Hbr
HbO2
Hbr
Absorbance
increasing

450 500 550 600 650 700


Wavelength (nm)

Figure 11–1. The absorption curve of oxyhemoglobin (HbO2 ) and deoxyhemoglobin


(Hbr). Note that at the wavelength of 525 nm, 545 nm, 570.5 nm, and 583 nm,
(indicated with solid rectangle) Hbr and HbO2 absorb light equally and optical
recording of intrinsic signal (ORIS) signal mainly shows changes in total hemoglobin
concentration. At wavelength 600–650 nm, the ORIS signal mainly shows changes in
Hbr concentration.
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 133

to CBV and CBF assuming that the concentration of Illumination


red blood cells remains constant.26,29 At higher wave-
lengths (600–650 nm), the majority of the signal arises
from the oxygenation state of hemoglobin, since Hbr
absorbs light with three times the absorption coefficient
Detector 1 3
of HbO2 .7,30 Hence, a decrease in light reflection indi-
cates an increase in Hbr. When intrinsic signals of mul-
tiple wavelengths are recorded simultaneously from the
same preparation, spectrum analysis can be performed
with the Beer-Lambert law to quantify relative changes
in Hbr, HbO2 , and Hbt. Because the reflectance changes
are small, usually approximately 0.1% of the illumina-
tion, they are not directly visible. In order to visualize
activated brain regions, images must be acquired when Signal
the brain is active and inactive for a relative compar- source
2 4
ison. The most straightforward way is to subtract im-
ages recorded during nonactive conditions from those
recorded during active conditions. Other methods, based Figure 11–2. Illustration of the path of light traveling
on frequency-dependent stimulus presentation and anal- through a scattering media. When nearly infrared light
ysis, have also been successful.31 Although ORIS only was shined into a scattering media, the light will be
records functional changes occurring near the cortical absorbed and scattered by the media. Each detector
will receive light traveling through a banana-shaped
surface in the supragranular levels, due to the short pen-
pathway. Detectors at different locations will receive
etration distance of visible light, ORIS offers a superior light from different pathways. When a signal source is
combination of spatial sampling (as wide a field as can located in the pathway, the light change will be
be imaged), spatial resolution (∼200 ␮m) and temporal detected by the relative detector. For example, the
resolution (∼100 milliseconds, based on the relatively signal source on the right could be detected by
slow temporal evolution of the signal, although faster Detector 4 and the signal source on the left can be
frame rates can be acquired). detected by Detector 2.

its spatial origin. DOT images contain three-dimensional


NEAR INFRARED SPECTROSCOPY information about the origin of Hbt, Hbr, and HbO2 aris-
AND DIFFUSE OPTICAL ing from a large volume of tissue (or the whole brain)
TOMOGRAPHY with high temporal resolution.32−38 Compared to other
optical imaging modalities, DOT has a number of promi-
Visible light penetrates tissue only short distances, thus nent advantages such as portability, long recording time,
little information is available about the activity in deep and an insensitivity to mechanical movement. Both tech-
tissue. In the NIR spectrum (700–1100 nm), however, niques work better when less skin and skull lies between
photons can penetrate deeper into tissue (∼3 cm), even the light emission source and the detector. For this rea-
through bone. Within this spectral region, light absorp- son, studies in infants are common.
tion is also sensitive to the oxygenation of hemoglobin
(Fig. 11–1). While Hbr has a peak absorption at 760 nm,
HbO2 has a peak absorption at 920 nm. The cortex is 䉴 IMAGING NORMAL
generally illuminated via fiberoptic bundles directed ver- ARCHITECTURE IN ANIMALS
tically at the cortical surface. Since photons do not travel AND HUMANS
in a straight line through tissue, they will be reflected and
scattered. Each photon entering the tissue will follow a The application of optical imaging to the study of
different path with only a small fraction reaching each normal brain architecture has lead to several major
detector simultaneously. These paths are generally mod- breakthroughs in our understanding of the functional
eled as banana-shaped (Fig. 11–2). The recorded signal architecture based on its ability to sample large areas
will only contain information within the banana-shaped of the cortex simultaneously. Since the intrinsic signal
region, which results in a lower spatial resolution from represents hemodynamic surrogates of neuronal activity,
which only two-dimensional topographic imaging can areas of activation are more widespread than the pop-
be generated. ulation of cells firing action potentials, corresponding
DOT uses the same illumination wavelengths as more closely with the area of subthreshold activation.
NIRS to monitor cerebral hemodynamics and oxygena- Hence, mapping functional architecture often requires a
tion. However, DOT techniques process the scattered detailed understanding of the variety of behaviors that
light in more detail to obtain spatial information about in combination will activate the entire cortex, in order to
134 SECTION I TECHNIQUES

perform the appropriate subtractions that will individu- and guinea pig auditory cortex and Hess and Scheich70
ally identify the cortical region associated with a partic- analyzed frequency- and intensity- dependent spatial-
ular behavior. For this reason, maps of primary cortex temporal activity in primary auditory cortex AE of awake
are more robust than maps of association cortex since Mongolian gerbils. Later on, Harel et al.71 defined three
the range of behaviors that activate association cortex separate auditory area, AI, AII, and AAF in chinchilla.
are not as clearly understood (i.e., all five fingers can be They showed a cochleotopic or tonotopic organization
stimulated individually to map the hand sensory area but based on the differences in intrinsic signal areas evoked
generating maps of every possible phonemic, semiotic, by pure tones at octave-spaced frequencies from 500 Hz
and grammatical unit in a particular language would not to 16 KHz. The maps in AI and AII are arranged orthog-
be possible). onal to each other. In a recent study, Mrsic-Flogel et al.72
use ORIS to study the maturation of functional maps in
primary auditory cortex (A1) after the onset of sensory
VISUAL CORTEX
experience and they found that neither the tonotopic or-
ganization nor the representation of inputs from each ear
ORIS was used initially to study the functional archi-
reach maturity until approximately 1 month after hearing
tecture of visual cortex, which lead to major discov-
onset.
eries such as the pinwheel organization of orientation
columns and relative locations of the ocular dominance
columns and spatiotemporal frequency domains within SOMATOSENSORY CORTEX
area 17 and 18 (Fig. 11–3).5,39−48 Subsequently, ORIS was
ORIS has been used to map the functional organization
used to map direction of motion preference,49,50 color
of the whisker barrel cortex in rodents39,73−76 as well as
selective neuronal clusters in V1,51 distribution of hue
forepaw and hindpaw representation in somatosensory
maps in macaque striate cortex (V1), thin cytochrome
cortex of rodents and cats.6,77−81 A recent study using
oxidase stripes of V252−54 as well as form processing
voltage-sensitive dye (VSD) and ORIS in the same animal
modules in V455 and complex object representation in
showed good correlation between the centers of the two
infratemporal cortex.56 With chronically implanted cham-
maps, with differences in spatial extent82 ).
bers and artificial dura, ORIS can be applied to study
The somatotopic hand representation in macaque
the stability of visual architecture in awake behaving
and squirrel monkey primary somatosensory cortex
monkeys57−59 and the role of experience in the devel-
(area S-I) has also been studied with ORIS.83,84 The ac-
opment of the columnar organization of the cortex.60,62
tivation evoked by stimulation of a single finger was
While ORIS requires a craniotomy and exposed cor-
strongest in a narrow transverse band across the postcen-
tex, NIRS and DOT can be performed though the in-
tral gyrus and a sequential organization of these bands
tact skull making studies of visual processing in humans
was observed (Fig. 11–4). In addition to finger-specific
more practical. Kato et al.63 first use NIRS to monitor hu-
bands, activation areas common to all animals were
man visual cortical function during photic stimulation.
identified as well as specific areas sensitive to flutter,
Both increases in CBV and Hbr were recorded. Soon
pressure, and vibratory stimuli.83,84
after, Meek et al.,64 Sakatani et al.,65 Takahashi et al.,66
In humans, Toga et al.85 analyzed the spatiotem-
and Wilcox et al.67 also showed that NIRS can record
poral evolution of hemodynamic changes in sensorimo-
hemodynamic changes from the visual cortex in adults
tor cortex in response to peripheral somesthetic stimu-
and in infants.
lation. The somatosensory cortex of seven anesthetized
DOT imaging has also been used for study of
patients was mapped in response to transcutaneous elec-
visual processing and can provide additional three-
trical median and ulnar nerve stimulation using optical
dimensional spatial information about the activated
reflectance imaging. The ORIS maps colocalized with
cortical regions. Taga et al.68 used DOT to image the
the largest evoked potentials in both motor and sen-
hemodynamic responses in occipital and frontal cor-
sory regions. Cannestra et al.86 mapped the cortical rep-
tex of newborn infants during visual stimulation. They
resentation of different fingers with ORIS. They found
found that visual stimulation induced statistically signifi-
that the peak ORIS response to the stimulation of dif-
cant increases in oxyhemoglobin not only in the occipital
ferent fingers did not overlap, but the nonpeak optical
cortex but also in the prefrontal cortex. These results
signals were more widespread with significant overlap.
suggest that the neurovascular coupling is already func-
Co-localization between peak ORIS responses and sites
tioning in the newborn’s brain.
where direct cortical stimulation induced finger move-
ment was also observed. Sato et al.87 used intraop-
AUDITORY CORTEX erative ORIS to study the representation of different
fingers in somatosensory cortex. Biphasic optical
In 1996, ORIS was first employed to study the functional changes were recorded with ORIS during the stimula-
architecture of auditory cortex. Bakin et al.69 investi- tion of the first and fifth digits. Near the central sulcus,
gated the suprathreshold tonotopic organization of rat the cortical response area to the first and fifth digit was
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 135

M L

B C

D E

F G

Figure 11–3. Optical imaging of intrinsic signals reveals the functional architecture in
ferret visual cortex. (A) Blood vessel pattern of the surface of the visual cortex.
(B) Differential map produced by dividing images obtained with 0◦ and 90◦ stimulation.
(C) Differential map produced by dividing images obtained with 45◦ and 135◦
stimulation. The size of the 45◦ and 135◦ iso-orientation domains is smaller than that of
the 0◦ and 90◦ domains. (D) The angle map is generated by color-coded vectorial
summation of each single condition map on a pixel-by-pixel basis. (E) Fracture map
generated from the two-dimensional derivative of the angle map demonstrates rapid
changes in orientation preference at pinwheel centers. (F) Ocular dominance map in
the ferret has a seemingly disorganized structure and column diameters vary widely
both between and within animals. The V1/V2 border correlates with the margin
between the caudal contralateral and rostral ipsilateral eye bands. (G) Spatial
frequency maps demonstrate high spatial frequency preference caudally and
low-spatial frequency preference rostrally. The spatial frequency border is roughly
parallel to the ocular dominance border but shifted caudally. R, rostral; C, caudal; L,
lateral; M, medial. Scale bar = 1 mm. From, Schwartz TH: Optical imaging of
epileptiform events in visual cortex in response to patterned photic stimulation. Cereb
Cortex 2003;13:1287-1298.

spatially distinct, whereas in the postcentral sulcus, the observed. Painful stimulation induced a significant de-
two fingers were represented in the same region. Their crease in oxygen saturation ( p < 0.0001), whereas tactile
results suggested a hierarchical organization of the pri- stimulation produced no changes in oxygen saturation.
mary somatosensory cortex. Both tactile and painful stimulation induced an increase
NIRS has also been used to map the representation in HbO2 bilaterally regardless of which hand was stimu-
of both painful and tactile stimulation in somatosensory lated. Pain-induced HbO2 increases in the contralateral
cortex in the preterm newborn brain.88 Different hemo- somatosensory cortex ( p < 0.05) were not mirrored in
dynamic responses in the somatosensory cortex were the occipital cortex ( p > 0.1).
136 SECTION I TECHNIQUES

som
atos
ens
ory
motor

5 mm

5 mm

Figure 11–4. Somatosensory organization of hand area in monkey. (A) Time course of
optical response to mechanical stimulation of a single finger. The first figure is the
image of the cortical surface (left hemisphere, monkey M3). The somatosensory and
motor areas are marked. The anterior part of the somatosensory strip is Brodmann’s
area 1, and the posterior part is area 2. An image series of the left hemisphere in
monkey M3 showing the temporal development of the optical response to a weak
mechanical indentation stimulation of finger 3 of the right hand. Each frame
represents 500 milliseconds of summation of collected video frames. The finger was
stimulated at the distal phalanx using pneumatic air pressure pulses (10 milliseconds)
delivered for 2 seconds at a rate of 10 Hz. The stimulus started concurrently with the
acquisition of the second frame (stimulus duration is marked by a black horizontal
bar). The full gray scale corresponds to a fractional change of 1 × 10−3 . The images
from 45 repetitions of the stimulus (and blank) presentations were averaged. (B)
Somatotopic organization of the hand area in monkey M5. Left hemisphere finger
maps for right-hand stimulation are summarized here in two formats. Left, The
images show contour maps from the three monkeys. Contour lines at the level of 30%
of peak activation for each finger are superimposed on the surface vasculature image
(imaged with green light). The thin contour lines were computed from a smoothed
version of the maps (low-pass filtered with a Gaussian filter, = 120 ␮). The contour for
each finger is colored according to the color code shown in the hand drawing below
the maps. To facilitate a comparison with the second type of analysis shown on the
right, the WTA patches are also superimposed. Right, The images show WTA maps
calculated from the same data. The information from the individual finger maps is
integrated here using a winner-takes-all rule. The color of each pixel is determined by
the finger that gave the strongest response, using the same color codes used for the
contours. The intensity encodes the amplitude of the response to the “winning” finger.
Only pixels in which this response was >30% of the peak activation were colored. The
other pixels show the underlying vascular pattern.
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 137

GLM
d d
coefficient

c r

coronal sagittal

GLM
d
d coefficient

c r

coronal sagitt

Figure 11–5. Diffuse optical tomography imaging of functional brain activity during
front paw stimulation. The hemodynamic change maps (color ) were overlaid onto
anatomical cross-sections of the rat brain. Top: HbO2 , bottom: deoxyhemoglobin
(Hbr). v, ventral; c, caudal; r, rostral; GLM, generalized linear mode.

DOT imaging has also been successfully used in performance can induce distinct spatial and temporal
the imaging of functional brain responses to paw stimu- response patterns in Broca’s and Wernicke’s area (Fig.
lation in rats.36 The spatial distribution and quantitative 11–6). Pouratian et al.91 also reported that cortical lan-
changes in different hemodynamic components could be guage representation in bilinguals may consist of both
clearly mapped in both superficially, in the somatosen- overlapping and distinct areas.
sory cortex and at a depth, in the thalamus (Fig. 11–5). NIRS and DOT imaging have also been used to
study the prefrontal hypo-oxygenation during language
processing and the cortical representation of sound
LANGUAGE CORTEX and language through an intact skull. Fallgatter et al.92
found that during language processing, Hbr increases
ORIS was also applied in humans to detect cortical lan- and HbO2 decreases were significant but no hemispheric
guage areas in awake patients undergoing neurosur- differences were found. Minagawa-Kawai et al.93 used
gical procedures. Haglund et al.89 first demonstrated NIRS to assess the cerebral representation of Japanese
cognitively evoked activity in a language area. Significant short- and long-vowel categories. Their results showed
activation was seen in essential language areas that cor- that NIRS could capture phoneme-specific information
related with areas where stimulation mapping induced and confirmed the left dominance in phoneme process-
errors. Intrinsic signal changes were more widespread, ing. Taga and Asakawa94 used DOT to localize the corti-
indication that the area of cortex participating in lan- cal response to auditory and visual stimulation in awake
guage processing was larger than the area essential for infants aged 2 to 4 months. They found that auditory
language. Cannestra et al. 200090 also used ORIS to stimulation could induce focal increases in HbO2 and
study the cortical response to different language tasks decreases in Hbr in both hemispheres, but visual stim-
in awake patients. They found that different tasks and ulation did not induce significant change is HbO2 and
Figure 11–6. Optical recording of intrinsic signal (ORIS) response over Wernicke’s
area demonstrates that topographic specificity depends on the language task. ORIS
was performed over the temporal/parietal junction during an awake neurosurgical
procedure. Images were acquired in a block paradigm scheme (10 sec rest, 20 sec
activation), and averaged across blocks to obtain reflectance changes. Overall,
regions 3 and 5 were activated by all paradigms (region WAc in lower right
schematic). When comparing object naming (upper left) and word discrimination
(upper right), object naming selectively activated anterior and inferior cortices (region
WAa in lower right schematic) while word discrimination activated more superior and
anterior anatomy (superior to ESM site 3; region WAs in lower right schematic).
Auditory responsive naming (lower right) tasks activated all of the regions
corresponding to both the object naming and the word discrimination paradigms, and
was more intense centrally (in WAc). In addition, the auditory responsive naming
discrimination paradigm also selectively activated more posterior cortex (near ESM
site 4; WAp in lower right schematic). The magnitude contours demarcate these
differences. Baseline activity is observed in the lower left. ESM mapping revealed
anomia for cortical regions 3, 4, 5, and 6. ESM mapping at site 1 (at upper left cranial
margin) reveal hand and arm movement, thereby localizing motor cortex. In this
patient (patient 8), angular gyrus and superior temporal gyrus were noted to be
shifted anterior and inferior due to the presence of an arteriovenous malformation
superior and deep to angular gyrus. Magnitude contours (in yellow ) indicate
increasing intensity. Magnitude contours were obtained after applying a 5-pixel
Gaussian blur. Images are averages of eight scans in one patient. A, anterior;
S, superior; CS, central sulcus; SF, superior frontal. The color bar is reflectance
change 10−4 for all paradigms. Scale bar: 1 cm.

138
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 139

Hbr, indicating staggered development of neurovascular with chronically implanted grids of electrodes. How-
coupling mechanisms. ever, these methods have limitations in their ability to
identify ictal onset zones, areas of propagation, and in-
terictal spikes because of low spatial sampling. ORIS
SUMMARY has been employed to study epilepsy in various ani-
mal models98−102 as well as in humans in the operating
Overall, optical imaging techniques have had a dramatic room.89,103
impact in our understanding of the topographic rep- Schwartz and Bonhoeffer101 showed in an animal
resentation of function in primary cortex such as vi- model that ORIS is an excellent method for in vivo map-
sual, auditory, and somatosensory cortex. This work has ping of clinically relevant epileptiform events, such as
been most clear in animal models in the laboratory. Hu- interictal spikes, ictal onsets, ictal propagation, and sec-
man studies of primary cortex are feasible but to date ondary homotopic foci. Using optical spectroscopy, Suh
have been mostly derivative of the laboratory studies et al.20 and Bahar et al.23 have shown that interictal
and lack the spatial resolution of the laboratory stud- and ictal events cause a rapid focal increase in Hbt as
ies due to large sources of noise, primarily from brain well as an increase in Hbr (Fig. 11–7). The increase in
pulsation and vasomotor fluctuations. Imaging the initial Hbr, or “epileptic dip” lasts longer than the initial dip
dip, which would increase the spatial resolution, is chal- reported after normal sensory processing and implies
lenging compared with the less spatially restricted BOLD that for a period of time, the reactive increase in CBF
signal (i.e., hyperperfusion and hyperoxygenation over- is not able to compensate for the metabolic demands
shoot). The operating room environment is also not con- of both ictal and interictal events. However, both the
ducive to these sensitive optical experiments based on Hbt and Hbr signals are useful for mapping either in-
time constraints. However, great promise exists for future terictal or ictal events. The later decrease in Hbr, which
work. In association cortex, the sources of the intrinsic comprises much of the BOLD signal demonstrated with
signal changes and the correlations between the signals fMRI, however, was much less focal and spread to larger
and behavior is less well-understood but still a fertile areas of cortex. Comparing the Hbr/Hbt maps with VSD
ground for future research. Chronic experiments with maps reveals that although there was a linear relation-
implanted optical arrays or skull mounted DOT helmets ship between the amplitude of the hemodynamic and
may hold scientific promise for future breakthroughs. voltage signals, both Hbr and Hbt spatially overshoot
the extent of the excitatory VSD signal by approximately
2×.24 Overall, ORIS has been shown in animal models
to be an excellent tool for mapping epilepsy, and as
䉴 IMAGING PATHOLOGY IN shown by Schwartz et al.19,104 can be used to simulta-
ANIMALS AND HUMANS neously derive maps of both epileptic and functional
architecture.
Many brain diseases, such as epilepsy, stroke, intracra- Experience with optical mapping of epilepsy in hu-
nial hemorrhage, and trauma, can significantly influence mans is limited since seizures do not routinely occur
CBF and hemoglobin oxygenation. Optical spectro- during surgery and cannot be reliably induced. Electri-
scopic imaging can be a powerful tool for identifying cally triggered afterdischarges have been imaged in hu-
and understanding these pathophysiological processes. man patients during craniotomies.89,105 Haglund et al.89
reported that the optical changes increased in magni-
tude as the intensity and duration of the afterdischarges
EPILEPSY increased. Spontaneous seizures have been recorded in
two reports. Haglund et al.106 localized a seizure using
Epilepsy is a clinical term referring to a disease which ef- the Hbt signal and Zhao et al.103 used both Hbr and Hbt
fects between 1% and 2% of the population of the United signals. In the latter publication, the authors confirmed
States involving recurrent seizures.95 Seizures consist of the presence of the epileptic dip in unanesthetized hu-
the paroxysmal, synchronous, rhythmic firing of a popu- man cortex during spontaneous seizures and also re-
lation of pathologically interconnected neurons capable ported focal changes in light reflection, which preceded
of demonstrating high-frequency oscillatory activity.96 the onset of seizures by approximately 20 seconds (Fig.
These events are caused by an imbalance in excitatory 11–8). This finding mirrors similar data reported in the
and inhibitory mechanisms leading to both hypersyn- rat107 and may offer new possibilities not only for seizure
chrony and hyperexcitability.97 localization but prediction.
Most studies in epilepsy have been done using elec- Another method of optical imaging, NIRS, is cur-
trophysiological recording from surface field and ex- rently being explored in experimental trials in the
tracellular single-unit electrodes. The current method clinic as a method for presurgical seizure localization
of identifying epileptogenic cortex in humans is also and is attractive because of its noninvasiveness. The
140 SECTION I TECHNIQUES

1 mV

50 ms

Hbr
0.2%
0.1%
0 ms 408.8 817.6 1226.4 1635.2 2044 2452.8
0

Hbt

0.1%
0 ms 408.8 817.6 1226.4 1635.2 2044 2452.8 0
A
–0.1%
4 mm

1 mv

10 s

Hbr

0.6
−1.4 s 0 0.8 1.2 2.0 5.0 8.1
0
–0.6
20.1 30.0 40.1 50.2 60.1 70.2 80.1

Hbt

0.6
−1.4 s 0 0.8 1.2 2.0 5.0 8.1
0

20.1 30.0 40.1 50.2 60.1 70.2 80.1 –0.6


B
4 mm

Figure 11–7. Optical recording of intrinsic signal of interictal and ictal events. (A) Time
course of optical response to interictal events. The top trace is the average signal
recorded from the local field potential of multiple interictal spikes. Below is the
evolution and propagation of the deoxyhemoglobin (Hbr) and total hemoglobin (Hbt)
signals. (B) Ictal events last for a longer period of time. Field potential above indicates
the duration of the seizure corresponding with the development of the Hbr and Hbt
signals shown in the figure.

first NIRS study on epileptic patients was performed used to monitor the hemodynamic responses to
by Villringer and his colleague.108 They used NIRS epilepsy,109−111 render a diagnosis of epilepsy,112 distin-
to continuously monitor hemodynamic changes during guish seizure types,113 and in the management of status
spontaneous complex-partial seizures. They found that epilepticus in infants.114 Although the spatial resolution
extremely large increases in CBV and HbO2 concentra- of NIRS is low, technical advances may overcome this
tion were measured during seizure, which were larger limitation. As a presurgical tool NIRS cannot yet replace
than during other cognitive functions such as calcu- the EEG but may provide complementary information to
lations and visual processing. NIRS was subsequently aid in the decision-making process.
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 141

LFP (mV)
2

0
0.1
–2
−40 −20 0 20 40 0.05

0
0.06
-0.05

0.03 -0.1

-0.03
A −40 −20 0 20 40

2
LFP (mV)

–2
−40 −20 0 20 40
0.2

0.1
0.2
0.15 0

0.1 -0.1

0.05 -0.2
0
-0.05
−40 −20 0 20 40
B

Figure 11–8. Hemodynamic change during spontaneous seizure in a human. The


electrocorticographic recording and optical signal (A, 610 nm, B, 570 nm) 40 seconds
before and after the onset of the seizure are presented above. Optical signals from a
linear region of interest (60 × 6 pixels (height × width); 2.3 × 0.23 cm) over the crest
of the gyrus from which the seizures arise are presented below. Six pixels in width
were averaged and each pixel was divided from the baseline, taken from an average
of 150 frames (4.5 sec) at the onset of each trial. The pixel changes in comparison to
the baseline pixels were color coded (see color bar: the increase in red and the
decrease in blue) and plotted as a function of time. The image on the right shows the
optical recording of intrinsic signal maps for each of the two seizures at the time of
maximum increase. Each image is divided by the baseline image. Scale bar: 1 cm

STROKE tical imaging techniques can provide real-time measure-


ments of perfusion and oxygenation in large areas of the
Stroke is the clinical designation for a rapidly devel- cortex.
oping loss of brain function due to an interruption in In an animal model, Wolf et al.115 first used NIRS in
the blood supply to all or parts of the brain. This phe- the monitoring of regional cerebral blood oxygenation
nomenon can be caused by thrombosis, embolism, or (CBO) during peri-infarct depolarization in a model of
hemorrhage. Traditional functional brain imaging meth- focal cerebral ischemia in the rat. Their work proved
ods such as fMRI, PET, and SPECT have provided insight that NIRS is a powerful tool to detect typical peri-infarct
into the functional changes associated with stroke. How- depolarization-associated changes in blood oxygena-
ever, these techniques have some common drawbacks tion. Chen et al.,116 further confirmed that in ischemic rat
such as the immobilization of the subject’s head and brain, the geometric shape and infarct area measured by
lack of real-time or long-term monitoring capability. Op- NIRS matched well with those measured with fMRI and
142 SECTION I TECHNIQUES

A B C D E

Figure 11–9. Orientation/Retinotopic map reorganization after an ischemic lesion.


(A) Blood vessel pattern. (B) Iso-orientation maps in response to gratings of 0◦ (C)
Retinotopic map in response to a 1◦ wide stimulus oriented at 0◦ . (D) Retinotopic map
in response to 1◦ wide stimulus oriented at 0◦ , adjacent to the stimulus presented in C.
(E) Overlap between responses in C and D: light gray domains correspond to active
zones in C, dark gray domains correspond to active zones in D, and black zones
correspond to active areas in both C and D, the “overlap.” Note that the overlapping
area fully recovers after 5wPL. Orientation of the imaged cortical area is shown. A,
anterior; P, posterior; M, medial; L, lateral.

postmortem histology. NIRS can monitor the hemody- reported. Kato et al.120 found that chronic stroke patients
namic change in real-time and may be a practical tool for exhibit a similar CBO response as do normal subjects
identifying the onset of stroke in high-risk patients. ORIS in the sensorimotor cortex during hand movement. In
has also been applied to study the effects of ischemic contrast, Sakatani et al.121 reported an atypical evoked
events on functional cortical maps. Zepeda et al. (2003)117 CBO change the left prefrontal cortex of a group of
induced a focal photochemical lesion in V1 of kittens stroke patients during language tasks. Murata et al.122
and analyzed the subsequent reorganization of the cor- also observed similar atypical evoked CBO changes in
tical maps using ORIS. They found that the retinotopic the primary sensorimotor cortex during contralateral mo-
and orientation preference maps reorganize around the tor tasks. A further study by Murata et al.123 on patients
lesion over a period of 5 weeks correlating with dendritic with a variety of cerebral circulatory conditions showed
spouting and biochemical changes (Fig. 11–9).118 that the affect of cerebral ischemia on the evoked CBO
Several studies in humans have examined the utility response patterns varies depending on the level of is-
of NIRS to monitor various aspects of a stroke in evolu- chemia. That is, moderate cerebral ischemia caused no
tion, such as the hemodynamic response patterns, and changes in the evoked CBO response pattern, whereas
recovery or rehabilitation after stroke. Benaron et al.119 severe cerebral ischemia caused an increase in Hbr con-
used NIRS to study hemoglobin oxygenation in neonates centration.
after acute stroke, and areas of decreased oxygenation The first NIRS study of stroke rehabilitation was
were found to spatially overlap with the region of is- by Saitou et al.124 They investigated the CBV and cere-
chemia as measured by CT scan. In chronic stroke pa- bral oxygen volume (COV) in 44 hemiplegic patients
tients, however, conflicting data about CBO have been and 24 normal control subjects. Each subject performed
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 143

specific rehabilitation tasks, including head-up tilt, cal- brain injury (ABI). The gold standard imaging test for TBI
culation, an ergometer task, facilitation, stand-up task, diagnosis in the emergency room is a CT scan, which
and gait. They found both increases and decreases in creates a series of cross-sectional X-ray images of the
CBV and COV during different tasks in all the sub- head and brain and can show bone fractures as well
jects. Their work clearly showed that NIRS is a useful as the presence of subdural and intraventricular hem-
tool for monitoring changes in regional cerebral hemo- orrhages intraparenchymal hematomas, contusions, and
dynamics and oxygenation during rehabilitation. Miyai brain swelling. MRI may be used after the initial assess-
et al.125 also used NIRS to study the rehabilitation of ment and treatment of the TBI patient to identify diffuse
gait following stroke. Six nonambulatory patients with axonal injury and more subtle changes in extracellular
severe stroke were monitored during a treadmill test water concentration (edema). Recently, several studies
approximately 3 months after stroke. Each patient per- have used optical methods to monitor the progression
formed the tasks (0.2 km/hr) for 30 seconds followed of intracranial hemorrhages, subdural hematomas, brain
by a 30-second rest period. The researchers found in- swelling, and increased intracranial pressure (ICP) in TBI
creased activity in the premotor and presupplementary patients.
motor cortex in the affected hemisphere during rehabili- In 1995, Robertson et al.127 used NIRS to detect
tation training. They also found that a therapist-facilitated the development of delayed hematomas in 167 patients.
swing induced greater activation than did mechanical The NIRS signals at 760 nm were measured in the
assistance. Their finding suggested that multiple motor hemisphere with the hematoma as well as the con-
areas, including the premotor cortex and presupplemen- tralateral hemisphere for comparison during the first
tary motor area might play important roles in restora- 3 days postinjury. Twenty-seven of the patients devel-
tion of gait in patients with hemiplegia following severe oped some type of late hematoma, of which 18 patients
stroke. required surgical evacuation. A significant increase in the
Kato et al.31 performed a study of hand grasp on light absorption occurred in 24 of the 27 patients prior to
six chronic stroke patients with minimal hemiparesis and other detectable symptoms, such as a change in the neu-
five similar age-matched control subjects. Both NIRS and rological examination or CT scan. Their work suggested
fMRI were used to evaluate the compensatory motor ac- that NIRS may allow early identification of progressive
tivation of cortical regions in patients who recovered hematomas with earlier institution of therapy.
from hemiparesis after cortical cerebral infarction. They NIRS has also been utilized to monitor brain phys-
found very similar data from NIRS and fMRI, although iology following TBI for prognostic purposes. Adelson
NIRS measurements seemed to be more sensitive than et al.128 performed a preliminary NIRS study in children
the fMRI. However, NIRS only measures superficial acti- after severe TBI. Changes in HbO2 , Hbr, and Hbt were
vation, whereas fMRI can measure from the entire vol- compared to, ICP mean arterial pressure and arterial
ume of the brain. PCO2 . They found that decreases in arterial PCO2 in-
Miyai et al.126 used NIRS in longitudinal studies of duced desaturation of cerebral oxygen regardless of ICP
recovery after stroke (Fig. 11–10). A total of eight pa- and high ICP correlated with increased Hbt and HbO2 .
tients with stroke were investigated during hemiparetic Kampfl et al.129 also used NIRS to measure changes in
gait training on the treadmill. They found that ambulat- regional cerebral oxygen saturation in TBI patients. They
ing was associated with increased levels of HbO2 in the found that NIRS measurements were able to differentiate
medial primary sensorimotor cortex, premotor cortex, high versus low ICP in spite of similar levels of arterial
and supplementary motor area, and that the activity was partial pressure of oxygen (pO2 ), partial pressure of car-
greater in the unaffected hemisphere than in the affected bon dioxide (pCO2 ), and peripheral oxygen saturation.
hemisphere. On the second examination, the asymmetry Regional cerebral oxygen saturation in the higher ICP
in sensorimotor activation was significantly diminished, group was significantly lower than those of the lower
and there was enhanced premotor cortex activation in ICP group. In addition, the low ICP group revealed a sig-
the affected hemispheres. This improvement in asym- nificant increase in regional cerebral oxygen saturation
metry significantly correlated with an improvement in following induced hyperoxygenation, which was not de-
gait. tectable in the high ICP group. Their results suggested
that NIRS can distinguish between patients with high
and low ICP. Dunham et al.130 also reported a correla-
TRAUMATIC BRAIN INJURY AND tion between cerebral oximetry and cerebral perfusion.
INTRACRANIAL HEMATOMAS In a small study of four patients, an increase in cere-
bral perfusion pressure was significantly correlated with
Traumatic brain injury (TBI) is a term used to describe an increase in cerebral oxygen saturation ( p < 0.0001).
the damage to the brain that results from head trauma. Their results suggested that the cerebral oxygen satura-
TBI can result from a closed head injury or a penetrating tion, measured with NIRS, could be a sensitive indica-
head injury and is also considered a subset of acquired tor to monitor the development of increased ICP due to
144 SECTION I TECHNIQUES

A B C D

Figure 11–10. Cortical mapping of hemiparetic gait in patients with stroke. Cortical
activation maps are based on changes in HbO2 levels during gait before (pre) and
after (post) inpatient rehabilitation. Images in bottom row show site of lesions on
T2-weighted magnetic resonance imaging. “L” indicates left. (A) Cortical mapping of
gait in Case 2, with infarction in the left corona radiata (arrow ). On day 53 after stroke,
the patient needed moderate assistance to take a step. SMC activation was much
less in affected hemisphere than in unaffected hemisphere. After inpatient
rehabilitation (118 days after stroke), the patient needed minimal assistance to
perform the task. Sensorimotor cortex (SMC) activation was symmetrical, and new
activation was seen in supplementary motor area (SMA) and premotor cortex (PMC),
especially in affected hemisphere. (B) Cortical mapping of gait in Case 3, with
infarction in left corona radiata (arrow ). On day 107 after stroke, the patient needed
mild assistance with gait. There was less SMC activation in affected hemisphere than
in unaffected hemisphere, but PMC and SMA were bilaterally activated. On the
second imaging (176 days after stroke), the patient needed little assistance with gait.
SMC were symmetrically activated, and there was persistent activation in PMC and
SMA. (C) Cortical mapping of gait in Case 6, with diffuse infarction in right
frontoparietal lobe. On day 102 after stroke, the patient needed maximal assistance
with gait; this was the patient’s first opportunity to walk after the ictus. Mild activation
was observed in PMC and prefrontal regions in affected hemisphere, parietal regions
in unaffected hemisphere, and bilateral pre-SMA. There was little activation in the
medial SMC. On the second evaluation (173 days after stroke), the patient needed
mild assistance. Near infrared spectroscopy (NIRS) imaging revealed enhanced
activation in bilateral PMC, pre-SMA, and prefrontal areas and mild SMC activation in
unaffected hemisphere. (D) Cortical mapping of gait in Case 8, with a large
hemorrhagic lesion centered in right parietal lobe (arrow ). On day 32 after stroke, the
patient needed moderate assistance with gait. NIRS imaging showed prominent
activation in unaffected hemisphere. After inpatient rehabilitation (98 days after
stroke), the patient needed minimal assistance with gait, and enhanced activation
was observed in the bilateral SMC, PMC, SMA, and prefrontal cortices.

increases in cerebral perfusion. Other investigators, ments in ten patients with severe closed head injury
however, have not found NIRS to be useful for TBI. (Glasgow Coma Scale score ≤8). Poor correlation (r 2 =
In 1996, Lewis and his colleague131 compared contin- 0.04) between paired measurements and wide limits
uous jugular venous bulb oximetry and NIRS measure- of agreement (−13% to +21%) were demonstrated. In
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 145

fact, 14 clinically significant episodes of jugular venous over multiple trails is an effective way to reduce this
bulb desaturation were not detected by NIRS. Likewise, noise, it is only applicable to events which are repetitive
Büchner et al.132 further investigated the possibility of us- and reproducible.
ing NIRS to measure cerebral oxygenation after severe Other limitations and obstacles must be overcome.
TBI in a larger population (n = 31). The NIRS measure- NIRS and DOT are influenced by the absorption of light
ments were compared to several different standard inva- of hair and reliable contact between the patient’s head
sive monitoring methods, such as ICP, CPP, and regional and the optodes. The subdural implantation of an op-
brain tissue–pO2 monitoring. Significant changes were tical grid might be a resolution of these limitations, but
observed in all the patients with these invasive monitor- it is invasive. When optical components are subdurally
ing methods, however, NIRS measurements were only implanted, imaging can be performed chronically for ex-
possible in 80% (using the INVOS oximeter) and 46% tended periods of time. Continuous imaging might then
(using the CRITIKON monitor). Only induced hyperoxia be possible while patients are hospitalized. Since light
(fraction of inspired oxygen (FiO2 ) = 1.0) revealed a scattering and absorption from the skull and hair will be
significant correlation between all modalities (r = 0.67, eliminated, the spatial resolution and imaging depth will
p < 0.01), while lower or no correlation was found after be increased.
changing arterial carbon dioxide tension (paCO2 ) and
administering mannitol.

REFERENCES
䉴 CONCLUSION
1. Cohen LB. Changes in neuron structure during action po-
While more commonly used in animal studies to study tential propagation and synaptic transmission. Physiol Rev
the functioning of the normal brain, the application 1973;53:373-418.
of optical spectroscopic to imaging pathological brain 2. Roy C, Sherrington C. On the regulation of the blood sup-
states in humans may be more useful and have more im- ply to the brain. J Physiol Lond 1890;11:85-108.
mediate clinical application than imaging normal func- 3. Fox PT, Raichle ME, Mintun MA, Dence C. Nonoxidative
tional architecture. For example, optical spectroscopy glucose consumption during focal physiologic neural ac-
tivity. Science 1988;241:462-464.
offers a unique and effective way to measure the spa-
4. Ogawa S, Lee TM, Kay AR, Tank DW. Brain magnetic res-
tiotemporal dynamics of neurovascular coupling during onance imaging with contrast dependent on blood oxy-
epileptic discharges,105 and the signals associated with genation. Proc Natl Acad Sci USA 1990;87:9868-9872.
epilepsy are much larger than those associated with sen- 5. Frostig RD, Lieke EE, Ts’o DY, Grinvald A. Cortical func-
sory processing and are easier to record.While the ini- tional architecture and local coupling between neuronal
tial dip is difficult to image in humans in the operating activity and the microcirculation revealed by in vivo high-
room, the epileptic dip has been identified more read- resolution optical imaging of intrinsic signals. Proc Natl
ily. Hence, optical spectroscopy holds promise for Acad Sci 1990;87:6082-6086.
identifying and predicting seizure onsets in humans. 6. Sheth SA, Nemoto M, Guiuo M, Walker M, Pouratian N,
Likewise, while the spatial resolution of transcranial Toga AW. Evaluation of coupling between optical intrinsic
techniques such as NIRS and DOT may be too low signals and neuronal activity in rat somatosensory cortex.
Neuroimage 2003;19:884-894.
to map functional architecture, these techniques offer
7. Malonek D, Grinvald A. Interactions between electrical
a noninvasive method for identifying large areas of is- activity and cortical microcirculation revealed by imaging
chemia, or hematomas in evolution and may become a spectroscopy: implications for functional brain mapping.
standard of care in intensive care monitoring unit. Sim- Science 1996;272:551-554.
ilarly, optical epilepsy monitoring with NIRS may offer 8. Mayhew JEW, Zheng Y, Hou Y, et al. Spectroscopic anal-
complementary data to EEG monitoring and have suffi- ysis of changes in remited illumination: the response to
cient spatial resolution to identify a regional onset zone increased neural activity in brain. Neuroimage 1999;10:
in preparation for further invasive monitoring. 304-326.
As the technology and our understanding of neu- 9. Vanzetta I, Grinvald A. Increased cortical oxidative
rovascular coupling evolve, optical spectroscopic imag- metabolism due to sensory stimulation: implications for
ing (ORIS, NIRS, and DOT) will likely play an increasing functional brain imaging. Science 1999;286:1555-1558.
10. Logothetis NK, Gugenberger H, Peled S, Pauls J. Func-
role in the clinic. Noise reduction will be a critical com-
tional imaging of the monkey brain. Nature Neurosci
ponent of this future work. Large signals arise from 1999;2:555-562.
vascular artifact84 such as the periodic dilation and con- 11. Kim DS, Duong TQ, Kim SG. High-resolution mapping
striction of blood vessels (vasomotor noise)133 and the of iso-orientation columns by fMRI. Nature Neurosci
oscillation of cerebral oxygenation.134 The amplitude of 2000;3:164-169.
these periodic fluctuations is often similar to, or higher 12. Lindauer U, Royl G, Leithner C, et al. No evidence for early
than the activity-related changes. Although averaging decrease in blood oxygenation in rat whisker cortex in
146 SECTION I TECHNIQUES

response to functional activation. Neuroimage 2001;13: 28. Vanzetta I, Slovin H, Omer DB, Grinvald A. Columnar
988-1001. resolution of blood volume and oximetry functional maps
13. Vanzetta I, Grinvald A. Evidence and lack of evidence for in the behaving monkey: Implications for fMRI. Neuron
the initial dip in the anesthetized rat: implications for hu- 2004;42:843-854.
man functional brain imaging. Neuroimage 2001;13:959- 29. Mayhew JEW, Johnston D, Berwixk J, et al. Spectroscopic
967. analysis of neural activity in brain: increased oxygen con-
14. Sheth SA, Nemoto M, Guiou G, Walker M, Pouratian N, sumption following activation of barrel cortex. Neuroim-
Toga AW. Linear and nonlinear relationships between age 2000;12:664-675.
neuronal activity, oxygen metabolism, and hemodynamic 30. Sato C, Nemoto M, Tamura M. Reassessment of activity-
response. Neuron 2004;42:347-355. related optical signals in somatosensory cortex by an algo-
15. Ances BM, Buerk DG, Greenberg JH, Detre JA. Temporal rith with wavelength-dependent path length. Jpn J Physiol
dynamics of the partial pressure of brain tissue oxygen 2002;52:301-312.
during functional forepaw stimulation in rats. Neurosci 31. Kalatsky VA, Stryker MP. New paradigm for optical imag-
Lett 2001;306:106-110. ing: temporally encoded maps of intrinsic signal. Neuron
16. Ances BM. Coupling of changes in cerebral blood flow 2003;38:529-545.
with neural activity: what must initially dip must come 32. Obrig H, Villringer A. Beyond the visible–imaging the hu-
back up. J Cereb Blood Flow Metab 2003;24:1-6. man brain with light. J Cereb Blood Flow Metab 2003;23:
17. Thompson JK, Peterson MR, Freeman RD. Single-neuron 1-18.
activity and tissue oxygenation in the cerebral cortex. Sci- 33. Boas DA, Dale AM, Franceschini MA. Diffuse optical
ence 2003;299:1070-1072. imaging of brain activation: approaches to optimizing
18. Thompson JK, Peterson MR, Freeman RD. High-resolution image sensitivity, resolution, and accuracy. Neuroimage
neurometabolic coupling revealed by focal activation of 2004;23(Suppl 1):S275-288.
visual neurons. Nat Neurosci 2004;7:919-920. 34. Gibson AP, Hebden JC, Arridge SR. Recent advances in
19. Schwartz TH. The application of optical recording of in- diffuse optical imaging. Phys Med Biol 2005;50:R1-R43.
trinsic signal to simultaneously acquire functional, patho- 35. Kotilahti K, Nissila I, Huotilainen M, et al. Bilateral hemo-
logical and localizing information and its potential role dynamic responses to auditory stimulation in newborn
in neurosurgery. Stereotac Funct Neurosurg 2005;83: infants. Neuroreport 2005;16:1373-1377.
36-44. 36. Schmitz CH, Graber HL, Pei Y, et al. Dynamic studies of
20. Suh M, Bahar S, Mehta AD, Schwartz TH. Temporal de- small animals with a four-color DOT imager. Rev Scie In-
pendence in uncoupling of blood volume and oxygena- strum 2005;76:094302.
tion during interictal epileptiform events in rat neocortex. 37. Barbour RL, Pei Y, Schmitz CH, et al. Functional multi-
J Neuroscience 2005;25:68-77. mode imaging in freely moving rats. In Fifth Inter-Institute
21. Suh M, Shariff S, Bahar S, Mehta AD, Schwartz TH. Intrin- Workshop on Optical Diagnostic Imaging from Bench to
sic optical signal imaging of normal and abnormal phys- Bedside at the National Institutes of Health. Bethesda,
iology in animals and humans-seeing the invisible. Clin MD, 2006.
Neurosurg 2005;52:135-149. 38. Huppert TJ, Hoge RD, Diamond SG, Franceschini MA,
22. Bahar S, Suh M, Mehta AD, Schwartz TH. In vivo intrinsic Boas DA. A temporal comparison of BOLD, ASL, and
optical signal imaging of neocortical epilepsy. In Bioimag- NIRS hemodynamic responses to motor stimuli in adult
ing in Neurodegeneration, edited by PA Broderick DN and humans. Neuroimage 2006;29:368-382.
EH Rahni Kolodny eds. Totowa, NJ: Humana Press, 2006, 39. Grinvald A, Lieke E, Frostig RD, Gilbert CD, Wiesel TN.
pp 149-175. Functional architecture of cortex revealed by optical imag-
23. Bahar S, Suh M, Zhao M, Schwartz TH. Intrinsic optical ing of intrinsic signals. Nature 1986;324:361-364.
signal imaging of neocortical seizures: The “epileptic dip.” 40. Ts’o DY, Gilbert CD, Wiesel TN. Relationships between
Neuroreport 2006;17:499-503. horizontal interactions and functional architecture in cat
24. Shariff S, Suh M, Zhao M, Ma H, Schwartz TH. Recent striate cortex as revealed by cross-correlation analysis. J
developments in oximetry and perfusion-based mapping Neurosci 1986;6:1160-1170.
techniques and their role in the surgical treatment of neo- 41. Ts’o DY, Frostig RD, Lieke EE, Grinvald A. Functional or-
cortical epilepsy. Epilepsy Behav 2006;8:363-375. ganization of primate visual cortex revealed by high res-
25. Suh M, Ma H, Zhao M, Shariff S, Schwartz TH. Neu- olution optical imaging. Science 1990;249:417-420.
rovascular coupling and oximetry during epileptic events. 42. Grinvald A, Frostig RD, Siegel RM, Bartfeld E. High-
Molec Neurobio 2006;33:1-17. resolution optical imaging of functional brain architec-
26. Nemoto M, Sheth S, Guiou M, Pouratian N, Chen JWY, ture in the awake monkey. Proc Natl Acad Sci U S A
Toga AW. Functional signal- and paradigm-dependent lin- 1991;88:11559-11563.
ear relationships between synaptic activity and hemody- 43. Bonhoeffer T, Kim DS, Malonek D, Shoham D, Grinvald
namic responses in rat somatosensory cortex. J Neurosci A. Optical imaging of the layout of functional domains
2004;24:3850-3861. in area 17 and across the area 17/18 border in cat visual
27. Sheth SA, Nemoto M, Guiou G, et al. Columnar Specificity cortex. Eur J Neurosci 1995;7:1973-1988.
of microvascular oxygenation and volume responses: 44. Hübener M, Shoham D, Grinvald A, Bonhoeffer T. Spatial
Implications for functional brain mapping. J Neurosci relationships among three columnar systems in cat area
2004;24:634-641. 17. J Neurosci 1997;17:9270-9284.
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 147

45. Shoham D, Hubener M, Schulze S, Grinvald A, Bonhoeffer means of near-infrared spectroscopy. J Cereb Blood Flow
T. Spatio-temporal frequency domains and their relation Metab 1993;13:516-520.
to cytochrome oxidase staining in cat visual cortex. Nature 64. Meek JH, Elwell CE, Khan MJ, et al. Regional changes in
1997;385:529-533. cerebral haemodynamics as a result of a visual stimulus
46. Shoham D, Hübener M, Schulze S, Grinvald A, Bonhoeffer measured by near infrared spectroscopy. Proc Biol Sci
T. Spatio-temporal frequency domains and their relation 1995;261:351-356.
to cytochrome oxidase staining in cat visual cortex. Nature 65. Sakatani K, Chen S, Lichty W, Zuo H, Wang YP. Cerebral
1997;385:529-533. blood oxygenation changes induced by auditory stimula-
47. Issa NP, Trepel C, Stryker MP.Spatial frequency maps in tion in newborn infants measured by near infrared spec-
cat visual cortex. J Neurosci 2000;20:8504-8514. troscopy. Early Hum Dev 1999;55:229-236.
48. Schwartz TH. Optical imaging of epileptiform events in 66. Takahashi K, Ogata S, Atsumi Y, et al. Activation of the
visual cortex in response to patterned photic stimulation. visual cortex imaged by 24-channel near-infrared spec-
Cer Cortex 2003;13:1287-1298. troscopy. J Biomed Opt 2000;5:93-96.
49. Shmuel A, Grinvald A. Functional organization for direc- 67. Wilcox T, Bortfeld H, Woods R, Wruck E, Boas DA. Us-
tion of motion and its relationship to orientation maps in ing near-infrared spectroscopy to assess neural activa-
cat area 18. J Neurosci 1996;16:6945-6964. tion during object processing in infants. J Biomed Opt
50. Weliky M, Bosking WH, Fitzpatrick D. A systematic map 2005;10:11010.
of direction preference in primary visual cortex. Nature 68. Taga G, Asakawa K, Maki A, Konishi Y, Koizumi H.
1996;379:725-728. Brain imaging in awake infants by near-infrared optical
51. Landisman CE, Ts’o DY. Color processing in macaque stri- topography. Proc Natl Acad Sci U S A 2003;100:10722-
ate cortex: relationships to ocular dominance, cytochrome 10727.
oxidase, and orientation. J Neurophysiol 2002;87:3126- 69. Bakin JS, Kwon MC, Masino SA, Weinberger NM, Frostig
3137. RD. Suprathreshold auditory cortex activation visual-
52. Xiao Y, Wang Y, Felleman DJ. A spatially organized rep- ized by intrinsic signal optical imaging. Cereb Cortex
resentation of colour in macaque cortical area V2. Nature 1996;6:120-130.
2003;421:535-539. 70. Hess A, Scheich H. Optical and FDG mapping of
53. Wang Y, Xiao Y, Felleman DJ. V2 thin stripes contain spa- frequency-specific activity in auditory cortex. Neuroreport
tially organized representations of achromatic luminance 1996;7:2643-2647.
change. Cereb Cortex 2007;17:116-129. 71. Harel N, Mori N, Sawada S, Mount RJ, Harrison RV. Three
54. Xiao Y, Casti A, Xiao J, Kaplan E. Hue maps in primate distinct auditory areas of cortex (AI, AII, and AAF) de-
striate cortex. Neuroimage 2007;35:771-786. fined by optical imaging of intrinsic signals. Neuroimage
55. Ghose GM, Ts’o DY. Form procesing modules in primate 2000;11:302-312.
area V4. J Neurophsyiol 1997;77:2191-2196. 72. Mrsic-Flogel TD, Versnel H, King AJ. Development of
56. Tsunoda K, Yamane Y, Nishizaki M, Tanifuji M. COmplex contralateral and ipsilateral frequency representations in
objects are represented in macaque inferotemporal cortex ferret primary auditory cortex. Eur J Neurosci 2006;23:780-
by the combination of feature columns. Nature Neurosci 792.
2001;4:832-838. 73. Masino SA, Kwon MC, Dory Y, Frostig RD. Characteri-
57. Vnek N, Ramsden BM, Hung CP, Goldman-Rakic PS, Roe zation of functional organization within rat barrel cortex
AW. Optical imaging of functional domains in the cortex using intrinsic signal optical imaging through a thinned
of the awake and behaving monkey. Proc Natl Acad Sci skull. Proc Natl Acad Sci U S A 1993;90:9998-10002.
U S A 1999;96:4057-4060. 74. Narayan SM, Santori EM, Blood AJ, Burton JS, Toga AW.
58. Shtoyerman E, Arieli A, Slovin H, Vanzetta I, Grinvald Imaging optical reflectance in rodent barrel and forelimb
A. Long-term optical imaging an spectroscopy reveal sensory cortex. Neuroimage 1994;1:181-190.
mechanisms underlying the intrinsic signal an stability 75. Narayan SM, Santori EM, Toga AW. Mapping functional ac-
of cortical maps in V1 of behaving monkeys. J Neurosci tivity in rodent cortex using optical intrinsic signals. Cereb
2000;20:8111-8121. Cortex 1994;4:195-204.
59. Raffi M, Siegel RM. Functional architecture of spatial at- 76. Blood AJ, Narayan SM, Toga AW. Stimulus parameters in-
tention in the parietal cortex of the behaving monkey. J fluence characteristics of optical intrinsic signal responses
Neurosci 2005;25:5171-5186. in somatosensory cortex. J Cereb Blood Flow Metab
60. Chapman B, Stryker MP, Bonhoeffer T. Development of 1995;15:1109-1121.
orientation preference maps in ferret primary visual cor- 77. Chen-Bee CH, Kwon MC, Masino SA, Frostig RD. Areal
tex. J Neuroscience 1996;16:6443-6453. extent quantification of functional representations us-
61. Sengpiel F, Stawinski P, Bonhoeffer T. Influence of expe- ing intrinsic signal optical imaging. J Neurosci Methods
rience on orientation maps in cat visual cortex. Nature 1996;68:27-37.
Neurosci 1999;2:727-732. 78. Tommerdahl M, Delemos KA, Vierck CJ, Favorov OV,
62. Trachtenberg JT, Stryker MP. Rapid anatomic plasticity of Whitsel BL. Anterior parietal cortical response to tactile
horizontal connections in the developng visual cortex. J and skin-heating stimuli applied to the same skin site.
Neurosci 2001;21:3476-3482. J Neurophysiol 1996;75:2662-2670.
63. Kato T, Kamei A, Takashima S, Ozaki T. Human visual 79. Tommerdahl M, Delemos KA, Favorov OV, Metz CB,
cortical function during photic stimulation monitoring by Vierck CJ, Whitsel BL. Response of anterior parietal cortex
148 SECTION I TECHNIQUES

to different modes of same-site skin stimulation. J Neuro- 97. Schwartzkroin PA. Epilepsy. Models, Mechanisms and
physiol. 1998;80:3272-3283. Concepts. Cambridge: Cambridge University Press, 1993.
80. Tommerdahl M, Delemos KA, Whitsel BL, Favorov OV, 98. Federico P, Borg SG, Salkauskus AG, MacVicar BA. Map-
Metz CB. Response of anterior parietal cortex to cuta- ping patterns of neuronal activity and seizure propagation
neous flutter versus vibration. J Neurophysiol 1999;82:16- by imaging intrinsic optical signals in the isolated whole
33. brain of the guinea-pig. Neuroscience 1994;58:461-480.
81. Brett-Green BA, Chen-Bee CH, Frostig RD. Compar- 99. Hochman DW, Baraban SC, Owens JW, Schwartzkroin
ing the functional representations of central and border PA. Dissociation of synchronization and excitability in
whiskers in rat primary somatosensory cortex. J Neurosci furosemide blockade of epileptiform activity. Science
2001;21:9944-9954. 1995;270:99-102.
82. Takashima I, Kajiwara R, Iijima T. Voltage-sensitive dye 100. Chen JW, O’Farrell AM, Toga AW. Optical intrinsic sig-
versus intrinsic signal optical imaging: comparison of op- nal imaging in a rodent seizure model. Neurology
tically determined functional maps from rat barrel cortex. 2000;55:312-315.
Neuroreport 2001;12:2889-2894. 101. Schwartz TH, Bonhoeffer T. In vivo optical mapping of
83. Chen LM, Friedman RM, Ramsden BM, LaMotte RH, Roe epileptic foci and surround inhibition in ferret cerebral
AW. Fine-scale oraganization of S1 (Area 3b) in the squir- cortex. Nat Med 2001;7:1063-1067.
rel monkey revealed with intrinsic optical imaging. J Neu- 102. Suh M, Bahar S, Mehta AD, Schwartz TH. Temporal de-
rophysiol 2001;86:3011-3029. pendence in uncoupling of blood volume and oxygena-
84. Shoham D, Grinvald A. The cortical representation of the tion during interictal epileptiform events in rat neocortex.
hand in macaque and human area S-I: high resolution J Neurosci 2005;25:68-77.
optical imaging. J Neurosci 2001;21:6820-6835. 103. Zhao M, Suh M, Ma H, Perry C, Geneslaw A, Schwartz TH.
85. Toga AW, Cannestra AF, Black KL. The temporal/spatial Decreases in perfusion and hemoglobin oxygenation pre-
evolution of optical signals in human cortex. Cereb Cortex cede focal seizure onset in spontaneous human lesional
1995;5:561-565. epilepsy. Epilepsia. 2007;48(11):2059-2067.
86. Cannestra AF, Black KL, Martin NA, et al. Topographical 104. Schwartz TH. Optical imaging of epileptiform events in
and temporal specificity of human intraoperative optical visual cortex in response to patterned photic stimulation.
intrinsic signals. Neuroreport 1998;9:2557-2563. Cereb Cortex 2003;13:1287-1298.
87. Sato K, Nariai T, Sasaki S, et al. Intraoperative intrinsic 105. Ma H, Geneslaw A, Zhao M, Suh M, Perry C, Schwartz
optical imaging of neuronal activity from subdivisions of TH. The importance of latency in the focality of perfusion
the human primary somatosensory cortex. Cereb Cortex and oxygenation changes associated with triggered after-
2002;12:269-280. discharges in human cortex. J Cereb Blood Flow Metab.
88. Bartocci M, Bergqvist LL, Lagercrantz H, Anand KJ. Pain 2009;29(5):1003-1014.
activates cortical areas in the preterm newborn brain. Pain 106. Haglund MM, Hochman DW. Optical imaging of epilep-
2006;122:109-117. tiform activity in human neocortex. Epilepsia 2004;45:43-
89. Haglund MM, Ojemann GA, Hochman DW. Optical imag- 47.
ing of epileptiform and functional activity in human cere- 107. Chen JWY, O’Farrell AM, Toga AW. Optical intrinsic
bral cortex. Nature 1992;358:668-671. signal imaging in a rodent seizure model. Neurology
90. Cannestra AF, Bookheimer SY, Pouratian N, et al. Tem- 2000;55:312-315.
poral and topographical characterization of language 108. Villringer A, Planck J, Stodieck S, Botzel K, Schleinkofer L,
cortices using intraoperative optical intrinsic signals. Neu- Dirnagl U. Noninvasive assessment of cerebral hemody-
roimage 2000;12:41-54. namics and tissue oxygenation during activation of brain
91. Pouratian N, Bookheimer SY, O’Farrell AM, et al. Optical cell function in human adults using near infrared spec-
imaging of bilingual cortical representations. Case report. troscopy. Adv Exp Med Biol 1994;345:559-565.
J Neurosurg 2000;93:676-681. 109. Steinhoff BJ, Herrendorf G, Kurth C. Ictal near infrared
92. Fallgatter AJ, Muller TJ, Strik WK. Prefrontal hypooxy- spectroscopy in temporal lobe epilepsy: a pilot study.
genation during language processing assessed with Seizure 1996;5:97-101.
near-infrared spectroscopy. Neuropsychobiology 1998;37: 110. Adelson PD, Nemoto E, Scheuer M, et al. Noninvasive
215-218. continuous monitoring of cerebral oxygenation periictally
93. Minagawa-Kawai Y, Mori K, Furuya I, Hayashi R, Sato using near-infrared spectroscopy: a preliminary report.
Y. Assessing cerebral representations of short and long Epilepsia 1999;40:1484-1489.
vowel categories by NIRS. Neuroreport 2002;13:581-584. 111. Watanabe E, Maki A, Kawaguchi F, Yamashita Y, Koizumi
94. Taga G, Asakawa K. Selectivity and localization of corti- H, Mayanagi Y. Noninvasive cerebral blood volume mea-
cal response to auditory and visual stimulation in awake surement during seizures using multichannel near infrared
infants aged 2 to 4 months. Neuroimage 2007;36(4):1246- spectroscopic topography. J Biomed Opt 2000;5:287-290.
1252. 112. Watanabe E, Nagahori Y, Mayanagi Y. Focus diagnosis
95. Hauser WA, Hesdorfer DC. Epilepsy: Frequency, Causes of epilepsy using near-infrared spectroscopy. Epilepsia
and Consequences. New York: Demos, 1990. 2002;43(Suppl 9):50-55.
96. Bragin A, Mody I, Wilson CL, Engel JJ. Local generation 113. Sokol DK, Markand ON, Daly EC, Luerssen TG, Malkoff
of fast ripples in epileptic brain. J Neurosci 2002;22:2012- MD. Near infrared spectroscopy (NIRS) distinguishes
2021. seizure types. Seizure 2000;9:323-327.
CHAPTER 11 OPTICAL SPECTROSCOPIC IMAGING OF THE HUMAN BRAIN—CLINICAL APPLICATIONS 149

114. Arca Diaz G, Cesaron E, Alfonso I, Dunoyer C, Yaylali 124. Saitou H, Yanagi H, Hara S, Tsuchiya S, Tomura S. Cere-
I. Near infrared spectroscopy in the management of sta- bral blood volume and oxygenation among poststroke
tus epilepticus in a young infant. Eur J Paediatr Neurol hemiplegic patients: effects of 13 rehabilitation tasks mea-
2006;10:19-21. sured by near-infrared spectroscopy. Arch Phys Med Re-
115. Wolf T, Lindauer U, Reuter U, et al. Noninvasive near in- habil 2000;81:1348-1356.
frared spectroscopy monitoring of regional cerebral blood 125. Miyai I, Yagura H, Oda I, et al. Premotor cortex is involved
oxygenation changes during peri-infarct depolarizations in restoration of gait in stroke. Ann Neurol 2002;52:188-
in focal cerebral ischemia in the rat. J Cereb Blood Flow 194.
Metab 1997;17:950-954. 126. Miyai I, Yagura H, Hatakenaka M, et al. Longitudinal op-
116. Chen WG, Li PC, Luo QM, Zeng SQ, Hu B. Hemody- tical imaging study for locomotor recovery after stroke.
namic assessment of ischemic stroke with near-infrared Stroke 2003;34:2866-2870.
spectroscopy. Space Med Med Eng (Beijing) 2000;13:84- 127. Robertson CS, Gopinath SP, Chance B. A new applica-
89. tion for near-infrared spectroscopy: detection of delayed
117. Zepeda A, Vaca L, Arias C, Sengpiel F. Reorganization of intracranial hematomas after head injury. J Neurotrauma
visual cortex maps after focal ischemic lesions. J Cereb 1995;12:591-600.
Blood Flow Metab 2003;23(7):811-920. 128. Adelson PD, Nemoto E, Colak A, Painter M. The use of
118. Zepeda A, Sengpiel F, Guagnelli MA, Vaca L, Arias C. near infrared spectroscopy (NIRS) in children after trau-
Functional reorganization of visual cortex maps after is- matic brain injury: a preliminary report. Acta Neurochir
chemic lesions is accompanied by changes in expression Suppl 1998;71:250-254.
of cytoskeletal proteins and NMDA and GABA-A receptor 129. Kampfl A, Pfausler B, Denchev D, Jaring HP, Schmutzhard
subunits. J Neurosci 2004;24:1812-1821. E. Near infrared spectroscopy (NIRS) in patients with se-
119. Benaron DA, Hintz SR, Villringer A, et al. Noninvasive vere brain injury and elevated intracranial pressure. A pi-
functional imaging of human brain using light. J Cereb lot study. Acta Neurochir Suppl 1997;70:112-114.
Blood Flow Metab 2000;20:469-477. 130. Dunham CM, Sosnowski C, Porter JM, Siegal J, Kohli C.
120. Kato H, Izumiyama M, Koizumi H, Takahashi A, Itoyama Correlation of noninvasive cerebral oximetry with cere-
Y. Near-infrared spectroscopic topography as a tool to bral perfusion in the severe head injured patient: a pilot
monitor motor reorganization after hemiparetic stroke: study. J Trauma 2002;52:40-46.
a comparison with functional MRI. Stroke 2002;33:2032- 131. Lewis SB, Myburgh JA, Thornton EL, Reilly PL. Cerebral
2036. oxygenation monitoring by near-infrared spectroscopy is
121. Sakatani K, Xie Y, Lichty W, Li S, Zuo H. Language- not clinically useful in patients with severe closed-head
activated cerebral blood oxygenation and hemodynamic injury: a comparison with jugular venous bulb oximetry.
changes of the left prefrontal cortex in poststroke apha- Crit Care Med 1996;24:1334-1338.
sic patients: a near-infrared spectroscopy study. Stroke 132. Buchner K, Meixensberger J, Dings J, Roosen K. Near-
1998;29:1299-1304. infrared spectroscopy–not useful to monitor cerebral oxy-
122. Murata Y, Sakatani K, Katayama Y, Fukaya C. Increase genation after severe brain injury. Zentralbl Neurochir
in focal concentration of deoxyhaemoglobin during neu- 2000;61:69-73.
ronal activity in cerebral ischaemic patients. J Neurol Neu- 133. Mayhew JE, Askew S, Zheng Y, et al. Cerebral vasomotion:
rosurg Psychiatry 2002;73:182-184. a 0.1-Hz oscillation in reflected light imaging of neural
123. Murata Y, Sakatani K, Hoshino T, et al. Effects of cere- activity. Neuroimage 1996;4:183-193.
bral ischemia on evoked cerebral blood oxygenation 134. Obrig H, Neufang M, Wenzel R, et al. Spontaneous low
responses and BOLD contrast functional MRI in stroke frequency oscillations of cerebral hemodynamics and me-
patients. Stroke 2006;37:2514-2520. tabolism in human adults. Neuroimage 2000;12:623-639.
This page intentionally left blank
Chapter 12
Electrocorticographic Spectral Analysis
Mackenzie C. Cervenka and Nathan E. Crone
Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland

䉴 INTRODUCTION AND later quantified by event-related analysis of changes in


HISTORICAL PERSPECTIVE band-specific alpha power, as discussed later.6,7 Sev-
eral studies have revealed event-related changes in scalp
Electrocorticographic (ECoG) spectral analysis is an EEG spectra that correlate temporally and anatomically
emerging method for mapping human brain function with the planning and execution of movement,6−14 au-
that originally developed from the clinical circumstances ditory activation,15−18 visual stimulation,19−22 and lan-
and needs of patients undergoing surgery for intractable guage processing.23−27
epilepsy. From almost the inception of epilepsy surgery, Recent clinical practice has evolved to allow for
identification of “eloquent” cortex relied primarily on placement of subdural electrodes directly on the sur-
electrocortical stimulation mapping (ESM), in which di- face of the brain to measure electrocortical activity
rect electrical stimulation of cortex interferes with mo- with improved spatial resolution and signal quality.28
tor, perceptual, or linguistic function. With the advent These ECoG recordings have yielded results similar to
of long-term intracranial electroencephalography (EEG) the previous noninvasive EEG findings. In addition,
monitoring for localization of the ictal seizure focus, ESM ECoG spectral analysis has revealed energy changes
was no longer limited to the stressful circumstances of in higher frequency bands such as gamma (30–60
awake craniotomies,1 but its clinical utility was still lim- Hz) and high-gamma (>60 Hz) frequency bands that
ited by its propensity for inducing seizures and some- were undetectable or uninterpretable with surface EEG
times pain during stimulation. Furthermore, because recordings.29,30
ESM had to be done sequentially at pairs of electrodes, ECoG spectral analysis is currently being used ex-
preferably finding the optimal stimulus intensity at each perimentally in humans to localize sensorimotor, audi-
pair, it was sometimes too time consuming to test more tory, and language cortices during extraoperative inva-
than a few tasks at every stimulation site. For these sive monitoring for epilepsy surgery. The clinical utility
reasons, clinicians explored the possibility of mapping of this technique, including its sensitivity and specificity
cortical function by utilizing the ECoG signals already with respect to ESM and other mapping techniques, is
being recorded for clinical purposes in these patients. Al- still under investigation and is expected to evolve with
though previous researchers had successfully exploited additional clinical research as well as continuing ad-
the phase-locked components of ECoG responses to vances in instrumentation, data acquisition, and data
stimuli or events (i.e., evoked and event-related poten- analysis. ESM is still widely considered to be the gold
tials [ERPs]) for this purpose, recent efforts toward standard for localizing eloquent cortex preoperatively.
ECoG functional mapping have focused on task-induced In the future, however, the information obtained from
changes in ongoing cerebral activity that are not neces- ECoG spectral analysis may complement the results ob-
sarily phase-locked to a stimulus or event. tained from ESM and other mapping techniques and
Broadly speaking, the general rationale and strat- could, in some circumstances, supplant ESM. In addi-
egy for analyzing ECoG spectra can be traced back to tion, ECoG continues to offer a rare opportunity to study
the early days of EEG, when neuroscientists first de- the working brain with unprecedented spatial and tem-
scribed specific background rhythms with distinct fre- poral resolution. The application of ECoG spectral anal-
quency ranges, reactivities, and spatial distributions over ysis to research in systems and cognitive neuroscience
the cortex.2−5 Suppression of occipital alpha activity (in has grown exponentially in recent years. In addition to
the 8–12 Hz range) and central mu activity was ob- studies of the brain mechanisms of selective attention,
served during functional activation of visual and senso- language, and memory, ECoG spectral analyses are being
rimotor cortices, respectively. These phenomena were used increasingly to drive brain–computer interfaces (BCI).

151
152 SECTION I TECHNIQUES

䉴 UNDERLYING PRINCIPLES cortical neurons into organized band-specific rhythms.


Band-limited power suppression during cortical activa-
Event-related changes in EEG spectral energy can be an- tion is thus assumed to represent a loss of this syn-
alyzed by averaging signals from multiple trials in the chronization, thus the term desynchronization. However,
time domain or in the frequency domain.7 Averaging in power suppression is not always restricted to frequen-
the time domain consists of averaging each sample of cies where there are well-defined peaks in the baseline
the raw EEG time series across multiple realizations or power spectrum, and in these circumstances it may be
trials of a task or behaviorally relevant event under study. more appropriate to adopt a more conservative, descrip-
This procedure discards response components that are tive terminology. Nevertheless, ERD may still be used by
not phase-locked to the event under study and neces- some investigators as a convenient shorthand to refer to
sarily yields phase-locked responses commonly known power suppression associated with cortical activation.
as evoked or event-related potentials. Averaging in the Likewise, event-related power increases, that is, power
frequency domain, in contrast, reveals changes in EEG augmentation, are often referred to as event-related syn-
spectral energy that are time-locked, but not necessar- chronization (ERS).
ily phase-locked, to events. The procedure consists of ERS originally referred to band-limited power in-
first converting the raw EEG time series into time- and creases in scalp EEG. For example, ERS was observed
frequency-dependent estimates of signal energy. The re- in beta frequencies immediately after ERD in motor cor-
sulting time–frequency spectra are then averaged with tex following movement,36,37 and it was observed in al-
respect to the task or event under study. Note that the en- pha frequencies over cortex not engaged by a task, that
ergy changes observed with this approach include both is, at rest or idling.38 Subsequent observations of band-
nonphase-locked and phase-locked components unless limited power increases in gamma frequencies some-
special procedures are used to mitigate the contribution times adopted the same ERS term for convenience. How-
of phase-locked components.31−33 ever, event-related spectral analyses may also reveal
After averaging EEG signals in the frequency do- power increases that are not band-limited. Because these
main, event-related spectral changes can be quanti- responses do not have a clear peak in the power spec-
fied and statistically tested by comparing time- and trum, they do not necessarily reflect synchronization of
frequency-specific energy estimates during events with neuronal networks into rhythmic or oscillatory behavior
those in a reference or “baseline” interval when the brain (see later), and thus the usual connotations of the ERS
is assumed to be “at rest,” that is, relatively inactive or terminology do not apply. For these reasons, it may be
at least not engaged in the event or task under study.7 most appropriate to limit the usage of ERD and ERS to
This reference interval is most commonly chosen imme- event-related power decreases or increases that are ob-
diately prior to the onset of each trial. Changes detected served in relatively narrow frequency bands. The distinc-
by this approach may consist of increases or decreases tion between narrow- and wide-band spectral responses,
of signal energy that depend on the frequency of the however, is not well defined in the literature, and it is
signal as well as its temporal relationship to the task or not clear what spectral profile a response should have
event under study and the location at which the signal in order to be called “oscillatory.”
is recorded with respect to functionally activated cortex. The physiologic frequency bands in which event-
The terminology used to describe different event- related spectral changes have been commonly inves-
related spectral changes is still evolving in the literature tigated with noninvasive scalp EEG include the theta
due to a lack of consensus about the functional signifi- (4–7 Hz), alpha (8–13 Hz), and beta (13–25 Hz) fre-
cance of different spectral phenomena, about the opti- quency bands. Although scalp EEG has also been used
mal balance between lumping and splitting these phe- to study event-related spectra at gamma frequencies
nomena, and about how to best reflect our evolving (>30 Hz),11,39 ECoG recordings have offered much bet-
understanding of their neural generators and generat- ter signal quality at high frequencies28,40,41 and have thus
ing mechanisms. For example, the term event-related been instrumental in studying the response properties
desynchronization (ERD), originally coined by Gert of event-related gamma responses, particularly those in
Pfurtscheller,34 is commonly used to refer to event- “high gamma” frequencies above the traditional band-
related suppression of power. This power suppression is limited responses at 40 Hz,30,39 to be discussed in greater
usually observed most prominently in frequency bands detail later.
that are defined by peaks in the power spectrum dur- Interpretations of event-related power changes
ing baseline intervals. These band-limited peaks typi- must always take into account the timing of these
cally correspond to “resting rhythms” such as the occipi- changes with respect to the timing of functional brain
tal alpha rhythm of resting visual cortex,2 the central mu activation. This time dependence of event-related spec-
rhythm of sensorimotor cortex,5 or the tau rhythm of tra was first demonstrated with scalp EEG recordings. For
auditory cortex.35 These spectral peaks are assumed to example, ERD was observed in the alpha and beta fre-
require the synchronization of a sizeable population of quency ranges over the sensorimotor cortex before and
CHAPTER 12 ELECTROCORTICOGRAPHIC SPECTRAL ANALYSIS 153

during movement.9 In contrast, alpha and beta ERS were test (Wada test), magnetoencephalography (MEG), and
observed over surrounding areas responsible for move- or ictal/interictal single positron emission tomography
ment of other body parts, that is, leg, suggesting that (SPECT). These findings help define the target of resec-
alpha and beta ERS may reflect cortical inhibition. Fol- tion and/or predict whether it is near functionally impor-
lowing movement, there was an increase in beta power tant motor, sensory, auditory, and/or language cortices.
over the same region where ERD was observed during Once patients have been selected for intracranial
movement, indicating a reset of the functional circuitry EEG monitoring, functional mapping with ECoG spec-
within motor cortex.42 In general, power suppression or tral analysis, like traditional ESM, requires patients to be
ERD in alpha and beta bands likely reflects functional willing and able to cooperate with testing. Patients must
activation of cortex while ERS in the same frequencies be able to respond quickly and consistently so that ei-
may in some cases index cortical inhibition. An impor- ther signal averaging, or stimulation in the case of ESM,
tant exception to this is the increase in low frequency can be temporally aligned to functional brain activation.
power that is often observed simultaneously with phase- Generally speaking, children younger than 5 years or
locked responses to stimulus onset.33 A broad, but often patients with severe cognitive or behavioral limitations
useful, generalization is that power changes in higher may have difficulty with this, though one recent study
(gamma) frequencies have an inverse relationship to suggested that ECoG might overcome some of the limita-
those of power changes in alpha/beta frequencies. That tions of ESM in children.46 Excessive sedation from pain
is, gamma power augmentation likely indexes corti- medications, or neurotoxic side effects from antiepileptic
cal activation,29,30,39,43 while gamma power suppression medications, can also interfere with testing.
may in some cases index cortical inhibition.44 However,
it is important to recognize that with any given cortical
activation the transition in the power spectrum between
power augmentation at higher frequencies and power INSTRUMENTATION
suppression in lower frequencies is quite variable, and
it is potentially dangerous to assume any fixed boundary Electrodes used for recording intracranial EEG vary be-
between the two. tween medical centers. Subdural electrode arrays are
most commonly used and consist of platinum–iridium
discs (4 mm diameter, 2.3-diameter exposed surface, 1.5
䉴 METHODOLOGY mm thickness) embedded in a soft silastic sheet with an
interelectrode distance of 1-cm (Ad-tech Medical Instru-
SUBJECT INCLUSION AND ment Corporation, Racine, WI). The electrode configura-
PREPARATION tion, including the position and number of electrodes, is
determined by the epilepsy and/or neurosurgical team
Intracranial EEG is indicated under a limited set of clin- based on a variety of clinical considerations. Electrode
ical circumstances. The most common indication is for configurations can include one-dimensional strips of 4 to
patients with intractable epilepsy, in whom seizures have 8 electrodes or two-dimensional arrays (grids) of up to
not been adequately controlled with medications and are 8 × 8 electrodes (see Fig. 12–1). Several electrode ar-
occurring with sufficient severity and frequency to dis- rays can be combined to provide adequate coverage
rupt the patient’s quality of life. In these patients, EEG of the cortical surface of interest and grids can be cut
electrodes may be surgically implanted in order to better to accommodate anatomic variability. Grids and strips
localize the seizure focus and/or to map cortical function with smaller interelectrode distances are available, but
when the seizure focus is suspected to be near or within require a larger number of electrodes and EEG ampli-
functionally critical cortex. Long-term monitoring with fiers to cover a comparable cortical region.
intracranial electrodes is particularly useful for localizing Stereotactically implanted depth electrodes placed
the ictal onset zone, which is more predictive of postsur- according to a Talairach atlas and/or a patient’s MRI
gical outcomes than interictal epileptiform discharges.45 findings provide another modality for obtaining invasive
Other potential indications arise in patients with brain EEG recordings. Depth electrodes are especially useful
tumors or vascular malformations in or near eloquent when recording from deep brain structures such as the
cortex, though many of these patients may be candi- amygdala, hippocampus, or a region of suspected cor-
dates for intraoperative stimulation mapping. tical dysplasia. Depth electrodes are typically smaller
The placement of subdural grids, strips, and/or than those used for subdural grid and strip implanta-
depth electrodes is dictated solely by clinical con- tion and may provide improved spatial resolution. Al-
siderations derived from the patient’s seizure semiol- though fewer studies of event-related spectral changes
ogy, ictal and interictal scalp EEG, structural and/or have used these electrodes than subdural electrodes, the
functional magnetic resonance imaging (MRI), positron results obtained with the depth electrodes do not appear
emission tomography (PET), intracarotid amobarbital to be fundamentally different.47,48
154 SECTION I TECHNIQUES

Figure 12–1. An example of a patient with electrocortical stimulation mapping (ESM)


and ECoG spectral analysis during a reading task. Subdural intracranial grid
electrodes are depicted over the dominant hemisphere with blue circles. The colored
bars indicate electrode pairs that were stimulated with bipolar ESM. Green indicates
that reading was not inhibited. Purple indicates that reading was inhibited. The yellow
lightning bolts indicate that ESM produced a seizure and that those electrodes could
not be analyzed further with ESM. ECoG spectral analysis revealed significant gamma
and high-gamma activation at the electrode that could not be tested with ESM,
indicating that the region is active during reading task performance. On the spectral
plot to the right, time in seconds is on the X-axis and frequency in Hertz is on the
Y-axis. An increase in power is shown on the red to yellow scale and a decrease in
power is shown on a light to dark blue scale (to the right of the diagram).

RECORDING TECHNIQUES AND DATA High gamma activity (>60 Hz) has a very low am-
ACQUISITION plitude and, therefore, requires significant signal am-
plification for adequate detection and evaluation. The
While scalp EEG has usually been able to accurately bit-depth of analog-to-digital (A/D) converters is ideally
detect frequency changes in the beta frequency range at least 16-bits. Because intracranial mapping can require
and recently even the lower gamma frequency bands,39 extensive use of multiple grids and strips, over 100 ECoG
electrocorticography accurately detects signal changes channels may be required to capture all of the relevant
in much higher frequency ranges.30,43 Sampling rates of data. Clinical video–EEG monitoring systems with up to
1000 Hz and higher are preferable to adequately study 128 ECoG channels are widely available, and a few sys-
these higher frequencies. The Nyquist–Shannon sam- tems now offer 256 ECoG channels. In our experience,
pling theorem49,50 states that the sampling rate frequency the more the better, particularly if one contemplates us-
must be greater than twice the maximum frequency be- ing high-density grid recordings.
ing sampled in order to reconstruct the signal, and higher Detection of event-related spectral changes (or
sampling rate frequencies are required to accurately de- event-related potentials) requires placement of precise
scribe the waveform characteristics. In practice, it is often record markers indicating the onset of a task elicit-
better to oversample as much as is practical. ing functional brain activation. This requires the use of
CHAPTER 12 ELECTROCORTICOGRAPHIC SPECTRAL ANALYSIS 155

dedicated channels that mark events within the ECoG 䉴 APPLICATIONS


recording for subsequent analyses in the time and fre-
quency domains. Detecting robust ECoG power changes ECoG spectral analysis has been used to map a va-
requires averaging over multiple trials; therefore, appro- riety of functional–anatomical domains of human cor-
priate stimuli and numbers of trials must be chosen to tex, including the frontal eye fields,44 visual cortex,71−74
identify cortical areas of interest (i.e., somatosensory, auditory cortex,32,33,75,76 premotor and primary motor
motor, language, auditory cortices). A minimum of 25 cortices,29,77−85 somatosensory cortex,86 and language
trials are recommended but greater than 50 trials may be cortex.46,58,69,70,87−91 ECoG spectral analysis has also
necessary, dictated by the quantity of interictal epilep- been used to study cortical mechanisms of attention and
tiform activity and patient performance, thus allowing memory.8,65,79,92−97
for rejection of artifact, bad trials, and inconsistent re-
sponses. In practice, we often use stimulus lists with no
fewer than 100 trials. MAPPING SENSORIMOTOR FUNCTION

Mapping sensorimotor cortex may be motivated not only


SIGNAL ANALYSIS by difficulties identifying the central sulcus by visual in-
spection through a craniotomy or with neuronavigation,
Event-related spectral analysis of ECoG signals is sim- but also by the variability among individuals in the exact
ilar to analysis performed on scalp EEG recordings. location of different motor and sensory representations,
However, ECoG signal analysis does not require the particularly in the setting of lesions that might distort
same degree of spatial reformatting as scalp EEG51 be- the anatomy or alter these representations. Localization
cause recordings from the cortical surface are already of hand area usually can be accomplished efficiently by
strongly biased in favor of local potentials. A reference- recording somatosensory-evoked potentials (SEPs) from
independent montage such as a common average refer- median nerve stimulation. This can be done intraoper-
ence is typically used to reformat ECoG data and remove atively with a strip of electrodes placed on the cortical
biases introduced by different distances between the ref- surface, or extraoperatively with subdural ECoG.86,98,99
erence electrode and active electrodes in the array.30 This procedure, which can also be done noninvasively
Placement of the reference electrode far from the active with MEG,100 is usually successful in localizing the cen-
electrodes can reduce this bias, but extracranial elec- tral sulcus in the hand area though care must be taken
trode placement is more permissive of noise from scalp in the interpretation of the resulting potentials.101 In ad-
myogenic potentials. Often, the most effective method is dition to median nerve stimulation, the tibial nerve and
to assign a reference electrode within the subdural array ulnar nerve can be stimulated to obtain SEPs at other
that is as distant as possible from sites of epileptiform sites along the central sulcus.102 Although this can be
activity and functional brain activity of interest. a very efficient and effective means of identifying peri-
ECoG signals are converted into the frequency do- Rolandic cortex, ESM is often used in addition to SEPs or
main in order to detect event-related changes in the in place of them to define the functional anatomy of sen-
power spectrum. Band-pass filtering was one of the sorimotor cortex. Under these circumstances, it may be
earliest approaches developed for quantitative analysis possible to complement ESM with maps of sensorimotor
of scalp ERD and ERS.7,52 Algorithms employing short- cortex obtained through ECoG spectral analysis.
term Fourier and wavelet transforms have also been A growing number of studies have suggested the
used.49,53 A matching pursuit (MP) algorithm is routinely clinical utility of ECoG spectral mapping for map-
being used by our group for ECoG spectral analysis.54−58 ping sensorimotor function in patients undergoing
The algorithm is designed to decompose the EEG sig- surgery.13,29,47,68,78,81,85,103,104 As with scalp EEG record-
nal by selecting wavelet packet vectors that sequentially ings, alpha and beta ERD has been observed during
optimize the signal approximation.59 It produces high- ECoG recordings of various tasks designed to stimulate
resolution maps of event-related power changes within and identify regions of the sensorimotor cortex.13,29,81,105
the time–frequency plane by assigning dictionaries con- In one such study of visually cued isometric muscle
structed from Gabor functions. contractions, alpha ERD occurred in a relatively diffuse
Once ECoG activity has been identified in various distribution at onset of the motor response and coa-
cortical regions, this information can be coregistered lesced into a more discrete, somatotopically localized
with surface renderings of three-dimensional MRI/CT pattern only later, as the isometric muscle contraction
(computed tomography) reconstructions of the brain was sustained.13 Beta ERD occurred in a more discrete
and electrode locations. This allows for direct compar- and somatotopically specific pattern but with a less ro-
isons to be made with other cortical mapping modalities bust, or absent, response at times. In addition, unilateral
such as PET, SPECT, MEG, functional MRI (fMRI), or ESM limb movements were sometimes associated with alpha
(see Fig. 12–1).60−70 and beta ERD over bilateral sensorimotor cortices with
156 SECTION I TECHNIQUES

overlapping patterns for different body parts. In contrast augmentation that correlated temporally and spatially
to these patterns of power suppression, the same mo- with the cortical populations responsible for movement.
tor task was also associated with event-related gamma Taken as a whole, these reports support the clinical util-
power increases in spatial distributions that were more ity of ECoG spectral mapping in patients undergoing sur-
discrete, more lateralized to contralateral sensorimotor gical resections near or within motor cortex.
cortex, and more somatotopically specific than those of ECoG high-gamma responses have also been ob-
alpha and beta ERD. In addition, power increases in “low served during activation of somatosensory cortex. When
gamma” frequencies (35–50 Hz), like alpha ERD, were vibrotactile stimuli were attended to, they were associ-
sustained during the muscle contraction, while power in- ated with greater high-gamma responses than when they
creases in “high gamma” frequencies (750–100 Hz) were were ignored.97 This was consistent with an MEG study
transient and covaried with the onset latency of motor by Gross et al.107 in which perceived nociceptive stimuli
responses. The functional localization of gamma power were associated with stronger 60–95-Hz gamma oscilla-
increases correlated well with ESM of motor function. tions in primary somatosensory cortex than unperceived
Several other studies have evaluated ECoG gamma stimuli of equal stimulus intensity. In another MEG study
responses in sensorimotor cortex using a variety of by Ihara et al.,108 source power in a high-gamma band
different tasks and methods for data acquisition and (70–90 Hz) increased simultaneously in contralateral SI
analysis.47,81,103,106 For example, Ohara et al.78 observed and contra/ipsilateral SII at 80–180 milliseconds, sug-
nonphase-locked gamma activity in SI and MI extending gesting a role in functional connectivity between SI and
up to 90 Hz during self-paced finger and wrist move- SII. In a subsequent MEG study by Hauck et al.,109
ments. Both low and high-gamma ERS were observed high-gamma responses to nociceptive stimuli were in-
only during contralateral movements, and high-gamma creased by directed attention to the stimuli, and coupling
activity (60–90 Hz) in particular was highly tempo- analysis of the high-gamma responses revealed stronger
rally correlated with movement onset. During self-paced functional interactions between ipsilateral and contralat-
tongue and finger movements, Pfurtscheller et al.104 also eral sites during attention. Similarly, another MEG study
observed broadband high-gamma responses (60–90 Hz) of normal subjects by Bauer et al.110 found that spa-
over sensorimotor cortex in a topographic pattern that tial tactile attention enhanced high-frequency gamma
was more discrete and somatotopically specific than the responses (60–95 Hz) over contralateral SI cortex. Al-
more widespread mu (alpha) and beta ERD, and in a though the upper boundary of the gamma responses
temporal pattern that also corresponded more closely to observed in this and several other MEG studies have
movement onset. ECoG high-gamma responses (60–200 been lower than those commonly observed with ECoG,
Hz) have also been demonstrated during activation of the sensitivity of MEG to higher frequency activity is
premotor cortex during a task designed to dissociate at- less than that of ECoG, and in some instances it may
tention from motor planning.79 A recent study correlated be difficult to distinguish between band-limited gamma
alpha and beta ERD/ERS during voluntary movement in oscillations and the broadband gamma responses more
a cognitive visuospatial task to functional MRI results commonly observed with ECoG.
during performance of a similar task77 and determined
that there was an “evident overlap in most results” but
that the ECoG mapping was more sensitive at detect- LOCALIZATION OF VISUAL FUNCTION
ing activity in the premotor and prefrontal cortices than
functional MRI. ECoG gamma activity has been demonstrated in vi-
In the largest series to date of ECoG spectral map- sual cortex during presentation of a variety of visual
ping of sensorimotor cortex, Miller et al.85 recorded sub- stimuli.71−74 Epilepsy patients with implanted poste-
dural ECoG in 22 subjects during a variety of motor tasks, rior depth electrodes were primarily studied. During
and the topographic patterns of ECoG power changes one experiment, patients performed a face detection
were compared for low frequencies (8–32 Hz) ver- task that revealed changes within a broad gamma fre-
sus high-gamma frequencies (76–100 Hz). High-gamma quency range (40–200 Hz) over the occipital, parietal,
power augmentation occurred in a more focused spatial and temporal cortices, most notably the fusiform gyrus,
distribution than did ERD and corresponded well to the the lateral occipital gyrus, and intraparietal sulcus.71
results of ESM, as well as the somatotopic organization The temporo-occipital changes were detected first, fol-
for movement of different body parts. Reddy et al.106 re- lowed by parietal activation. Alpha and beta activity
cently demonstrated in six patients that changes in high- was notably suppressed during the increase in gamma
gamma activity (50–160 Hz) can be accurately differenti- activity. Tallon-Baudry et al.72 demonstrated variable
ated during hand movement of a joystick in four cardinal gamma activity in the lateral occipital regions and the
directions (up, down, left, and right). fusiform gyrus during presentation of simple visual stim-
Despite the varying techniques used in the afore- uli. Gamma oscillations were enhanced by increased at-
mentioned studies, all revealed high-gamma power tention during visual stimulation in the fusiform gyrus,
CHAPTER 12 ELECTROCORTICOGRAPHIC SPECTRAL ANALYSIS 157

whereas attention increased baseline gamma activity in regions assessed in the previous studies described (see
the lateral occipital cortex prior to stimulus presentation. Sections “Localization of Visual Function” and “Localiza-
The authors concluded that the functional significance tion of Auditory Function”). Subsequent processing is re-
of changes in gamma activity depends on the region of quired, however, in dominant frontal, parietal, and tem-
activation. poral cortices. Several studies have directly compared
Vidal et al.111 used MEG to demonstrate differen- cortical mapping results with ECoG gamma activity map-
tial gamma frequency activation in response to various ping to ESM (for example, see Fig. 12–1).69,88,90 In one
visual task demands. During a visual task, high-gamma such study,88 ECoG high-gamma changes were studied
band (70–120 Hz) activation was found over the central during visual object naming, word reading, and word
occipital regions and low gamma activity (44–66 Hz) oc- repetition in a patient fluent in English and American
curred over the parietal regions during attentional tasks. Sign Language by eliciting spoken and signed responses.
Asano et al.74 recently demonstrated high-gamma (50– High-gamma power changes during auditory word rep-
150 Hz) augmentation in the anterior–medial occipital etition had the greatest magnitude in the posterior su-
cortex, followed by the lateral–polar occipital cortex in perior temporal gyrus, while activation during naming
response to stroboscopic flash-stimuli in nine children and reading was most prominent in the basal temporal–
undergoing intracranial monitoring in preparation for occipital cortex, including fusiform gyrus. Spoken re-
epilepsy surgery. Their results not only confirmed previ- sponses activated tongue regions of motor cortex and
ous findings demonstrating high-gamma activation that manual sign language responses activated more hand re-
temporally and spatially correlated with visual cortical gions. These ECoG results were found to correlate with
activity, but demonstrated the safety and tolerability of language ESM at many, but not all, sites.
this technique in the pediatric population. Tanji et al.89 observed ECoG high-gamma (80–120
Hz) activity during picture-naming of line drawings of
animals and tools and during lexical–decision tasks in a
LOCALIZATION OF AUDITORY
patient with bilateral lateral and basal temporal subdu-
FUNCTION
ral grid implantation. They demonstrated bilateral high-
gamma activity during the naming tasks with significant
ECoG spectral mapping has been applied in several stud-
differences during naming of tool and animal stimuli.
ies to auditory function.32,33,75,76 In one such study,32
The lexical–decision task comprise two distinct written
four left hemisphere-dominant patients with subdural
forms of Japanese, Kanji and Kana. Task stimuli included
electrodes over left superior temporal gyrus performed
words and pseudowords using each of these forms.
auditory tone and phoneme discrimination tasks, and
There was greater high-gamma activity over the dom-
event-related ECoG band power changes were investi-
inant left basal temporal region than the right during
gated. Phoneme discrimination produced a higher mag-
the lexical–decision tasks with significant differences be-
nitude of gamma augmentation over a more widespread
tween Kanji and Kana words and also between words
region of the superior temporal gyrus than did tone dis-
and pseudowords. The authors concluded that changes
crimination. Gamma augmentation was detected at not
in high-gamma activity could accurately map language
only 40 Hz but also high-gamma frequencies, extending
function within the dominant basal temporal region.
from ∼80 Hz to 200 Hz.
Sinai et al.69 directly compared ESM and ECoG high-
Edwards et al.75 performed ECoG recordings intra-
gamma activity (80–100 Hz) during visual object naming
operatively on 11 patients undergoing tumor resection
and found that the specificity of ECoG mapping relative
with five to nine epipial electrodes placed over the left
to ESM was 78%, with a relatively modest sensitivity of
lateral surface of the frontal, temporal, or inferior pari-
38%. The authors concluded that ECoG mapping could
etal lobe. High-gamma activity was detected over the
be used to create a preliminary map of language cortex
superior temporal gyrus during repetitive tone stimuli
that could later be assessed with electrocortical stimula-
(500 Hz) and more anteriorly in response to deviant
tion. This would enable a more focused evaluation when
stimuli (550 Hz), suggesting that goal-directed activity is
performing ESM of language function, limiting potential
not required to produce a detectable high-gamma power
complications such as inflicting pain, stimulating after-
change during auditory stimulation.
discharges, or inducing seizures. ECoG mapping could
also be used to evaluate sites at which ESM results are
MAPPING LANGUAGE CORTEX equivocal or could not be obtained.
Towle et al. investigated the localization of ECoG
ECoG spectral analysis has also been used to evaluate gamma power augmentation during auditory stimulation
the temporal and spatial characteristics of cortical activa- (warning tones), followed by word repetition and recall
tion during language tasks.8,46,58,69,88−90,112 Performance tasks in left hemisphere-dominant epilepsy patients.90
of these complex tasks requires perception of auditory They demonstrated ECoG high-gamma (70–100 Hz)
and visual stimuli, utilizing some of the same cortical power increases in primary auditory cortex in response
158 SECTION I TECHNIQUES

to auditory tones, in posterior temporal and parietal cor- homologous structures have assumed critical memory
tices during the presentation of word stimuli, and in lat- functions. Preoperative clinical assessment routinely in-
eral frontal and anterior parietal cortex during verbal re- cludes structural MRI, neuropsychological testing, and
sponses. Comparing these results to language ESM, they the intracarotid amobarbital test (Wada test). fMRI has
reported a sensitivity of 63%, with a specificity of 57%. also been used for this purpose.118−122 There have also
In another recent study, Sinai et al.70 compared au- been several recent studies of ECoG spectral changes
ditory high-gamma power changes with ESM over the during memory tasks.65,92−96 However, as when inter-
left lateral cortex of six patients with left hemisphere lan- preting language maps, care must be taken when in-
guage dominance based on Wada testing during speech terpreting the results of activation-based maps of mem-
comprehension and tones detection. Electrocortical stim- ory function, especially for ECoG assessment of memory
ulation interfered with speech comprehension but not function since electrode implantations are usually uni-
with tone detection in the posterior temporal region. lateral or asymmetric, making it difficult to compare the
Statistical analysis revealed high spatial concordance be- relative contribution of left versus right mesial temporal
tween ESM and high-gamma (>60 Hz) power changes structures.
with a specificity of 98%, a sensitivity of 67%, and a pos- Sederberg et al.93 studied ten patients with im-
itive predictive value of 67%. planted subdural grids and measured ECoG activity
The excellent specificity with relatively modest sen- while they memorized a list of common nouns. There
sitivity compared to ESM as identified here as well as in was an increase in theta (4–8 Hz) activity distributed
other studies previously discussed illustrates one of the over the right temporal and frontal cortices, and more
important potential limitations of mapping with electro- widespread power increases in higher frequencies (28–
corticography. Ideally, fMRI, ESM, and ECoG mapping 64 Hz) during successful encoding. In a follow-up study
results based on performance of identical tasks should of 39 epilepsy patients with surgically implanted sub-
be compared directly to postsurgical outcome results to dural and depth electrodes, Sederberg et al.96 demon-
determine which modality provides the most accurate strated increased gamma power (44–64 Hz) over the hip-
functional language map. pocampus, left temporal, and left frontal cortices during
Brown et al.46 evaluated three children with in- a similar recall task. Finally, Sederberg et al.95 recorded
tractable left-hemispheric focal epilepsy and demon- intracranial EEG activity in 52 patients during memory
strated gamma-power increases in the posterior superior retrieval tasks and using a broader frequency range for
temporal gyrus when they were presented questions, signal analysis. This study demonstrated a broadband in-
posterior lateral temporal and posterior frontal activation crease in gamma activity (28–100 Hz) in the hippocam-
between questions and responses, and pre- and post- pus and left temporal lobe as well as the prefrontal
central gyri activation immediately preceding and dur- cortex that distinguished retrieval of true versus false
ing responses. Their findings correlated with previous memories.
results found in the adult population and indicated that Although short-term memory, that is, working mem-
not only is ECoG analysis safe and reliable in children, ory, likely depends more on prefrontal cortex and is
but that the language localization results described in not usually a major concern for epilepsy surgery, it is
adults can be generalizable to the pediatric population. of great interest to cognitive neuroscientists and, there-
Previous pediatric studies indicated that ESM often fails fore, has also been studied with ECoG spectral analysis.
to localize language in children, and even Wada testing Howard et al.92 demonstrated an increase in gamma (30–
is less sensitive than in the adult population.113,114 There- 60 Hz) oscillatory activity during a working memory task
fore, ECoG analysis could potentially play a particularly and showed a linear correlation between an increase in
important role in functional language mapping prior to gamma activity and memory load over multiple trials.
surgical resection in the pediatric population. Mainy et al.112 evaluated nine patients with implanted
electrodes in mesial temporal structures and perisylvian
and prefrontal regions during a task of short-term con-
MAPPING MEMORY FUNCTION solidation in order to map encoding in working mem-
ory. They found an increase in high-gamma (50–150 Hz)
Preservation of memory function is a key concern for activity within an area referred to as the “phonological
planning epilepsy surgery, particularly in the temporal loop,” including within the frontal lobe, the prefrontal
lobe, and much of the preoperative assessment of pa- cortex, the precentral gyrus, and Broca’s area; in the
tients is focused on determining the structural and func- temporal lobe, the auditory cortex, the fusiform gyrus,
tional integrity of the neural substrates of memory, espe- and the hippocampus; and in the postcentral gyrus of
cially the hippocampus.115−117 In general, the likelihood the parietal lobe. They also concluded that these find-
of postoperative memory impairment depends on the ings correlated with previous fMRI localization of work-
amount of disease in mesial temporal structures to be ing memory. In another study, Axmacher et al.65 com-
resected, as well as the degree to which contralateral bined ECoG spectral analysis and fMRI results to provide
CHAPTER 12 ELECTROCORTICOGRAPHIC SPECTRAL ANALYSIS 159

evidence that the medial temporal lobe controlled ac- vasive than competing techniques for human functional
tivity within the inferior temporal cortex during visual mapping. An important consequence of this is that it
working memory. can only be done under limited clinical circumstances in
patients with lesions and/or seizure foci that could po-
tentially result in functional reorganization and/or alter
䉴 MAPPING FUNCTIONAL normal neurophysiological responses. A practical conse-
CONNECTIVITY quence of this is that epileptiform discharges may con-
taminate ECoG signals and produce inaccurate or unus-
Although clinical brain mapping has usually emphasized able data at specific electrodes that may be of interest.
the localization of function at individual cortical sites This can be addressed by excluding segments or whole
or collections of these sites, there is a growing interest channels of ECoG data contaminated by this activity, but
in and appreciation for the role of cortical networks in concerns about the effects of epilepsy on normal neu-
brain function. To this end, fMRI studies have used co- rophysiological responses can only be addressed by the
variations in BOLD signal to identify networks of brain reproducibility of ECoG responses across a variety of
sites that tend to be coactivated under different task con- subjects with different seizure foci and/or lesions, or by
ditions, including at rest (i.e., default networks).123−126 similar findings in normal populations using noninvasive
In contrast to fMRI, however, the temporal resolution techniques. Compared to high-density scalp EEG and
of electrophysiological measures, such as EEG, MEG, MEG, ECoG mapping offers significant improvements in
and ECoG, make them ideal for studying not only the signal-to-noise ratio and far greater sensitivity for high-
structure of these networks, that is, their anatomic com- frequency cortical activity.28
As the previous sections have indicated, high-
ponents, but also their event-related dynamics, that is,
frequency spectral responses represent an important
how their components interact on the time scale of func-
electrophysiological signature of cortical activation with
tional tasks. A variety of methods have been devised
excellent temporal and spatial resolution. Several recent
to study these interactions, including event-related co-
MEG studies have reported spectral responses that sig-
herence and partial directed coherence.127−129 One such
nificantly overlap the broadband gamma responses ob-
approach is based on the concept of Granger causality,
served in ECoG studies107–111 , but direct comparisons
whereby a causal influence of one site on another can
be inferred if the ECoG signal of one can be used to pre- with ECoG are still forthcoming. A recent recording
dict that of the other. An adaptation of this method called of both high-density EEG and ECoG in the same sub-
short-time directed transfer function (SDTF) can be used jects found that with some enhancements of record-
to estimate the magnitude, frequency, timing, and direc- ing techniques, scalp EEG is also capable of measuring
tionality of task-related changes in causal interactions be- high-gamma responses, albeit with lower sensitivity.39 In
tween ECoG recording sites.130,131 Variations of this ap- addition to its greater sensitivity to high-frequency ac-
tivity, artifacts from eye blinks and eye movements are
proach have been used to analyze scalp EEG recordings
greatly attenuated, albeit not eliminated, in ECoG sig-
during movement imagery,82 to analyze ECoG record-
nals. A recent study comparing artifacts in invasive and
ings during visually cued movement,132 and to analyze
noninvasive EEG estimated that the signal quality of in-
ECoG recordings during language processing.133 The
vasive EEG is 20 to above 100 times superior to that of
clinical relevance of these findings, obtained in a rel-
simultaneously recorded noninvasive EEG. fMRI is also
atively small number of subjects to date, is yet to be
clearly less invasive than ECoG, and its clinical utility in
determined. However, these and other studies like them
patients undergoing epilepsy surgery has been studied
illustrate the potential of ECoG (and other electrophysi-
extensively, both for the purpose of lateralizing lan-
ological measures) not only to identify the regions of the
guage function134,135 and for localizing language func-
brain responsible for processing different types of infor-
tion within the language dominant hemisphere.136−138
mation, but also better define the functional role of these
Although the fMRI scanning environment can limit the
regions by estimating the temporal sequence by which
they are activated and the direction of information flow functional tasks that can be performed during evalua-
tion, including motor tasks, and can be prohibitive for
among them.
claustrophobic patients, its noninvasive nature and its
growing availability are leading to more widespread ac-
ceptance. However, like ECoG and other electrophysio-
䉴 STRENGTHS AND WEAKNESSES logical measures, fMRI relies on measures of functional
RELATIVE TO OTHER MAPPING activation, and care must be taken when using it to pre-
TECHNIQUES dict the effect of surgical resection, that is, lesioning. The
main advantage of ECoG (and other electrophysiological
With the exception of ESM, which also relies on surgi- techniques) over fMRI is their superior temporal resolu-
cally implanted intracranial electrodes, ECoG is more in- tion. The fMRI BOLD response is an indirect and delayed
160 SECTION I TECHNIQUES

measure of synaptic activity that necessarily integrates of the safety of resecting any single site. Nevertheless,
activity over time scales of seconds.30 At first blush, this there is mounting evidence that ECoG mapping can be
may not seem like an important consideration for clini- used to complement ESM and other mapping modali-
cal functional mapping, but the latencies and sequences ties. Furthermore, recent studies have shown that ECoG
by which different cortical regions are activated during functional mapping can potentially be done in real- or
a functional task can give important clues as to their re- near real-time.
spective functional roles and could, in theory, allow finer An important illustrative example of the clinical ap-
functional–anatomical segregation. Whether this added plication for ECoG functional mapping was provided
information could be used to make better predictions of recently by Lachaux et al.141 The authors displayed real-
the functional impact of surgical resection remains to be time spectral power changes during intracranial lan-
seen. guage mapping of two epilepsy patients with implanted
Because ESM can induce a brief, reversible, and depth electrodes for visualization by the patients and ex-
reproducible disruption of cortical function, in essence perimenters on a computer monitor. During evaluation
mimicking the effects of a surgical resection, it remains of one of the patients, investigators were able to iden-
the gold standard for localizing language function prior tify high-gamma power changes in the middle portion
to resective surgery. However, this procedure can in- of the upper left superior temporal sulcus that occurred
flict pain through the stimulation of trigeminal afferents during spontaneous conversation and performed addi-
in cortical blood vessels and can induce afterdischarges tional language testing to further isolate the functional
and seizures. Time constraints during intraoperative ESM characteristics of this region based on their initial ob-
can prohibit comprehensive mapping of language cor- servations. There were no measurable changes in high-
tex with multiple language tasks. Although more time is gamma activity when the patient was speaking, reading
available for ESM when it is done extraoperatively with aloud, or listening to a loud noise. The high-gamma
implanted electrodes, comprehensive language mapping power changes were seen only when the experimenter
can still be quite time consuming, particularly with a bat- spoke, and increased more in response to sentences spo-
tery of language tasks, since each task must be repeated ken in the patient’s primary language than in languages
at each pair of electrodes, ideally after determining the in which the patient was not fluent. These findings sug-
optimal stimulation intensity at each site. At times, re- gested not only that the region being evaluated was
peated comparisons may produce equivocal results, re- functionally responsible for phonological processing of
quiring even further testing. spoken language, but also that the spectral changes
There is no possibility of provoking pain or seizure could be detected in real-time and the testing paradigm
activity with ECoG mapping since it relies on passive adapted during a single session to obtain a more precise
recordings and does not require cortical stimulation. In functional evaluation.
addition, the entire electrode array can be recorded si- In the same patient, the ECoG mapping results were
multaneously during each functional task. Although av- compared to traditional ESM findings. Initially, no lan-
eraging in the time and/or frequency domains requires guage impairment had been demonstrated in the region
more trials of each task than ESM, a battery of language of interest during ESM. On review of the ESM language
tasks can be recorded in 1–2 hours, yielding compre- tasks performed, the investigators discovered that the
hensive functional maps of the entire recording array in patient had been asked to count aloud or to listen to
much less time than ESM. Recent advances in software an examiner count, but had not been evaluated dur-
for ECoG signal analysis have made it possible to obtain ing any sentence comprehension tasks. Because of the
results from ECoG almost instantaneously.68,139−141 positive ECoG language mapping results, a sentence
comprehension task was performed with ESM and also
demonstrated interference with language comprehen-
sion during cortical stimulation. Had the ECoG language
䉴 EXAMPLES IN CLINICAL mapping not been performed, the treating neurosurgeon
PRACTICE might have resected this portion of the left superior
temporal sulcus on the basis of negative ESM, possibly
Functional cortical mapping with ECoG spectral analysis resulting in a profound language deficit in sentence com-
is being used with increasing frequency during surgical prehension.
planning prior to resection of epileptic foci and/or struc- Miller et al.140 also demonstrated that ECoG high-
tural lesions such as tumors or vascular malformations. gamma power augmentation is robust enough to be de-
ECoG mapping is safer than ESM and is potentially faster tected during single trials, in this case, of a motor task
since it can be done at all electrode sites at the same such as a handshake. More recently, Brunner et al.68 de-
time. However, its positive predictive value for surgi- scribed a new ECoG mapping procedure that creates a
cal outcomes has not been fully evaluated, and at least functional cortical map in only a few minutes with a false
for now it should not be used as the sole determinant positive rate of 0.46% and 1.10% for hand and tongue
CHAPTER 12 ELECTROCORTICOGRAPHIC SPECTRAL ANALYSIS 161

maps, respectively, compared to ESM, and no false nega- ronment, thinking, planning, etc. Experimental control
tives. These findings indicate that ECoG functional map- of the patient’s attention is, therefore, paramount. The
ping can be used in near real-time to investigate func- task itself should be relatively brief and simple so that
tional cortical activation with much higher precision than the timing of task-related cortical processing can be eas-
previously found. ily inferred from the timing of stimuli and/or responses.
ECoG spectral changes have also been investigated Event-related designs are useful for this purpose and for
for their utility in controlling BCI. Many neuromuscular approximating on a trial-by-trial basis a “resting base-
diseases such as amyotrophic lateral sclerosis, muscular line”, with which task-related ECoG temporal segments
dystrophy, neuromuscular junction diseases, as well as can be compared. Proper task design should be used to
spinal cord trauma and brainstem lesions, leave patients avoid contamination of the intertrial baseline with extra-
with a loss of voluntary motor control but with rela- neous speech or noise, particularly when employing au-
tively preserved cortical motor and premotor functions. ditory stimuli. Efforts should be made to keep the testing
Tai et al. reviewed a series of articles addressing the po- room, usually the patient’s hospital room, as free from
tential use of BCI for patients with locked-in syndrome, unnecessary distractions as possible. These efforts may
including BCI using electrocorticography.142 Leuthardt include sound treatments to reduce noise from hallway
et al.143 demonstrated the use of ECoG signal detection traffic and next-door neighbors, signs on the door noti-
in controlling a one-dimensional cursor through real and fying visitors to wait until testing is finished, and simple
imagined motor and speech tasks in four patients under- measures like unplugging the phone and turning off the
going intracranial electrode implantation in preparation television.
for epilepsy surgery. Additional studies have shown that During testing, the patient should be continuously
patients can utilize ECoG signal changes during auditory monitored for their level of arousal and attentiveness to
as well as motor imagery to accurately control the move- the task. Delayed or missing responses can be a sign of
ment of a cursor.139,144 Ramsey et al.67 observed gamma fatigue and should be excluded from subsequent signal
power increases within the dorsolateral prefrontal cortex analyses. Video recordings synchronized with ECoG and
during mental calculation and proposed that this mea- test markers are useful for post-hoc review to exclude
surable change could also be used as an input for BCI trials with experimental artifacts or abnormal ECoG activ-
applications. ity. During testing, the patient should be encouraged to
Finally, studies investigating the treatment of in- notify the testing staff if they have difficulty with testing
tractable pain with motor cortex stimulation have and/or need to take a break. Because ECoG mapping,
demonstrated the potential safety of long-term intracra- like ESM, is sensitive to the patient’s level of coopera-
nial electrode implantation.145,146 Combining these re- tion, every effort should be made to ensure that the pa-
sults, it may be feasible to implant subdural electrodes tient is comfortable. Patients vary considerably in how
over the cortex for the sole purpose of detecting ECoG quickly they recover from the surgery to implant intracra-
spectral indices of cortical activation associated with in- nial electrodes. Some patients are ready for testing the
tent to move that can in turn be translated into the use next day, while others have persistent headache, nausea,
of a prosthetic device. fatigue, and/or somnolence that prevent effective testing
for several days. Notwithstanding concerns about bleed-
ing in the immediate perioperative period, nonsteroidal
䉴 PEARLS AND PITFALLS anti-inflammatory analgesics such as Toradol are usually
more effective than opiates for postcraniotomy pain and
ECoG spectral analysis is still an emerging technique are less likely to cause excessive sedation, nausea, and
for functional mapping, and its clinical applications are constipation, or to suppress the patient’s appetite. These
still evolving, albeit rapidly. There remains much to be medications should be withdrawn prior to surgical resec-
learned about the neural mechanisms underlying the dif- tion to avoid increased bleeding risk.
ferent event-related spectral changes that have been ob- The technical specifications for data acquisition dur-
served during functional brain activation, and about how ing ECoG spectral mapping are somewhat more rig-
they relate to behavioral and experimental variables, as orous than are customary for the clinical purposes of
well as to other measures of neural function. It is, there- capturing interictal and ictal ECoG activity. Although
fore, important to take care with experimental design there is growing interest in high-frequency epilepto-
and data acquisition, and yet keep an open mind with genic activity,147,148 most commercially available long-
respect to data analysis and interpretation. term video–EEG monitoring systems have not been
The design of experiments for both research and optimized for the high-gamma activity that appears to be
clinical testing must always take into account that ECoG, so useful for ECoG spectral mapping. In addition to ad-
like EEG, MEG, fMRI, and PET, is a measure of functional equate sampling rates (preferably greater than 1000 Hz)
brain activation. When not engaged in an experimental and appropriate anti-aliasing filter settings (preferably
task, the human brain is still actively sensing the envi- greater than 300 Hz), it is critical, as with ERPs, to
162 SECTION I TECHNIQUES

accurately record experimental markers indicating the terpretation of its results should continue to take into
timing of experimental stimuli and/or responses rela- account the potential limitations of this new technology.
tive to the recorded ECoG. For this purpose, it is prefer- There is still much to be done to demonstrate the positive
able to record the markers directly into the ECoG data predictive value of ECoG with respect to surgical out-
stream and to avoid relying on complicated synchro- comes, and until then it is important to continue to com-
nization schemes or the promises of software vendors. pare ECoG functional maps with those derived from ESM
The markers themselves should be as direct a measure and other established and emerging techniques. When-
of events as possible. For example, instead of relying ever possible, clinical decisions regarding the extent of
solely on markers for the onset of visual stimuli based resection should be based on consistent findings across
on Transistor-Transistor Logic (TTL) pulses, the timing of multiple independent lines of evidence, always taking
which may depend on the computer operating system into consideration the relative strengths and weaknesses
and the experimental software, one can use a photodi- of the techniques that produced them.
ode to measure the luminance of a patch that is obscured
from view in a corner of the monitor and that changes
from black to white at stimulus onset.
When approaching analysis and interpretation of
REFERENCES
ECoG data, there are a plethora of choices to be made.
1. Lesser RP, Lüders H, Klem G, et al. Extraoperative corti-
Whenever possible, however, it is best to avoid precon-
cal functional localization in patients with epilepsy. J Clin
ceptions about what aspect of the ECoG signal is relevant Neurophysiol 1987;4(1):27-53.
to brain function. Although there is growing evidence 2. Adrian ED, Matthews BHC. The Berger rhythm: po-
supporting broadband high gamma as an important in- tential changes from the occipital lobes in man. Brain
dex of cortical processing, research continues on its re- 1934;57:355-385.
sponse properties and generating mechanisms, as well 3. Jasper HH, Andrews HL. Electro-encephalography. III.
as those of other spectral phenomena such as band- Normal differentiation of occipital and precentral regions
limited gamma responses and ERD/ERS in lower fre- in man. Arch Neurol Psychiatry 1938;39:96-115.
quencies. This research is driven in part by the exciting 4. Gastaut H. Etude electrocorticographique de la reactivite
prospect that different spectral changes in the ECoG sig- des rythmes rolandiques. Rev Neurol (Paris) 1952;87:176-
nal may reflect different populations of neurons and/or 182.
5. Chatrian GE, Petersen MC, Lazarte JA. The blocking of
different levels of neural integration across cortical net-
the rolandic wicket rhythm and some central changes re-
works, possibly even reflecting different modes of in- lated to movement. Electroencephalogr Clin Neurophysiol
formation processing. It is, therefore, prudent to keep 1959;11:497-510.
an open mind when analyzing and interpreting experi- 6. Pfurtscheller G, Aranibar A. Evaluation of event-related
mental ECoG data, particularly for research purposes. A desynchronization (ERD) preceding and following volun-
common pitfall in spectral analysis of EEG or ECoG is tary self-paced movement. Electroencephalogr Clin Neu-
to choose, a priori, particular frequency bands for anal- rophysiol 1979;46:138-146.
ysis of power changes. Because of the variability across 7. Pfurtscheller G, Lopes da Silva FH. Event-related
individuals, experiments, and studies in the frequency EEG/MEG synchronization and desynchronization: basic
bands that are optimally reactive to functional activation, principles. Clin Neurophysiol 1999;110(11):1842-1857.
this approach may lead to suboptimal or even uninter- 8. Jung J, Mainy N, Kahane P, et al. The neural bases of at-
tentive reading. Hum Brain Mapp 2008;29(10):1193-1206.
pretable event-related spectral analyses. Instead, prelim-
9. Pfurtscheller G. Functional topography during sensorimo-
inary exploratory analyses should be considered using tor activation studied with event-related desynchroniza-
time–frequency analyses of event-related power changes tion mapping. J Clin Neurophysiol 1989;6:75-84.
in all frequencies at all task-relevant times. Subsequent 10. Pfurtscheller G, Berghold A. Patterns of cortical activa-
analyses can then focus on bands empirically derived tion during planning of voluntary movement. Electroen-
from these exploratory analyses. On the other hand, cephalogr Clin Neurophysiol 1989;72:250-258.
because algorithms for the aforementioned exploratory 11. Pfurtscheller G, Flotzinger D, Neuper C. Differentiation
analyses can be computationally intensive and statis- between finger, toe and tongue movement in man based
tically challenging, analyses for clinical mapping may on 40 Hz EEG. Electroencephalogr Clin Neurophysiol
require pragmatic choices to be made. One of the ad- 1994;90:456-460.
vantages of using broadband high-gamma responses for 12. Pfurtscheller G, Stancak A Jr, Neuper C. Post-movement
beta synchronization. A correlate of an idling motor area?
functional mapping, as illustrated in the previous sec-
Electroencephalogr Clin Neurophysiol 1996;98(4):281-
tion, is that there is a greater tolerance for choosing 293.
the frequency range for analysis as long as one avoids 13. Crone NE, Miglioretti DL, Gordon B, et al. Functional
power suppression (ERD) in lower frequencies, which mapping of human sensorimotor cortex with electrocor-
can, however, range as high as 40–50 Hz. ticographic spectral analysis. I. Alpha and beta event-
Finally, because ECoG spectral mapping remains in related desynchronization. Brain 1998;121(Pt 12):2271-
the early stage of scientific and clinical investigation, in- 2299.
CHAPTER 12 ELECTROCORTICOGRAPHIC SPECTRAL ANALYSIS 163

14. Wheaton LA, Carpenter M, Mizelle JC, et al. Prepara- 31. Kalcher J, Pfurtscheller G. Discrimination between phase-
tory band specific premotor cortical activity differenti- locked and non-phase-locked event-related EEG ac-
ates upper and lower extremity movement. Exp Brain Res tivity. Electroencephalogr Clin Neurophysiol 1995;94:
2008;184(1):121-126. 381-384.
15. Krause CM, Lang HA, Laine M, et al. Event-related desyn- 32. Crone NE, Boatman D, Gordon B, et al. Induced electro-
chronization evoked by auditory stimuli. Brain Topogr corticographic gamma activity during auditory perception.
1994;7(2):107-112. Clin Neurophysiol 2001;112(4):565-582.
16. Krause CM, Lang H, Laine M, et al. Cortical processing 33. Trautner P, Rosburg T, Dietl T, et al. Sensory gating
of vowels and tones as measured by event-related desyn- of auditory evoked and induced gamma band activity
chronization. Brain Topogr 1995;8(1):47-56. in intracranial recordings. Neuroimage 2006;32(2):790-
17. Krause CM, Astrom T, Karrasch M, et al. Cortical activa- 798.
tion related to auditory semantic matching of concrete 34. Pfurtscheller G. Graphical display and statistical evalua-
versus abstract words [In Process Citation]. Clin Neuro- tion of event-related desynchronization (ERD). Electroen-
physiol 1999;110(8):1371-1377. cephalogr Clin Neurophysiol 1977;43:757-760.
18. Krause CM. Cognition- and memory-related ERD/ERS re- 35. Lehtela L, Salmelin R, Hari R. Evidence for reactive mag-
sponses in the auditory stimulus modality. Prog Brain Res netic 10-Hz rhythm in the human auditory cortex. Neu-
2006;159:197-207. rosci Lett 1997;222(2):111-114.
19. Pfurtscheller G. [Spontaneous and evoked EEG-activity 36. Stancak A Jr, Pfurtscheller G. Desynchronization and
during one-second light- stimulation (author’s transl)]. recovery of beta rhythms during brisk and slow self-
[German]. EEG-EMG Zeitschrift fur Elektroenzephalo- paced finger movements in man. Neurosci Lett 1995;196:
graphie Elektromyographie und Verwandte Gebiete 21-24.
1977;8:145-154. 37. Neuper C, Pfurtscheller G. Post-movement synchroniza-
20. Aranibar A, Pfurtscheller G. On and off effects in the back- tion of beta rhythms in the EEG over the cortical foot
ground EEG activity during one- second photic stimula- area in man. Neurosci Lett 1996;216(1):17-20.
tion. Electroencephalogr Clin Neurophysiol 1978;44:307- 38. Pfurtscheller G, Stancak A Jr, Neuper C. Event-related syn-
316. chronization (ERS) in the alpha band–an electrophysio-
21. Pfurtscheller G, Klimesch W. Functional topography dur- logical correlate of cortical idling: a review. [Review] [47
ing a visuoverbal judgment task studied with event- refs]. Int J Psychophysiol 1996;24:39-46.
related desynchronization mapping. J Clin Neurophysiol 39. Ball T, Demandt E, Mutschler I, et al. Movement related
1992;9(1):120-131. activity in the high gamma range of the human EEG. Neu-
22. Pfurtscheller G, Neuper C, Mohl W. Event-related desyn- roimage 2008;41(2):302-310.
chronization (ERD) during visual processing. Int J Psy- 40. Cooper R, Winter AL, Crow HJ, et al. Comparison of sub-
chophysiol 1994;16:147-153. cortical, cortical and scalp activity using chronically in-
23. Karrasch M, Krause CM, Laine M, et al. Event-related dwelling electrodes in man. Electroencephalogr Clin Neu-
desynchronization and synchronization during an audi- rophysiol 1965;18:217-228.
tory lexical matching task. Electroencephalogr Clin Neu- 41. Pfurtscheller G, Cooper, R. Frequency dependence of the
rophysiol 1998;107(2):112-121. transmission of the EEG from cortex to scalp. Electroen-
24. Krause CM, Porn B, Lang AH, et al. Relative alpha desyn- cephalogr Clin Neurophysiol 1975;38:93-96.
chronization and synchronization during speech percep- 42. Pfurtscheller G, Stancak AJ, Neuper C. Post-movement
tion. Brain Res Cogn Brain Res 1997;5(4):295-299. beta synchronization. A correlate of an idling motor area?
25. Krause CM, Gronholm P, Leinonen A, et al. Modality mat- Electroencephalogr Clin Neurophysiol 1996;98:281-293.
ters: the effects of stimulus modality on the 4- to 30-Hz 43. Crone NE, Korzeniewska A, Sinai A, et al. ERD/ERS
brain electric oscillations during a lexical decision task. and ERPs. In Event-Related Potentials in Patients with
Brain Res 2006;1110(1):182-192. Epilepsy: from Current State to Future Prospects, edited
26. Bastiaansen MC, van der Linden M, Ter Keurs M, et al. by A Ikeda and Y Inoue. Montrouge: Libbey, 2008,
Theta responses are involved in lexical-semantic retrieval pp. 233-246.
during language processing. J Cogn Neurosci 2005;17(3): 44. Lachaux JP, Jung J, Mainy, N, et al. Silence is golden: tran-
530-541. sient neural deactivation in the prefrontal cortex during
27. Grabner RH, Brunner C, Leeb R, et al. Event-related EEG attentive reading. Cereb Cortex 2008;18(2):443-450.
theta and alpha band oscillatory responses during lan- 45. Asano E, Juhasz C, Shah A, et al. Role of subdural electro-
guage translation. Brain Res Bull 2007;72(1):57-65. corticography in prediction of long-term seizure outcome
28. Ball T, Kern M, Mutschler I, et al. Signal quality of si- in epilepsy surgery. Brain 2009;132(Pt 4):1038-1047.
multaneously recorded invasive and non-invasive EEG. 46. Brown EC, Rothermel R, Nishida M, et al. In vivo ani-
Neuroimage 2009;46(3):708-716. mation of auditory-language-induced gamma-oscillations
29. Crone NE, Miglioretti DL, Gordon B, et al. Functional in children with intractable focal epilepsy. Neuroimage
mapping of human sensorimotor cortex with electrocor- 2008;41(3):1120-1131.
ticographic spectral analysis. II. Event-related synchro- 47. Szurhaj W, Bourriez JL, Kahane P, et al. Intracerebral study
nization in the gamma band. Brain 1998;121(Pt 12):2301- of gamma rhythm reactivity in the sensorimotor cortex.
2315. Eur J Neurosci 2005;21(5):1223-1235.
30. Crone NE, Sinai AS, Korzeniewska, A. High-frequency 48. Lachaux JP, Rudrauf D, Kahane P. Intracranial EEG and
gamma oscillations and human brain mapping with elec- human brain mapping. J Physiol (Paris) 2003;97(4–6):613-
trocorticography. Prog Brain Res 2006;159:279-302. 628.
164 SECTION I TECHNIQUES

49. Nilsson J, Panizza M, Hallett M. Principles of digital frequency dynamics of cortical activity. Neuroimage
sampling of a physiologic signal. [Review]. Electroen- 2008;40(4):1686-1700.
cephalogr Clin Neurophysiol 1993;89:349-358. 67. Ramsey NF, van de Heuvel MP, Kho KH, et al. Towards
50. Shannon CE. Communication in the presence of noise. human BCI applications based on cognitive brain systems:
Proc IEEE 1998;86(2):447-457. an investigation of neural signals recorded from the dor-
51. Pfurtscheller G. Mapping of event-related desynchroniza- solateral prefrontal cortex. IEEE Trans Neural Syst Rehabil
tion and type of derivation. Electroencephalography and Eng 2006;14(2):214-217.
clinical Neurophysiol Suppl 1988;70:190-193. 68. Brunner P, Ritaccio AL, Lynch TM, et al. A practical proce-
52. Pfurtscheller G, Aranibar A. Event-related cortical desyn- dure for real-time functional mapping of eloquent cortex
chronization detected by power measurements of scalp using electrocorticographic signals in humans. Epilepsy
EEG. Electroencephalogr Clin Neurophysiol 1977;42:817- Behav 2009;15(3):278-286.
826. 69. Sinai A, Bowers CW, Crainiceanu CM, et al. Electrocortico-
53. Mallat S. A Wavelet Tour of Signal Processing, 2nd edition. graphic high gamma activity versus electrical cortical stim-
New York: Academic Press, 1998. ulation mapping of naming. Brain 2005;128(Pt 7):1556-
54. Mallat S, Zhang Z. Matching pursuit with time-frequency 1570.
dictionaries. IEEE Trans Signal Process 1993;41:3397-3415. 70. Sinai A, Crone NE, Wied HM, et al. Intracranial mapping of
55. Durka PJ, Blinowska KJ. Analysis of EEG transients auditory perception: event-related responses and electro-
by means of matching pursuit. Ann Biomed Eng 1995; cortical stimulation. Clin Neurophysiol 2009;120(1):140-
23(5):608-611. 149.
56. Durka PJ, Ircha D, Blinowska KJ. Stochastic time- 71. Lachaux JP, George N, Tallon-Baudry C, et al. The many
frequency dictionaries for matching pursuit. IEEE Trans faces of the gamma band response to complex visual stim-
Signal Process 2001;49(3):507-510. uli. Neuroimage 2005;25(2):491-501.
57. Durka PJ. Time-frequency microstructure and statistical 72. Tallon-Baudry C, Bertrand O, Henaff MA, et al. Atten-
significance of ERD and ERS. In Event-Related Dynamics tion modulates gamma-band oscillations differently in the
of Brain Oscillations, edited by C Neuper and W Klimesch. human lateral occipital cortex and fusiform gyrus. Cereb
Oxford: Elsevier, 2006, pp. 121-133. Cortex 2005;15(5):654-662.
58. Ray S, Jouny CC, Crone NE, et al. Human ECoG analysis 73. Hoogenboom N, Schoffelen JM, Oostenveld R, et al.
during speech perception using matching pursuit: a com- Localizing human visual gamma-band activity in fre-
parison between stochastic and dyadic dictionaries. IEEE quency, time and space. Neuroimage 2006;29(3):764-
Trans Biomed Eng 2003;50(12):1371-1373. 773.
59. Zygierewicz J, Durka PJ, Klekowicz H, et al. Compu- 74. Asano E, Nishida M, Fukuda M, et al. Differential visually-
tationally efficient approaches to calculating significant induced gamma-oscillations in human cerebral cortex.
ERD/ERS changes in the time–frequency plane. J Neurosci Neuroimage 2009;45(2):477-489.
Methods 2005;145(1–2):267-276. 75. Edwards E, Soltani M, Deouell LY, et al. High
60. Sutherling WW, Levesque MF, Crandall PH, et al. Localiza- gamma activity in response to deviant auditory stimuli
tion of partial epilepsy using magnetic and electric mea- recorded directly from human cortex. J Neurophysiol
surements. Epilepsia 1991;32(Suppl 5):S29-S40. 2005;94(6):4269-4280.
61. Nunez PL, Silberstein RB. On the relationship of synap- 76. Kaiser J, Hertrich I, Ackennann H, et al. Gamma-
tic activity to macroscopic measurements: does co- band activity over early sensory areas predicts detection
registration of EEG with fMRI make sense? Brain Topogr of changes in audiovisual speech stimuli. Neuroimage
2000;13(2):79-96. 2006;30(4):1376-1382.
62. Foucher JR, Otzenberger H, Gounot D. The BOLD re- 77. Rektor I, Sochurkova D, Bockova M. Intracerebral
sponse and the gamma oscillations respond differently ERD/ERS in voluntary movement and in cognitive visuo-
than evoked potentials: an interleaved EEG-fMRI study. motor task. In Event-Related Dynamics of Brain Oscil-
BMC Neurosci 2003;4(1):22. lations, edited by C Neuper and W Klimesch. Oxford:
63. Babiloni F, Cincotti F, Babiloni C, et al. Estimation of the Elsevier, 2006, pp. 311-330.
cortical functional connectivity with the multimodal inte- 78. Ohara S, Ikeda A, Kunieda T, et al. Movement-related
gration of high-resolution EEG and fMRI data by directed change of electrocorticographic activity in human sup-
transfer function. Neuroimage 2005;24(1):118-131. plementary motor area proper. Brain 2000;123(Pt 6):1203-
64. Lachaux JP, Fonlupt P, Kahane P, et al. Relationship be- 1215.
tween task-related gamma oscillations and BOLD signal: 79. Brovelli A, Lachaux JP, Kahane P, et al. High gamma
new insights from combined fMRI and intracranial EEG. frequency oscillatory activity dissociates attention from
Hum Brain Mapp 2007;28(12):1368-1375. intention in the human premotor cortex. Neuroimage
65. Axmacher N, Schmitz DP, Wagner T, et al. Interactions 2005;28(1):154-164.
between medial temporal lobe, prefrontal cortex, and in- 80. Leuthardt EC, Schalk G, Wolpaw JR, et al. A brain-
ferior temporal regions during visual working memory: computer interface using electrocorticographic signals in
a combined intracranial EEG and functional magnetic humans. J Neural Eng 2004;1(2):63-71.
resonance imaging study. J Neurosci 2008;28(29):7304- 81. Leuthardt EC, Miller K, Anderson NR, et al. Electrocortico-
7312. graphic frequency alteration mapping: a clinical technique
66. Dalal SS, Guggisberg AG, Edwards E, et al. Five- for mapping the motor cortex. Neurosurgery 2007;60(4
dimensional neuroimaging: localization of the time- Suppl 2):260-270; discussion 270-271.
CHAPTER 12 ELECTROCORTICOGRAPHIC SPECTRAL ANALYSIS 165

82. Ginter J Jr, Blinowska KJ, Kaminski M, et al. Propaga- 100. Baumgartner C, Doppelbauer A, Deecke L, et al. Neu-
tion of EEG activity in the beta and gamma band dur- romagnetic investigation of somatotopy of human hand
ing movement imagery in humans. Methods Inf Med somatosensory cortex. Exp Brain Res 1991;8:641-648.
2005;44(1):106-113. 101. Allison T, McCarthy G, Wood CC, et al. Potentials evoked
83. Szurhaj W, Derambure P. Intracerebral study of gamma in human and monkey cerebral cortex by stimulation
oscillations in the human sensorimotor cortex. In Event- of the median nerve. A review of scalp and intracranial
Related Dynamics of Brain Oscillations, edited by C Neu- recordings. Brain 1991;114(Pt 6):2465-2503.
per and W Klimesch. Oxford: Elsevier, 2006, pp. 297-310. 102. Baumgartner U, Vogel H, Ellrich J, et al. Brain electri-
84. Gonzalez SL, de Peralta RG, Thut G, et al. Very high fre- cal source analysis of primary cortical components of
quency oscillations (VHFO) as a predictor of movement the tibial nerve somatosensory evoked potential using
intentions. Neuroimage 2006;32(1):170-179. regional sources. Electroencephalogr Clin Neurophysiol
85. Miller KJ, Leuthardt EC, Schalk G, et al. Spectral changes 1998;108(6):588-599.
in cortical surface potentials during motor movement. J 103. Aoki F, Fetz EE, Shupe L, et al. Increased gamma-range ac-
Neurosci 2007;27(9):2424-2432. tivity in human sensorimotor cortex during performance
86. Baumgartner C, Barth DS, Levesque MF, et al. Human of visuomotor tasks. Clin Neurophysiol 1999;110(3):524-
hand and lip sensorimotor cortex as studied on elec- 537.
trocorticography. Electroencephalogr Clin Neurophysiol 104. Pfurtscheller G, Graimann B, Huggins JE, et al. Spatiotem-
1992;84(2):115-126. poral patterns of beta desynchronization and gamma
87. Hart J Jr, Crone NE, Lesser RP, et al. Temporal dynamics synchronization in corticographic data during self-paced
of verbal object comprehension. Proc Natl Acad Sci USA movement. Clin Neurophysiol 2003;114(7):1226-1236.
1998;95(11):6498-6503. 105. Toro C, Deuschl G, Thatcher R, et al. Event-related desyn-
88. Crone NE, Hao L, Hart J Jr, et al. Electrocorticographic chronization and movement-related cortical potentials on
gamma activity during word production in spoken and the ECoG and EEG. Electroencephalogr Clin Neurophys-
sign language. Neurology 2001;57(11):2045-2053. iol 1994;93:380-389.
89. Tanji K, Suzuki K, Delorme A, et al. High-frequency 106. Reddy CG, Reddy GG, Kawasaki H, et al. Decod-
gamma-band activity in the basal temporal cortex dur- ing movement-related cortical potentials from electrocor-
ing picture-naming and lexical-decision tasks. J Neurosci ticography. Neurosurg Focus 2009;27(1):E11.
2005;25(13):3287-3293. 107. Gross J, Schnitzler A, Timmermann L, et al. Gamma os-
90. Towle VL, Yoon HA, Castelle M, et al. ECoG gamma activ- cillations in human primary somatosensory cortex reflect
ity during a language task: differentiating expressive and pain perception. PLoS Biol 2007;5(5):1168-1173.
receptive speech areas. Brain 2008;131(Pt 8):2013-2027. 108. Ihara A, Hirata M, Yanagihara K, et al. Neuromagnetic
91. Mainy N, Jung J, Baciu M, et al. Cortical dynamics of word gamma-band activity in the primary and secondary so-
recognition. Hum Brain Mapp 2008;29(11):1215-1230. matosensory areas. Neuroreport 2003;14(2):273-277.
92. Howard MW, Rizzuto DS, Caplan JB, et al. Gamma oscil- 109. Hauck, M, Lorenz J, Engel, AK. Attention to painful
lations correlate with working memory load in humans. stimulation enhances gamma-band activity and syn-
Cereb Cortex 2003;13(12):1369-1374. chronization in human sensorimotor cortex. J Neurosci
93. Sederberg PB, Kahana MJ, Howard MW, et al. Theta and 2007;27(35):9270-9277.
gamma oscillations during encoding predict subsequent 110. Bauer M, Oostenveld R, Peeters M, et al. Tactile spatial
recall. J Neurosci 2003;23(34):10809-10814. attention enhances gamma-band activity in somatosen-
94. Mormann F, Fell J, Axmacher N, et al. Phase/amplitude re- sory cortex and reduces low-frequency activity in parieto-
set and theta-gamma interaction in the human medial tem- occipital areas. J Neurosci 2006;26(2):490-501.
poral lobe during a continuous word recognition memory 111. Vidal JR, Chaumon M, O’Regan JK, et al. Visual group-
task. Hippocampus 2005;15(7):890-900. ing and the focusing of attention induce gamma-band
95. Sederberg PB, Schulze-Bonhage A, Madsen JR, et al. oscillations at different frequencies in human magnetoen-
Gamma oscillations distinguish true from false memories. cephalogram signals. J Cogn Neurosci 2006;18(11):1850-
Psychol Sci 2007;18(11):927-932. 1862.
96. Sederberg PB, Schulze-Bonhage A, Madsen JR, et 112. Mainy N, Kahane P, Minotti L, et al. Neural correlates
al. Hippocampal and neocortical gamma oscillations of consolidation in working memory. Hum Brain Mapp
predict memory formation in humans. Cereb Cortex 2007;28(3):183-193.
2007;17(5):1190-1196. 113. Hamberger MJ. Cortical language mapping in epilepsy: a
97. Ray S, Niebur E, Hsiao SS, et al. High-frequency gamma critical review. Neuropsychol Rev 2007;17(4):477-489.
activity (80–150 Hz) is increased in human cortex during 114. Schevon CA, Carlson C, Zaroff CM, et al. Pediatric lan-
selective attention. Clin Neurophysiol 2008;119(1):116- guage mapping: sensitivity of neurostimulation and Wada
133. testing in epilepsy surgery. Epilepsia 2007;48(3):539-545.
98. Lueders H, Lesser RP, Hahn J, et al. Cortical somatosen- 115. Guerreiro C, Cendes F, Li LM, et al. Clinical patterns of
sory evoked potentials in response to hand stimulation. J patients with temporal lobe epilepsy and pure amygdalar
Neurosurg 1983;58:885-894. atrophy. Epilepsia 1999;40(4):453-461.
99. Baumgartner C, Barth DS, Levesque MF, et al. Functional 116. Glikmann-Johnston Y, Saling MM, Chen J, et al. Struc-
anatomy of human hand sensorimotor cortex from spa- tural and functional correlates of unilateral mesial tem-
tiotemporal analysis of electrocorticography. Electroen- poral lobe spatial memory impairment. Brain 2008;131(Pt
cephalogr Clin Neurophysiol 1991;78(1):56-65. 11):3006-3018.
166 SECTION I TECHNIQUES

117. Busch RM, Chapin JS, Umashankar G, et al. Poor presur- 132. Kaminski M, Liang H. Causal influence: advances in neu-
gical performance on both verbal and visual memory rosignal analysis. Crit Rev Biomed Eng 2005;33(4):347-
measures is associated with low risk for memory decline 430.
following left temporal lobectomy for intractable epilepsy. 133. Korzeniewska A, Crainiceanu CM, Kus R, et al. Dynamics
Epileptic Disord 2008;10(3):199-205. of event-related causality in brain electrical activity. Hum
118. Golby AJ, Poldrack RA, Illes J, et al. Memory lateralization Brain Mapp 2008;29(10):1170-1192.
in medial temporal lobe epilepsy assessed by functional 134. Gaillard WD, Balsamo L, Xu B, et al. fMRI language task
MRI. Epilepsia 2002;43(8):855-863. panel improves determination of language dominance.
119. Richardson MP, Strange BA, Thompson PJ, et al. Pre- Neurology 2004;63(8):1403-1408.
operative verbal memory fMRI predicts post-operative 135. Binder JR, Rao SM, Hammeke TA, et al. Lateralized human
memory decline after left temporal lobe resection. Brain brain language systems demonstrated by task subtrac-
2004;127(Pt 11):2419-2426. tion functional magnetic resonance imaging. Arch Neurol
120. Janszky J, Jokeit H, Kontopoulou K, et al. Functional MRI 1995;52:593-601.
predicts memory performance after right mesiotemporal 136. Binder JR, Frost JA, Hammeke TA, et al. Human brain lan-
epilepsy surgery. Epilepsia 2005;46(2):244-250. guage areas identified by functional magnetic resonance
121. Binder JR, Sabsevitz DS, Swanson SJ, et al. Use of imaging. J Neurosci 1997;17(1):353-362.
preoperative functional MRI to predict verbal memory 137. FitzGerald DB, Cosgrove GR, Ronner S, et al. Location of
decline after temporal lobe epilepsy surgery. Epilepsia language in the cortex: a comparison between functional
2008;49(8):1377-1394. MR imaging and electrocortical stimulation. Am J Neuro-
122. Powell HW, Richardson MP, Symms MR, et al. Preoper- radiol 1997;18(8):1529-1539.
ative fMRI predicts memory decline following anterior 138. Rutten GJ, Ramsey NF, van Rijen PC, et al. Development
temporal lobe resection. J Neurol Neurosurg Psychiatry of a functional magnetic resonance imaging protocol for
2008;79(6):686-693. intraoperative localization of critical temporoparietal lan-
123. Schicke T, Muckli L, Beer AL, et al. Tight covariation of guage areas. Ann Neurol 2002;51(3):350-360.
BOLD signal changes and slow ERPs in the parietal cortex 139. Felton EA, Wilson JA, Williams JC, et al. Electrocortico-
in a parametric spatial imagery task with haptic acquisi- graphically controlled brain-computer interfaces using
tion. Eur J Neurosci 2006;23(7):1910-1918. motor and sensory imagery in patients with temporary
124. Arthurs OJ, Donovan T, Spiegelhalter DJ, et al. In- subdural electrode implants. Report of four cases. J Neu-
tracortically distributed neurovascular coupling relation- rosurg 2007;106(3):495-500.
ships within and between human somatosensory cortices. 140. Miller KJ, denNijs M, Shenoy P, et al. Real-time functional
Cereb Cortex 2007;17(3):661-668. brain mapping using electrocorticography. Neuroimage
125. Sigman M, Jobert A, Lebihan D, et al. Parsing a sequence 2007;37(2):504-507.
of brain activations at psychological times using fMRI. 141. Lachaux JP, Jerbi K, Bertrand O, et al. A Blueprint for Real-
Neuroimage 2007;35(2):655-668. Time Functional Mapping via Human Intracranial Record-
126. Thompson WK, Siegle G. A stimulus-locked vector au- ings. PLoS ONE 2007;2(10):e1094.
toregressive model for slow event-related fMRI designs. 142. Tai K, Blain S, Chau T. A review of emerging access tech-
Neuroimage 2009;46(3):739-748. nologies for individuals with severe motor impairments.
127. Sameshima K, Baccala LA. Using partial directed coher- Assist Technol 2008;20(4):204–219; quiz 220-221.
ence to describe neuronal ensemble interactions. J Neu- 143. Leuthardt EC, Miller KJ, Schalk G, et al. Electrocortico-
rosci Methods 1999;94(1):93-103. graphy-based brain computer interface–the Seattle experi-
128. Baccala LA, Sameshima K. Partial directed coherence: a ence. IEEE Trans Neural Syst Rehabil Eng 2006;14(2):194-
new concept in neural structure determination. Biol Cy- 198.
bern 2001;84(6):463-474. 144. Wilson JA, Felton EA, Garell PC, et al. ECoG factors under-
129. Schelter B, Winterhalder M, Eichler M, et al. Testing for lying multimodal control of a brain-computer interface.
directed influences among neural signals using partial di- IEEE Trans Neural Syst Rehabil Eng 2006;14(2):246-250.
rected coherence. J Neurosci Methods 2006;152(1–2):210- 145. Osenbach RK. Motor cortex stimulation for intractable
219. pain. Neurosurg Focus 2006;21(6):E7.
130. Ding M, Bressler SL, Yang W, et al. Short-window spectral 146. Rasche D, Ruppolt M, Stippich C, et al. Motor cortex stim-
analysis of cortical event-related potentials by adaptive ulation for long-term relief of chronic neuropathic pain: a
multivariate autoregressive modeling: data preprocessing, 10 year experience. Pain 2006;121(1–2):43-52.
model validation, and variability assessment. Biol Cybern 147. Zijlmans M, Jacobs J, Zelmann R, et al. High frequency
2000;83(1):35-45. oscillations and seizure frequency in patients with focal
131. Korzeniewska A, Manczak M, Kaminski M, et al. Determi- epilepsy. Epilepsy Res 2009;85(2-3):287-292.
nation of information flow direction among brain struc- 148. Zijlmans M, Jacobs J, Zelmann R, et al. High-frequency os-
tures by a modified directed transfer function (dDTF) cillations mirror disease activity in patients with epilepsy.
method. J Neurosci Methods 2003;125(1–2):195-207. Neurology 2009;72(11):979-986.
Chapter 13
Pediatric Brain Mapping: Special
Considerations
Robert J. Bollo1 , Chad Carlson2 , Orrin Devinsky 2 , and Howard L. Weiner 2,3
1
Department of Neurosurgery, Baylor College of Medicine and Neurosurgical Service, Texas Children’s Hospital,
Houston, Texas
2
Comprehensive Epilepsy Center, Department of Neurology, New York University School of Medicine,
New York, New York
3
Department of Neurosurgery, Division of Pediatric Neurosurgery, New York University School of Medicine,
New York, New York

䉴 INTRODUCTION subcortical white matter tracts critical to motor, sen-


sory and language function is essential to safe resec-
BRAIN MAPPING IN THE tion of tumors and ictal foci near eloquent cortical
PEDIATRIC PATIENT areas.2,7−10 Functional reorganization in the context of
long-standing pathology (e.g., low-grade tumors or long-
Advancements in noninvasive imaging of the substruc- standing epilepsy) may be more frequent in the devel-
tures and function of the developing brain, including oping brain.2,11,12
new applications of magnetic resonance imaging (MRI)
and the development of other imaging modalities includ-
ing magnetoencephalography (MEG), positron emission SPECIAL CONSIDERATIONS
tomography (PET), and single photon emission com-
puted tomography (SPECT), have profoundly impacted Pediatric patients present unique challenges to brain
clinical neuroscience. Advanced MRI techniques are mapping. Immaturity and anxiety can lead to head
rapidly becoming a standard part of pediatric neurol- movement, lack of cooperation, or an inability to follow
ogy and neurosurgery, including functional MRI (fMRI), directions during prolonged testing.13−15 Very young,
diffusion-weighted imaging (DWI), perfusion-weighted autistic, and developmentally delayed children may not
imaging (PWI), and diffusion tensor imaging (DTI). The be candidates for current methods of fMRI or awake in-
ability to noninvasively map eloquent brain function traoperative mapping if they cannot comprehend or par-
(fMRI, MEG, PET), ictal foci (SPECT, MEG, PET), and ticipate in these tasks.15
white matter tracts (DTI), as well as the coregistration Functional loci are poorly consolidated in the de-
of multiple imaging modalities, has changed how we veloping brain, with diffuse anatomical representation
diagnose, evaluate, and manage epilepsy and brain tu- compared to adults. Direct cortical stimulation mapping
mors in children.1−3 These techniques help guide and of sensorimotor and language function is less sensitive in
in some cases replace direct mapping via electrocortical children younger than 10–12 years, independent of pa-
stimulation (ECS), which remains the gold standard. tient cooperation.16−19 The Wada (intracarotid amobar-
Advances in functional neuroimaging hold signif- bital) test for hemispheric dominance is also less sensi-
icant promise in the diagnosis and management of tive in children younger than 10 years.16 Wada testing is
other disorders, such as traumatic brain injury (TBI) and also complicated in children by limited cooperation.20,21
psychiatric diseases.4−5 These techniques can provide PET and SPECT involve the administration of a radioiso-
important insights into plasticity during brain develop- tope, limiting research in children.1 Short seizure dura-
ment.2,6 However, noninvasive brain mapping tech- tion (less than 1 minute), common in children, can make
niques have limitations, especially in children, and few ictal SPECT injections difficult and less informative.1
have been rigorously tested in the pediatric population.1
The most common clinical indication for brain map- SPECIFIC TECHNIQUES
ping in children is preoperative planning for the re-
section of a brain tumor or seizure focus. For exam- Clinical brain mapping techniques in children can be
ple, a detailed knowledge of the cortical regions and categorized by substrate (eloquent function, seizure

167
168 SECTION I TECHNIQUES

modality invasive test measured variable mapping substrate special considerations in children clinical applications in pediatrics references
MRI no
requires cooperative, still patient;
sensorimotor & language function, 2, 11, 22–41
sensitivity diminished by sedation
fMRI BOLD signal language lateralization & in children <7 year-old, language mapping surgical planning
(hemispheric dominance) requires active task paradigms

neonatal hypoxic-ischemic reperfusion injury,


DWI extrocellular water diffusion acute ischemia/infection none 42, 43, 44–49
ADEM, ictal focus localization (peri-ictal DWI)

DTI axonal water diffusion (large) white matter tracts none surgical planning TBI 48–52
tumor grading, differentiating tumor
PWI rCBV brain tumors, ischemic penumbra improved sensitivity in children recurrence from treatment response, 53, 54
stroke, TBI

MEG no
requires a cooperative, still patient;
sensotimotor & language function, sensitivity diminished by sedation
MSI magnetic source dipole ictal focus localization, surgical planning 23–88
ictal foci & in children under 6 years-old

PET no requires radioisotope

sensitivity diminished by sedation; tumor grading, targeting sterentactic biopsy,


FDG glucose metobolism brain tumors, ictal foci cannot differentiate ictal onset zones from differentiating tumor recurrence from 23, 33, 89–109
regions of rapid seizure propagation treatment response; identifying ictal foci

MET protein metobolism brain tumors, ictal foci none tumor grading, deliniating tumor boundaries 91, 97, 100–103
identifying ictal foci
FMZ GABA receptor density ictal foci none (more specific than FDG-PET), 90
especially crypotgenic cortical dysplasia
identifying ictal foci, especially among
AMT serotonin receptor density ictal foci none
children with multifocal pathelogy (e.g., TSC) 90, 105, 110

SPECT no requires radioisotope


no tracer uptake in many common pediatric differentiating treatment response from
MIBI cerebral perfusion brain tumors, relative perfusion deficits 91, 111–120
tumors recurrent neoplasm; Moya-Moya disease
ictal injection technically demanding
HMPAO cerebral perfusion ictal foci, relative perfusion deficits identifying ictal foci, Moya-Moya disease 106, 114, 121–125
sensitivty improved with SISCOM
Wada yes requires cerebral angiography
requires a cooperative patient;
10, 12, 16, 20–21, 24, 41, 89,
amobarbital language and memory function language and memory lateralization diminished sensitivity in children surgical planning
92, 126–129
<10 years-old

ECS yes requires open craniotomy


diminished sensitivity in children <5 years-
functional inhibition, sensorimotor & language function, old; risk of seizures, diminished sensitivity of 2, 7-9, 13–14, 16, 19, 130,
intraoperative ictal foci EEG under anesthesia, awake craniosomy for
(intraoperative) surgical planning
131–139
interictal EEG recording
language mapping poorly tolerated
diminished sensitivity in children <7 years-
functional inhibition, sensorimotor & language function, 2, 16–19, 22, 73, 75, 134, 135,
extraoperative old; multiple surgeries with prolonged surgical planning
ictal & interictal EEG recording ictal foci invasive monitoring 141–145, 146–152

Figure 13–1. Common techniques for pediatric brain mapping.

activity, white matter pathways) or technique (noninva- Figure 13–1 describes the most common techniques
sive and invasive). Noninvasive techniques assay relative for pediatric brain mapping, their clinical indications
changes in local perfusion (fMRI and SPECT), metabolism in children, and their present limitations in this patient
(PET), or magnetic flux (MEG) as surrogate markers of population.
increased neuronal activity during either spontaneous
activity or a specific task. DTI together with axonal trac-
tography allows the specific delineation of white matter 䉴 NONINVASIVE TECHNIQUES
tracts.
Invasive testing, including the Wada procedure for OVERVIEW
determination of hemispheric dominance and direct cor-
tical stimulation for mapping eloquent function, remain PET, fMRI, and MEG are powerful, noninvasive means
the “gold standards.”2,10,22 These studies use pharmaco- of mapping sensorimotor and language function.
logical (Wada) or electrical (ECS) methods to produce Frequently used in adults to guide surgical resec-
spatially and temporally restricted neuronal inhibition, tion of supratentorial lesions near eloquent cortical
while assessing the patient for a corresponding tem- regions,1,2,10,23−26,55,56,153−158 these techniques may be
porary clinical deficit. In addition, invasive electroen- difficult to perform in young children. In addition, the
cephalography (EEG) using subdural grid and strip elec- reliability of detailed functional maps obtained from
trodes and intraparenchymal depth electrodes remains these techniques remains controversial in children, es-
the “gold standard” for identifying the epileptogenic pecially for functional mapping of cortex near tumors,
zone. in patients with refractory epilepsy, and for language
There is strong interest in noninvasive techniques mapping.2,10,23,25,27,158,130
to identify the epileptogenic zone as well as eloquent Noninvasive functional imaging techniques differ
cortex. Experimental studies in children have focused fundamentally from their invasive counterparts. Nonin-
on developing clinical protocols that improve their suc- vasive mapping paradigms measure activation of a re-
cess and accuracy, with validation through comparison gion(s) in response to a particular task or series of tasks.
to invasive means of brain mapping. In contrast, invasive techniques rely on the inhibition
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 169

of function using drugs (Wada) or inhibition or electric children. Factors that decrease the signal-to-noise ratio
current (ECS). As a result, direct comparisons of func- include poor cooperation, anxiety and head movement,
tional regions identified using invasive and noninva- sedation, small patient size, structural immaturity, and
sive testing should be carefully considered. Noninva- physiological differences in cerebrovascular response.23
sive methods may overestimate the area of functional Behavioral effects may present the most significant ob-
cortex for a given function which, in turn, may limit stacle to fMRI in children, especially those with focal
resection.2 Noninvasive tests are limited by spatial res- neurological signs, developmental delay, or psychiatric
olution, changes in brain hemodynamics secondary to disorders such as autism. Children often experience rest-
tumors or arteriovenous malformations (SPECT, fMRI), lessness and anxiety and have difficulty following com-
signal-to-noise limitations for paradigms relying on pa- mands, sustaining attention to functional tasks, and re-
tient cooperation, and source localization secondary to maining still for extended periods in the MRI scanner.
the underlying assumptions of the model (MEG). Discomfort or anxiety leads to poor attention to func-
A common clinical application of noninvasive func- tional tasks, head movement, and the recruitment of
tional brain imaging is localizing a cryptogenic seizure neural networks that are not function-specific. Behav-
focus in children with refractory epilepsy. Seizures be- ioral conditioning including the use of mock scanners
gin during childhood or adolescence in 60% of epilepsy may diminish anxiety and decrease mapping failures.15
patients.106 Approximately 30% of children become re- Having a child view a cartoon videotape in the scan-
fractory to medical treatment with multiple agents. Surgi- ner reduces motion and improves successful functional
cal resection of the ictal onset zone leads to either com- scanning, especially in children younger than 5 years
plete elimination of seizures or a significant reduction without sedation.30
in seizure frequency in 80% of children with temporal Inferior frontal speech areas (Broca’s area) usually
lobe epilepsy. Surgery is less successful for extratempo- require activation by a verb generation task, because in-
ral seizures, due to difficulty in localizing the focus with- consistent activation is seen with passive stimuli (which
out a structural lesion on MRI, multifocal pathology, or commonly activate posterior temporal primary auditory
discordant localizing data from noninvasive studies.121 cortex and receptive language centers). This may be
Functional neuroimaging techniques assessing focal ab- very difficult for young children. Passive tasks in chil-
normalities of brain perfusion or metabolism (SPECT, dren with cerebral pathology may not be reliable, as
PET), or ictal and interictal foci of abnormal electro- bilateral activation and failure to determine dominance
physiology (invasive video EEG (VEEG), MEG) have are frequently observed.23 However, creative passive
improved detection of the epileptogenic zone and in- tasks, such a listening to a story with key words omit-
creased the number of children who may benefit from ted, have activated unilateral inferior frontal speech cen-
surgical treatment. Microscopic areas of focal cortical ters in small studies. Continued development of testing
dysplasia are a common source of partial seizures in paradigms may improve fMRI-based language mapping
children with a normal MRI.121 in young children.31 Despite efforts to improve coop-
eration and decrease anxiety, the failure rate of fMRI is
approximately 30% children less than 8 years, and almost
FUNCTIONAL MRI 60% in children less than 6 years.29
The developing brain has higher synaptic density
fMRI measures microvascular changes in deoxyhe- and relative gray matter volume compared to the mature
moglobin concentration, known as the blood oxygen adult. In addition, there are differences in cerebral vascu-
level dependent (BOLD) signal. Changes in oxyhe- lar responsiveness, especially in neonates.15 These fac-
moglobin and deoxyhemoglobin ratios are a surro- tors affect the BOLD signal response, confounding fMRI
gate for neuronal activity, because increased neuronal data in children.15,28,32,33 Patients younger than 12 years
metabolism is coupled to local increases in perfusion un- have significantly more variation in the BOLD signal,
der physiological conditions.22,28 One limitation of fMRI, leading to increased noise, diminished signal-to-noise
especially when used for language mapping, is that it ratio, and smaller activation volumes.28
reveals cortex specifically recruited during a particular The majority of brain growth is finished by 5 years,
task. This may vary significantly according to the task but myelination continues into adulthood. Major white
performed. Areas of activation may not be required for matter tracts are visible by 3 years, and the ratio of gray to
task performance; conversely, direct cortical stimulation white matter is comparable to adults at 7 years. Synap-
only identifies essential cortex.23 tic density peaks at 1 year and subsequently declines
until age 16; this is mirrored by changes in cerebral glu-
cose metabolism.15 A faster heart rate, thinner skull, and
General Considerations in Pediatrics
smaller body prevent dampening of cardiac and respira-
fMRI is noninvasive, does not involve ionizing radiation, tory motion, which contribute to increased physiological
and has no known adverse effects,23,29 making it safe for noise in children.33 In addition, fetal hemoglobin
170 SECTION I TECHNIQUES

accounts for 20% all hemoglobin at 4 months, decreas- identified within a structural abnormality or the epilep-
ing the sensitivity of fMRI in neonates.33 Comparing fMRI togenic zone.40 Determining hemispheric dominance
data between children and adults may be confounded in children is challenging. Clinical data and structural
by differences in brain size. This introduces error in the MRI may not accurately predict language lateralization,
localization of functional activity when images are trans- possibly due to functional reorganization.32,40 Preoper-
formed into a common stereotactic space. Spatial nor- ative fMRI may help direct the placement of subdural
malization of MRI structural data is similar to the adult electrode arrays, or surgery may be deferred if func-
by approximately 6 years of age.15 These factors are neg- tional activation is identified within the epileptogenic
ligible in children more than the age of 7 years. zone.40
Half of all pediatric MRI scans, and the majority in
children less than the age of 7 years, require sedation.
Cortical Plasticity and Reorganization
Commonly used agents impact cerebral perfusion and
diminish the sensitivity of fMRI. Propofol causes a dose- A better understanding of age-dependent plasticity may
dependent inhibition of functional activation. Primary inform surgical decision-making and help predict neu-
motor and auditory cortex are easily activated, even at rological recovery after traumatic brain injury. For ex-
high doses and in very young children.32,33 However, ample, the age at which complete disconnection of the
association centers such as language cortex are highly dominant hemisphere will result in permanent language
sensitive to propofol; in addition, less function-specific dysfunction is not precisely defined. Historically, a cutoff
passive tasks are required in the sedated patient.34,35 The of 6 years of age has been used to define the estimated
effects of other medications have not been studied. end of the critical period of language acquisition.41 How-
Given these limitations, fMRI techniques must ever, fMRI studies in children as old as nine have corre-
be verified against direct cortical mapping in chil- lated recovery of language with recruitment of contralat-
dren. Several studies in children undergoing craniotomy eral perisylvian cortex.12
have compared fMRI sensorimotor maps with intra-
operative cortical stimulation, with generally excellent
agreement.36,37 Future clinical applications of fMRI are DIFFUSION AND PERFUSION MRI
likely to evolve if normative patterns of brain devel-
opment can be established. This may enable the early DWI and DTI are increasingly being used in children.
diagnosis of functional deficits, psychiatric disease, and DWI images the degree of random motion of extracel-
cognitive deficits in children, facilitating early interven- lular water molecules; pathology that restricts extracel-
tion and possibly improving outcomes.38 lular water motion (e.g., cytotoxic edema) creates con-
trast on DWI. This technique is most commonly used
for the early detection of stroke in adults,42,43 but is be-
Brain Tumors
coming part of routine MRI in young children, especially
Changes in cerebral perfusion and metabolism in or neonates.159
near a focal lesion (e.g., tumor, arteriovenous malfor- DTI measures diffusion of water molecules along
mation) or chronic seizures may alter the BOLD sig- axons, and it can display large white matter tracts as dis-
nal and diminish the sensitivity of fMRI-based functional tinct structures.50−52,160 DWI and DTI do not require pa-
mapping.2,11,32,35,39 This translates into 20% discordance tient cooperation. PWI utilizes intravenous gadolinium
between fMRI and intraoperative ECS maps of sensori- to create maps of relative cerebral blood volume (rCBV)
motor cortex, and as much as 66% discordance for lang- and relative cerebral blood flow (rCBF). It is used in
uage maps.2,26 Malignant gliomas of the ipsilateral hemi- assessing cerebral ischemia, brain tumors, and TBI.53
sphere are associated with decreased fMRI activation
volumes and increased anatomic variability.24 However,
Diffusion-Weighted Imaging
fMRI may be complementary to ECS among children
with tumors near eloquent cortex. fMRI may localize DWI is superior to conventional MRI sequences in the
function to areas inaccessible by ECS, such as dis- early differentiation of hypoxic-ischemic reperfusion in-
tant from the craniotomy site or deep within cortical jury patterns in the neonate.43,44 For example, a modest
sulci.11 decrease in cerebral perfusion produces a “border zone”
pattern of restricted diffusion before changes are seen on
T1- or T2-weighted imaging. Prolonged hypoperfusion
Epilepsy
leads to a more diffuse pattern, affecting the brainstem,
Functional reorganization is common in children with re- thalamus, paracentral white matter, and optic radiations.
fractory epilepsy.12,25,40,41 fMRI may improve the safety DWI is more sensitive than conventional MRI techniques
of ictal focus resection by sparing functional cortex, for detection of diffuse ischemic injury, and differentiat-
as well as inform treatment decisions when function is ing this pattern from focal infarction in the newborn.42−44
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 171

Among young children with traumatic brain injury, le- Perfusion-Weighted Imaging
sions on DWI correlate with an increased likelihood of
The most common technique for PWI used in children
seizures, neurosurgical intervention, mechanical ventila-
is dynamic susceptibility contrast imaging. During the
tion, and inpatient rehabilitation.44
first pass of gadolinium through the cerebral vascular
DWI may better distinguish cellular from cystic
bed, changes in T2 signal occur in the surrounding brain
brain tumors, such as medulloblastoma and pilocytic as-
parenchyma due to the high concentration of paramag-
trocytoma. DWI also aids in the differentiation of tumor
netic contrast. The intensity of change in T2 signal is
recurrence and radiation necrosis. Tumor recurrence is
strongly correlated with perfusion. This technique has
associated with restricted diffusion, whereas radiation
several advantages in children over other noninvasive
necrosis typically displays higher diffusion. In addition,
methods (e.g., PET and SPECT). This includes the su-
diffusion changes precede T2 signal change in children
perior spatial resolution of MRI, rapid image acquisi-
with leukoencephalopathy. DWI may also differentiate
tion, and the absence of ionizing radiation. In addition,
posterior reversible encephalopathy syndrome from ir-
PWI is highly sensitive to the microvasculature, which is
reversible neurological injury in children undergoing
the anatomic location of many types of cerebrovascular
chemotherapy. Preserved diffusion predicts reversible
pathology observed in children.53
injury, and restricted diffusion suggests irreversible dam-
Dynamic susceptibility imaging is wellsuited for
age due to cytotoxic edema, and correlates with a poor
children. The smaller cross-sectional area of cerebral
prognosis.43,45
vasculature and higher pulse combine to concentrate
As in adults, DWI is helpful in the diagnosis of
gadolinium as a rapid bolus in a relatively small vol-
cerebritis, encephalitis, or brain abscess, as well as mon-
ume, improving the signal-to-noise ratio. In children
itoring treatment response to antimicrobial therapy. DWI
younger than 5 years, these advantages may be offset
may reveal focal diffusion abnormalities with herpes
by the need for manual injection due to small venous
encephalitis prior to signal changes on routine MRI.
caliber.53 Clinical applications of PWI include the evalua-
Restricted diffusion among patients with acute dis-
tion and management of brain tumors, cerebral ischemia,
seminated encephalomyelitis (ADEM) predicts a poor
and traumatic brain injury. Higher maximum rCBV corre-
long-term outcome.43,46 Peri-ictal DWI shows decreased
lates with higher grade in gliomas. In children diagnosed
diffusion in the epileptogenic zone, and may help in lo-
with medulloblastoma, PWI is sensitive at differentiating
calizing this region in patients with refractory seizures
tumor recurrence from radiation necrosis, which may
and multifocal pathology or discordant MRI, EEG, and
be indistinguishable by conventional MRI.53 In the con-
seizure semiology.43 In addition, early studies using DWI
text of acute cerebral ischemia, such as Moyamoya syn-
to lateralize nonlesional temporal lobe epilepsy have
drome, PWI in combination with DWI may delineate the
demonstrated encouraging results.47
ischemic penumbra, or viable but dysfunctional tissue at
risk for infarction.53 Finally, the number and depth of
Diffusion Tensor Imaging microhemorrhages due to diffuse axonal injury correlate
with neurological outcome in children.54
DTI often demonstrates significantly diminished sig-
nal along frontal and supracallosal white matter tracts
among children following TBI. These changes correlate
with clinical deficits of executive function and motor MAGNETOENCEPHALOGRAPHY
speed.48 Among children with congenital malformations
such as holoprosencephaly, the size of the white mat- MEG records magnetic flux produced by electrical cur-
ter tracts correlates with disease type and severity.49−51 rents that result from ion flow across excited neu-
Among children undergoing surgery for brain tumor re- ronal membranes.57 The magnetic field generated by
section, DTI may assist in preoperative planning and these currents is not distorted or degraded by the
intraoperative navigation by delineating the relation- skull or scalp, so MEG has better spatial resolution
ship between neoplasm and functionally eloquent white with similar temporal resolution compared to scalp EEG
matter.51,52 DTI can also distinguish tumor infiltration recordings.56 MEG and EEG are complementary, be-
from compression of white matter tracts, which may im- cause they are each most sensitive to currents from dif-
pact surgical decision-making.52 Unlike DWI, DTI is not ferent sources. MEG is sensitive to tangential flux, gen-
sensitive to acute changes in extracellular diffusion. In erated from dipole sources within sulci, whereas EEG is
addition, present DTI techniques have relatively poor more sensitive to radial dipoles.58
spatial resolution compared to conventional MRI tech-
niques. This limits the clinical utility of DTI as a diagnos-
General Considerations in Pediatrics
tic tool.51 However, as more DTI studies are performed
in children with white matter pathology, additional clin- MEG is noninvasive and safe in children, with no known
ical applications are likely to emerge.50 side effects. Compared to fMRI, MEG has the advantage
172 SECTION I TECHNIQUES

of millisecond temporal resolution. Like fMRI, MEG Functional Mapping


can accurately localize interictal and ictal foci in rela-
Early studies in children with refractory epilepsy re-
tion to eloquent cortex. MEG is frequently an impor-
ported successful sensorimotor mapping with MEG in
tant part of the presurgical evaluation for intractable
54%.63 More recent studies comparing MEG-based sen-
epilepsy.59,60 Functional mapping with MEG also em-
sorimotor mapping with ECS in children with epilepsy
ploys task-oriented patient cooperation, and head move-
have demonstrated near-perfect concordance.56 In chil-
ment is a common source of error in children.23 MEG
dren younger than 6 years, MEG-based sensorimotor
directly monitors electrophysiology, rather than using a
mapping is less sensitive, because small patient size am-
surrogate marker such as perfusion or metabolism like
plifies physiological noise. Promising technical solutions
fMRI, SPECT, and PET. Sources of interictal spikes and
include spatial filtering, which has successfully mapped
functional activity can be accurately projected onto MRI
sensorimotor cortex in healthy children as young as
images using fiducial markers to coregister the mod-
3 years.64 Simultaneous EEG and MEG data may im-
eled activity to a structural MRI. This technique is mag-
prove the spatial accuracy of functional mapping of so-
netic source imaging (MSI). MEG may noninvasively
matosensory cortex in children with ipsilateral structural
map both functional cortex and ictal foci, and guide inva-
lesions.65 MEG has very high sensitivity (98%) and speci-
sive cortical mapping (Fig. 13–2).59 The use of sedation is
ficity (93%) for determining hemispheric dominance in
often required to ensure patient compliance during MEG
children older than 8 years.66 MEG may be a noninvasive
studies in young children and those with developmental
alternative to the Wada procedure. However, Wada test-
delay and behavioral problems.61,62 Midazolam signifi-
ing is still necessary if memory assessment is needed.66
cantly decreases the sensitivity of MEG for spike activity
Functional mapping of receptive language centers
and sensorimotor mapping, with reported failure rates
has demonstrated excellent test–retest reliability in small
of almost 30%. The combination of chloral hydrate and
studies including children older than 10 years.57 MEG-
propofol is associated with much lower failure rates in
based receptive language maps of healthy volunteers
children.61,62

B1 B2

B3 C

Figure 13–2. (A) Averaged response for 512 pneumatic tactile stimulation of the left
index finger from all 248 magnetometer sensors (4-D Neuroimaging, San Diego, CA)
with peaks at 24 milliseconds, 51 milliseconds, and 93 milliseconds. (B) The magnetic
source images (MSI) demonstrating localizations for the 24 (anterior ) and 51
(posterior) millisecond responses in the sagittal (B1 ), coronal (B 2 ) and axial planes
(B 3 ). (C) Composite three-dimensional rendering of digital imaging and
communications in medicine images with multiple left and right upper extremity digit
trials. Responses are shown in red.
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 173

reveals adult patterns of cortical activation in children In children, refractory epilepsy is more often neo-
more than the age of 7 years.67 In addition, sensorimo- cortical than mesial temporal in origin.74 MEG is less
tor mapping via MEG correlates well with intraoperative sensitive than invasive monitoring with intracranial elec-
ECS in older children undergoing surgery for brain tu- trodes in identifying extratemporal seizure foci.75 How-
mor resection.34,68 MEG-based functional maps inform ever, MEG has a significantly better yield at identifying
surgical decision-making and intraoperative navigation neocortical foci compared to medial temporal disease
in children with tumor infiltrating or adjacent to MEG- among patients with cryptogenic seizures.56,79 In addi-
defined functional cortex.26,69 tion, asymmetric MEG dipole clusters demonstrated high
MEG studies have shown a high incidence of sen- concordance with intraoperative electrocorticography
sorimotor reorganization in children as young as 2 years (ECoG) in children with lesional, neocortical epilepsy.
with abnormalities of cortical proliferation (e.g., corti- The resection of remote ictal zones identified by MEG
cal dysplasia), including relocation in the contralateral and ECoG does not contribute to epilepsy outcome in
hemisphere. Similar changes were not observed in chil- patients with tumors.74 However, in patients with corti-
dren with abnormalities of cortical organization, such cal dysplasia, the resection of remote ictal regions may
as polymicrogyria. This stresses the importance of func- be critical to seizure outcome, as small populations of
tional mapping in the preoperative evaluation of patients intrinsically epileptogenic, dysplastic neurons are likely
with cortical dysplasia for epilepsy surgery.70 Cortical to exist at these locations.74
reorganization may also inform negative mappings via Most authors recommended the use of iVEEG to
ECS, which is subject to sampling error due to its focal define the epileptogenic zone in patients with multi-
employment and limited cortical coverage. ple clusters on MEG.79 An emerging technique to map
MEG/MSI studies are limited by the most common the temporal and spatial spread of interictal spike ac-
method of localizing sources of evoked waveforms, the tivity is gradient magnetic field topography, which may
equivalent current source dipole algorithm. This assumes complement the interpretation of interictal MEG in pa-
the source to be a zero-volume point in a homogeneous tients with multiple clusters by defining epileptogenic
sphere.71 Good correlation between ECS and motor, sen- networks.80 Invasive or scalp VEEG monitoring and MEG
sory, and language point dipole localization on MEG provide complementary data. Simultaneous EEG record-
has been demonstrated in adults.26,55,68 However, this ing during MEG sessions have been shown to improve
method cannot precisely define the boundary between sensitivity for the detection of interictal epileptogenic
a lesion and functionally critical brain.72 activity.58,81 Like all noninvasive techniques of ictal focus
mapping, MEG provides the most information in the con-
text of a multimodality evaluation for refractory seizures.
MEG may be especially useful in identifying the
Epilepsy
epileptogenic tuber in patients with tuberous sclerosis
MEG measures only interictal activity over short time complex (TSC) and focal seizures. 90% of patients suf-
intervals (less than 1 hour), compared to video EEG fering from TSC experience seizures, and these are re-
(VEEG) monitoring which records both ictal and inter- fractory to medical treatment in 50% of cases.58 Focal
ictal activity over a period of days. Despite this, MEG is seizures are usually mapped to tubers and adjacent cor-
a sensitive noninvasive method of predicting the epilep- tex. However, the majority of these patients have multi-
togenic zone.73 The location of interictal spikes detected ple tubers, and identifying the epileptogenic lesion may
by MEG accurately predicts the epileptogenic zone in be challenging.58 MEG has demonstrated high accuracy
children,73−76 but cannot delineate the extent of abnor- in predicting the epileptogenic zone based on prelimi-
mal tissue.72 Like MRI, MEG acquisition is generally lim- nary studies, and is superior to other noninvasive tech-
ited to the interictal period. However, in select children niques such as scalp VEEG or ictal SPECT.82,83 The pres-
with frequent, motionless partial seizures, ictal MEG may ence of a single cluster compared to multiple clusters
be possible, good results compared to ictal SPECT and or bilateral discharges was able to accurately differenti-
EEG.77 ate patients with a single, primary ictal onset zone from
Approximately one-third of children with refractory those with multiple or bilateral ictal onset zones that are
partial epilepsy have a normal MRI.72 Outcome studies part of a larger epileptogenic network.84 Taken together,
demonstrate that MEG has a sensitivity of 50–60% and this data suggests that MEG may identify a subset of pa-
a positive predictive value of 80–90% for identifying the tients with focal epilepsy and TSC with a single epilep-
epileptogenic zone.73,75,76 The concordance of EEG and togenic tuber, who may not require invasive monitoring
MEG localizing a single seizure focus predicts postop- with intracranial electrodes prior to tuber and ictal focus
erative seizure freedom.74,78 Bilateral MEG clusters or resection.
the absence of an MEG cluster within an area of cor- In addition to TSC, MEG is part of the clinical
tex targeted for surgical resection correlates with poor evaluation of Landau-Kleffner syndrome (LKS), acquired
outcome.72,74 epileptiform aphasia. MEG shows perisylvian interictal
174 SECTION I TECHNIQUES

discharges in many children.85 The temporal resolution been studied extensively in children undergoing cran-
and sensitivity to interictal sources deep within the syl- iotomy. Indications include functional mapping ([15 O]-
vian fissure of MEG has been combined with pharma- H2 O), seizure focus mapping (FDG, MET, FMZ), and the
cological sleep and methohexital suppression to iden- delineation of the boundaries of intra-axial neoplasms
tify a single sylvian pacemaker in some children with (FDG, MET). In some centers, overlap between a seizure
LKS, and multiple subpial transections in this region have focus and eloquent brain regions defined by PET is used
stopped all interictal spike activity in a small number of as a criteria for nonsurgical management of children with
patients.86 Similar techniques have been used to iden- medically refractory epilepsy.89
tify unilateral interictal sources in patients with benign
rolandic epilepsy of childhood,87 as well as to study dif-
ferences between focal seizures and hypsarrhythmia in Functional Mapping
children with West syndrome.88 Functional mapping with PET has a lower spatial reso-
lution compared to fMRI. Small studies in children have
demonstrated excellent concordance between ([15 O]-
POSITRON EMISSION H2 O)-PET and hemispheric dominance on Wada test.92
TOMOGRAPHY Language mapping using PET was reported in children
as young as 3 years, using unverified speech activation
PET uses radiolabeled tracers to image cerebral perfu- tasks.92
sion ([15 O]-H2 O), glucose metabolism (2-[18 F]fluoro-2-
deoxy-D-glucose, or FDG), amino acid transport and pro-
tein metabolism ([11 C]-L-methionine, or MET), and GABA Brain Tumors
receptor density ([11 C]-flumazenil, or FMZ).89−91 PET im- In adults and children, FDG and MET uptake in tu-
ages coregistered to standard MRI allow anatomic lo- mors are predictors of outcome and tumor grade in-
calization of perfusion changes (functional mapping), dependent of histopathology.91,92,97,98 FDG uptake is a
glucose or amino acid metabolism (ictal focus or tumor more sensitive predictor of tumor grade than gadolinium
mapping), or GABA receptor concentration (ictal focus enhancement.98,99 However, low-grade tumors common
mapping).89 in children such as pilocytic astrocytoma and choroid
plexus papilloma, demonstrate brisk FDG uptake.98 Clin-
ically, MET and FDG-PET can assess response to treat-
General Considerations in Pediatrics ment and identify tumor recurrence.100 MET uptake is
PET has the advantage of an open environment reducing more sensitive than MRI with gadolinium for identi-
the potential for claustrophobia and anxiety.92 Because fying the tumor boundaries.97 Late changes on MET-
PET requires administration of radioisotopes, normative PET in children with a remote history of low-grade
pediatric data is scarce. This is especially problematic for glioma may represent treatment-related pathology, such
functional mapping of language during normal devel- as meningioangiomatosis.101
opment. In addition, a poor signal-to-noise ratio means PET imaging with FDG and MET tracers coregis-
studies are often repeated.23 PET imaging is not widely tered to MRI is used in some centers to select stereo-
available, limiting its clinical use, especially in children.91 tactic biopsy targets in children with infiltrative tumors
FDG, which delineates specific cortical regions of that display heterogeneous signal changes on traditional
high or low metabolism, is the most common PET tracer MRI. This approach may decrease the incidence of non-
used for brain mapping. Compared to technetium-99m diagnostic biopsies (estimated to be approximately 5% of
SPECT and FDG-PET studies in adults, relatively low- MRI-guided stereotactic brain biopsies) and increase the
radiation doses to the brain and bladder are adminis- safety by decreasing the sampling rate.91,99 The safety
tered with FDG-PET studies in neonates. This may be may be further improved with functional mapping us-
attributed to the short half-life of FDG compared to ing ([15 O]-H2 O)-PET. PET may also demonstrate resid-
technetium-99m DTPA, and the high ratio of brain to ual tumor following resection, informing decisions about
total body mass in infants.93 However, clinical studies of early second-look operations and assisting in radiother-
long-term outcome that quantify the risk to children are apy planning.99,102 Functional and metabolic PET maps
lacking. Like fMRI and MEG, streamlined FDG-PET pro- may guide intraoperative tumor resection.103
tocols (e.g., venipuncture instead of arterial sampling)
are used for infants and small children.94
Epilepsy
Sedation with propofol in children undergoing
FDG-PET produces dose-dependent hypometabolism in A strong correlation exists between foci of interictal hy-
both parietal and occipital lobes. This may diminish the pometabolism revealed by FDG-PET and epileptic foci
sensitivity of FDG-PET in sedated children.95,96 PET has on iVEEG.90,104 Interictal FDG-PET has a sensitivity of
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 175

60–80% for identifying foci of hypometabolism among SINGLE PHOTON EMISSION


patients with chronic, refractory extratemporal epilepsy TOMOGRAPHY
and a normal MRI.90,105 This sensitivity is similar to ictal
SPECT.90,105 The sensitivity of interictal FDG-PET for pa- SPECT has become a commonly used tool in the
tients with neocortical temporal lobe seizures is approx- management of pediatric brain tumors and refractory
imately 90%.104,106 FDG-PET is more sensitive but less epilepsy.91 Unlike PET, a cyclotron is not required to
specific in detecting cortical dysplasia.107 Areas of FDG- generate tracers and therefore SPECT is more widely
PET hypometabolism include regions of rapid seizure available. Although PET is reported to be more sensi-
propagation in addition to the ictal onset zone. This poor tive for the detection of perfusion abnormalities, SPECT
specificity is exaggerated in extratemporal epilepsy.90,105 is much more widely available in the clinical setting,111
FDG-PET also has a low sensitivity in children with new- and is more sensitive than FDG-PET in the clinical de-
onset partial epilepsy; abnormalities on FDG-PET corre- tection of brain metabolism.91,112,113
late with chronic seizure activity.33,105
Among neonates with infantile spasms, includ-
General Considerations in Pediatrics
ing those with an EEG characterized by hypsarrhyth-
mia, FDG-PET may demonstrate one or more abnormal Brain perfusion SPECT imaging is most commonly per-
foci (65% patients have multiple foci). The significance formed with 99m technetium-hexamethylene propylene
of these changes in infants with generalized spasms amine (99m Tc-HMPAO), 99m technetium-ethyl cysteinate
remains controversial.105,108 Longitudinal studies failed dimer (99m Tc-ECD),114 or N-isopropyl-(123 I) p-iodoamp-
to demonstrate a prognostic significance.108 However, hetamine (NPI).111 Less common tracers include 99m Tc-
children with a single FDG-PET abnormality that corre- methoxyisobutylisonitrile (MIBI), 99m Tc-tetrofosmin, and
99m
lates with focal changes on EEG typically have an ex- Tc-glucoheptonate.113 HMPAO is more sensitive than
cellent outcome following seizure focus resection in- ECD in identifying the epileptic zone, and is the most
clusive of the PET hypometabolic focus.105 FDG-PET common tracer used in pediatric epilepsy.115 SPECT
may have prognostic value in patients with infantile is commonly used to assess cerebral perfusion in
spasms diagnosed with West syndrome. Persistent fo- the pre- and postoperative management of Moyamoya
cal hypometabolism on FDG-PET after medical treat- disease,116 and is increasingly being used to study the
ment is correlated with developmental delay. Persistent response to psychiatric treatment117 and injury118 in chil-
regions of hypometabolism after therapy that correlate dren.
with focal EEG abnormalities represent structural ab-
normalities such as cortical dysplasia.109 This is con-
Brain Tumors
sistent with other data showing a higher incidence of
FDG-PET abnormalities in children with longer seizure Studies with multiple different tracers have failed to
duration.33 establish SPECT as a valuable tool for predicting tu-
The introduction of [11 C]-flumazenil, or FMZ, which mor histology or recurrence in many pediatric brain
images GABAA receptor density, has refined the use tumors.119,120 MIBI-SPECT studies show tracer accumu-
of PET in the identification of the ictal onset zone.90 lation in both low- and high-grade infiltrative gliomas,
Changes on FMZ-PET are more specific to the ictal on- as well as brainstem gliomas. Other common pediatric
set zone when compared to FDG-PET. FMZ may demon- brain tumors including medulloblastoma, optic pathway
strate mesial temporal dysfunction when MRI does not glioma, and ependymoma do not accumulate MIBI.119
clearly demonstrate hippocampal sclerosis. In addition, Clinical studies using 99m Tc-tetrofosmin among children
FMZ-PET has a high sensitivity for demonstrating small with suspected tumor recurrence in the posterior fossa
foci of cryptogenic cortical dysplasia (decreased FMZ up- failed to show tracer accumulation in most cases of tu-
take due to decreased density of GABAA receptors).90 mor recurrence.120 However, preliminary results suggest
The novel PET tracer ␣-[11 C]-methyl-serotonin, or 99m
Tc-glucoheptonate SPECT may differentiate tumor re-
AMT, which images tryptophan metabolism, has shown currence from radiation necrosis among children treated
promising results in preliminary clinical studies aimed for medulloblastoma with an equivocal MRI.113
at distinguishing epileptogenic and electrically silent le-
sions in children with multifocal pathology.90,105 Stud-
Epilepsy
ies in children with tuberous sclerosis and refractory
epilepsy have shown FDG-PET may complement MRI Ictal and interictal perfusion mapping with HMPAO-
with DTI in differentiating epileptogenic from clini- SPECT may improve detection of the epileptogenic zone
cally silent tubers.110 Higher apparent diffusion coeffi- in children.106 Like PET, ictal SPECT is clinically useful
cient (ADC) volumes on DTI and larger volumes of hy- among patients with refractory seizures and conflicting
pometabolism on interictal FDG-PET compared to tuber clinical and MRI data or multifocal disease.114,121,122 Ictal
size distinguished epileptogenic tubers.110 SPECT, where tracer injection is performed during a
176 SECTION I TECHNIQUES

seizure may demonstrate focal hyperperfusion corre- Patients often develop focal neurological deficits and
sponding to the ictal onset zone. This sensitivity of this developmental delay secondary to cerebral hypoper-
technique is comparable to FDG-PET (50–80%) for lo- fusion and infarction. This is most commonly treated
calization of the ictal onset zone.121 Interictal SPECT, by revascularization via indirect surgical bypass (e.g.,
which may demonstrate focal hypoperfusion, is much pial synangiosis, encephaloduroarteriosynangiosis). Dy-
less sensitive.122 namic studies quantifying cerebrovascular reserve with
The yield of ictal SPECT is optimal when the tracer HMPAO-SPECT before and after acetazolamide stress
injection occurs within 45 seconds of seizure onset. Late may predict ischemia, and patients with decreased
injections often lead to false positive or negative results reserve by SPECT benefit from surgical revascu-
in children due to rapid seizure spread. Therefore, ic- larization.116,162 In addition, dynamic SPECT after
tal SPECT requires a monitored, inpatient setting with revascularization predicts clinical outcome, and postop-
trained staff and available tracer. Ictal injection failure is erative surveillance permits early intervention before the
minimized when patients are monitored in a dedicated development of neurological symptoms among patients
video EEG monitoring unit.114,121 Ictal injection is more with persistently diminished cerebrovascular reserve.116
technically challenging in children because of the high SPECT has been used to characterize cortical plas-
incidence of extratemporal seizures, short seizure dura- ticity and functional recovery among children after TBI.
tion, and frequent absence of an aura or warning signs Higher perfusion after injury correlates with improved
of an impending seizure.106 In addition, interictal injec- language and cognitive outcome 3 years later.118 Corti-
tions may correspond to early or subclinical ictal activity cal regions that subserve language normally demonstrate
in 5% of patients.123 late decreases in regional perfusion, corresponding to
Combining interictal and ictal SPECT, such as by late maturation of complex associative function.118,163
subtraction ictal SPECT coregistered to MRI (SISCOM), Significant increases in overall cerebral perfusion among
improves the sensitivity of ictal SPECT for localization children with good language outcome following TBI
of the ictal onset zone.106,114,122 Among children with may reflect delayed but preserved functional maturation.
cryptogenic focal epilepsy, SISCOM improved the sen- Finally, SPECT may help monitor treatment re-
sitivity of ictal SPECT from 80% to greater than 90% in sponse among children with attention deficit hyperac-
multiple studies.106,121 Like other noninvasive mapping tivity disorder (ADHD).117 Children with ADHD have
techniques, SISCOM may help plan intracranial elec- regional hyperperfusion in the prefrontal and temporal
trode placement, and in select patients may eliminate the cortex on SPECT. Methylphenidate treatment is associ-
need for invasive monitoring.38,123 Frontal lobe epilepsy ated with lasting improvement in perfusion in the frontal
(FLE) is often characterized by extremely rapid spread of and temporal cortex among responders.117
epileptic activity. Ictal SPECT may be especially useful
in providing preoperative guidance for intracranial elec-
trode placement in children with FLE, as rapid seizure 䉴 INVASIVE TECHNIQUES
spread often renders false-localizing clinical and elec-
trophysiological data. Ictal SPECT has a sensitivity of OVERVIEW
approximately 60% in detecting frontal ictal foci, which
may be higher with SISCOM.124,125 Direct ECS remains the gold standard for mapping elo-
Similar to PET, NPI-SPECT has been used in in- quent cortex.2,10,22,164 It is performed intraoperatively
fants with West syndrome to monitor the effectiveness with a hand-held stimulator (requiring an awake patient
of medical therapy, and normalization of multifocal hy- in the case of language mapping), or extraoperatively
perperfusion abnormalities correlates with neurological across implanted subdural electrodes (Fig. 13–3). ECS
outcome.111 This is concordant with PET data suggesting identifies discrete regions essential for a specific task,
perfusion abnormalities before treatment primarily rep- which if resected, are likely to produce permanent neu-
resent transient phenomena rather than foci structural rological deficits. Noninvasive tests discussed in the pre-
lesions.109 A minority of patients with West syndrome vious section highlight regions actively recruited during
may have a single focus of hyperperfusion demonstrated a particular task, but not necessarily essential.2,140 Wada
by ECD-SPECT or PET studies.105 Surgical resection of testing, also known as the intracarotid Amytal test, is a
this focus abolishes seizure activity in many patients.161 means of establishing hemispheric dominance for lan-
guage and memory function by asking the patient to
perform specific tests while one hemisphere is anes-
Additional Clinical Applications
thetized by sodium amobarbital infusion directly into the
Moyamoya disease is an inflammatory vasculopathy in ipsilateral internal carotid artery.16,20,21 Both techniques
children characterized by progressive occlusion of the are used clinically in children undergoing evaluation for
carotid systems, and the development of a fine network craniotomy, especially for the resection of a seizure fo-
of small collateral vessels along the base of the brain. cus or brain tumor.
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 177

A B

Figure 13–3. Photographs depicting intraoperative stimulation mapping. (A)


Hand-held bipolar stimulator applied to exposed frontal cortex to identify
sensorimotor function during craniotomy for epilepsy in a child under general
anesthesia. (B) A four-contact strip electrode (Adtech, Inc., Racine, WI) laid on
exposed posterior frontal cortex for identification of sensorimotor cortex by
stimulation across the strip electrodes prior to cortical resection.

WADA TESTING epilepsy or brain tumors, where functional reorganiza-


tion is common.12,24 However, the external validity of
The Wada test can determine hemispheric dominance published data is limited by small sample size and vari-
for language and memory, with a sensitivity greater than able criteria for establishing hemispheric dominance or
90% in children more than the age of 10 years.16 It “successful” Wada testing.12,16,20,21,24,41 Wada testing of-
requires invasive cerebral angiography, with a compli- ten assists in predicting prognosis and counseling pa-
cation rate of approximately 3% (major complications tients and families regarding the risks to memory and
include groin hematoma, vascular injury, and stroke). cognitive function of proposed epilepsy surgery.129
Wada testing is safe in children.16,20,21,126,127 However,
an alert and cooperative patient is required. In chil-
dren younger than 10 years, the sensitivity is signifi- INTRAOPERATIVE CORTICAL
cantly diminished (46–86%), although successful Wada STIMULATION
testing for language dominance has been reported in
children as young as 3-years-old.16,20,21,126 Patient coach- Intraoperative cortical and subcortical stimulation to map
ing before the procedure and propofol anesthesia dur- motor cortex is performed routinely in the anesthetized
ing groin puncture and initial angiography can improve pediatric patient to guide the resection of lesions near
cooperation with functional testing.126 There is a very sensorimotor cortex.2,7−9,131 In addition, bipolar stimu-
low false-positive rate, and successful determination of lation can map motor nuclei on the floor of the fourth
hemispheric dominance in children correlates well with ventricle,132 which may enhance the safety of tumor re-
ECS.20 section in the dorsal brainstem.
The Wada procedure cannot localize eloquent func- The most common method of direct electrocorti-
tions within the hemisphere or define their anatomic cal and subcortical stimulation (ECS) employs a bipo-
relationship to pathology.89,92 This is important in chil- lar electrode with 5 mm between contacts, delivering a
dren who often have functional reorganization due to train of biphasic square pulses at 50–60 Hz with a single
chronic seizures, low-grade neoplasms, or congenital pulse duration of 1 millisecond. When strip electrodes
lesions.12,41,89,128 Most pediatric Wada studies report a are used, contacts are typically 1 cm apart (Fig. 13–3).
high rate of right hemisphere dominance, or signif- Current is increased in a stepwise fashion from 2 to 16
icant bilateral speech representation.16,20,21 This sup- mA until motor function is detected via electromyogra-
ports fMRI and MEG studies in children with chronic phy or direct visual observation of muscular movements.
178 SECTION I TECHNIQUES

Lower stimulation currents are used to map descending ated in the pediatric population, and successful extra-
subcortical motor pathways.8,19,132 Using this method, operative language and motor mapping have been re-
current spread is less than 3 mm, allowing precise def- ported in children as young as 4 years and 18 months,
inition of functional regions.132 While motor mapping respectively.2,17,18,22,134,141,142 Stimulation is most often
under general anesthesia is reliable, stimulation map- performed with a continuous train of stimulation at
ping of sensorimotor cortex requires higher stimulation 50 Hz, single pulse duration 0.3 milliseconds, for 3–5
currents (4–16 mA); this technique is more sensitive in seconds (Fig. 13–4).17
the awake patient,133 where much lower stimulation cur- Studies in children indicate iVEEG has a sensitivity
rents are usually sufficient.132 of approximately 90%, which may be lower in extratem-
A limiting factor of ECS is seizures, which occur in poral or cryptogenic epilepsy.73,75,143,144 Specific indica-
as many as 20% of patients undergoing this procedure.132 tions for invasive monitoring in children vary consid-
Anticonvulsants are not fully protective. Seizures are erably between epilepsy centers. Generally, candidates
usually stopped by irrigation of the cortical surface with have discordant noninvasive data, multifocal pathology,
cold lactated ringer’s solution.132 Short-acting GABA ag- a defined ictal onset zone in near eloquent regions, or
onists may also be effective, but additional mapping may a structural lesion with chronic seizures, where the ic-
often need to be deferred.8,19,132 An alternative to bipo- tal onset zone is often localized to cortex around the
lar stimulation is multipulse stimulation, which may have lesion.145 Quantitative spectral analysis of peri-ictal EEG
several advantages. A lower stimulation frequency of changes may improve the sensitivity of this technique in
2 Hz is used to deliver a short, discrete train of stim- children.135
uli (5–7 pulses of 1 millisecond each), rather than con- Extraoperative ECS of sensorimotor and language
tinuous stimulation. This procedure has a lower risk of function is less sensitive in children younger than 10
seizures and can continuously monitor descending mo- years, compared to older children and adults.8,16,17,19,134
tor pathways during resection.132 Studies show an inverse relationship between age and
Similar to Wada testing and fMRI, ECS of both senso- current thresholds required for motor stimulation, a find-
rimotor and language function is less reliable in children ing magnified in children with disorders of neuronal
less than the age of 10 years.16,134 In children younger migration.132 One possible explanation is a higher per-
than 5 years, the cortex is less excitable. Because of this, centage of small, unmyelinated fibers, requiring an in-
motor mapping via direct cortical stimulation is less sen- creased charge density to elicit a response to stimu-
sitive in young children compared to adults, and suc- lation. The charge density is inversely proportional to
cessful mapping may not be possible.8,16,19,132 Children the area being stimulated, determined by the distance
younger than 1 year typically require very high current between electrodes with bipolar stimulation.16,17 Thus,
thresholds for functional activation, significantly increas- placing the contacts closer together in implanted subdu-
ing the risk of seizures.132,134 If ECS is unsuccessful, the ral electrode arrays increases the charge density without
central sulcus may be identified via phase reversal of so- increasing the applied current.16 We use 64-contact grids
matosensory evoked potentials, which has a sensitivity with 5 mm between electrodes (Adtech, Inc., Racine, WI)
of greater than 90%.8,132 in children. In addition, stimulation protocols that vary
both current intensity and pulse duration may improve
the sensitivity of extraoperative functional mapping in
EXTRAOPERATIVE CORTICAL young children.17 Cortical plasticity and reorganization
STIMULATION in children with chronic seizures, a congenital malfor-
mation, or a brain tumor may lead to negative map-
Extraoperative functional mapping is performed at the pings, because functional centers may not be covered by
bedside by stimulation across implanted subdural elec- electrodes.16,19 Preoperative noninvasive mapping with
trode arrays (2.5 mm platinum electrodes 5–10 mm apart MEG, fMRI, or PET may overcome this limitation.
mounted on silastic and implanted via craniotomy), and Extraoperative functional mapping carries the risks
is a well-established alternative to intraoperative corti- inherent to a second operation for electrode implanta-
cal stimulation. This technique is used in children who tion and prolonged invasive monitoring.126 In addition
are candidates for seizure focus or tumor resection, and to the general risks of additional surgery and anesthesia,
allows prolonged extraoperative iVEEG monitoring.2,140 studies of grid implantations in children report a com-
Functional mapping can demonstrate the relationship plication rate of 10–15%.142,146−150 The most common
between the ictal onset zone and eloquent cortex. In complications include CSF leak and positive CSF culture
young children or patients with developmental delay, in the absence of clinically evident meningitis.22,142,149
awake craniotomy for language mapping may not be Other complications include transient neurological
feasible. The implantation of subdural electrodes allows deficit, cerebral edema, epidural or subdural hematoma,
sensorimotor and language mapping in the awake pa- and stroke. However, most complications do not cause
tient under more comfortable conditions outside of the morbidity or require treatment.142,147 Complications are
operating room. This technique is safe and well toler- less frequent in children compared to adults, and
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 179

Figure 13–4. Ictal focus map and functional map generated from extraoperative
monitoring and stimulation across an implanted subdural grid and intraparenchymal
depth electrodes. The maps clearly delineate the anatomical relationship of a left
temporal tumor to the ictal onset zone, regions of rapid seizure spread, and interictal
spikes (A) as well as functional language cortex (B). (AT: anterior temporal; MT: mesial
temporal).

complication rates decrease with increasing surgical rays have revealed focal, transient inflammation in the
experience.146,148−150 Factors correlated with complica- vast majority of patients, but the severity of this reac-
tions include a large number of electrodes, a long du- tion does not correlate with the incidence of infection
ration of extraoperative monitoring, and left-sided or or long-term surgical outcome.152
bilateral electrode placement.146 Low-grade fevers or To minimize the risks from subdural electrode
aseptic meningitis may occur more frequently.150 Three- placement and chronic invasive monitoring, we employ
stage procedures with two extraoperative monitoring pe- meticulous hemostasis at each operative stage, subdu-
riods does not appear to significantly increase the com- ral and epidural surveillance cultures at each operative
plication rate in children.22 stage, autograft duraplasty using pericranium or tempo-
Dexamethasone is effective at reducing cerebral ralis fascia, watertight dural closure around the electrode
edema in children with implanted subdural grid elec- cables, tunneling of the electrode cables through a sepa-
trode arrays. However, it also may increase the risk of rate stab incision in the scalp, purse-string sutures placed
infection and potentially decrease the seizure frequency, around each cable at the skin, continuous subgaleal
leading to longer extraoperative monitoring periods to drainage for the duration of each monitoring period, and
capture sufficient data to localize the ictal focus.151 broad-spectrum antibiotics for 72 hours perioperatively
Pathological studies of cortex underlying subdural ar- for all procedures after the initial implantation. The bone
180 SECTION I TECHNIQUES

flap is replaced in-situ during monitoring, and reaffixed Conversely, invasive mapping techniques are also chal-
with titanium miniplates after the final operative stage.144 lenging to apply in children, due to higher thresholds
for cortical activation with stimulation and sustained co-
operation required for the Wada test. Despite these chal-
AWAKE CRANIOTOMY IN CHILDREN lenges, the thoughtful combination of noninvasive and
invasive techniques in an experienced center is often
Intraoperative awake language mapping in children highly effective and useful in clinical mapping of brain
presents many challenges.13,14,130,136 Although possible function in children.
with an experienced surgical team in a select group
of cooperative children,130 it lacks safety and reliabil-
ity in children less than the age of 12 years.8,13,14 Prob- REFERENCES
lems include pain, anxiety, nausea, and seizures.137 In
adults, the complication rate is approximately 16.5%, 1. Billingsley-Marshall RL, Simos PG, Papanicolaou AC. Re-
similar to that for staged resection with extraoperative liability and validity of functional neuroimaging tech-
mapping.138 The youngest patient reported to undergo niques for identifying language-critical areas in children
awake craniotomy with successful language mapping and adults. Dev Neuropsychol 2004;26:541.
was 9 years old.130 Recent advancements include the 2. Duffau H. Lessons from brain mapping in surgery for low-
use of dexmedetomidine (␣2 -adrenergic receptor ago- grade glioma: insights into associations between tumour
nist) during the awake portion of the procedure to re- and brain plasticity. Lancet Neurol 2005;4:476.
duce anxiety without excessive sedation, as well as use 3. Hertz-Pannier L, Chiron C, Vera P, et al. Functional
imaging in the work-up of epilepsy. Childs Nerv Syst
of the laryngeal mask airway.8,13,14,136 Successful awake
2001;17:223.
intraoperative language mapping has been performed in 4. Munson S, Schroth E, Ernst M. The role of func-
children as young as 12 years with dexmedetomidine.13 tional neuroimaging in pediatric brain injury. Pediatrics
2006;117:1372.
5. O’Tuama LA, Dickstein DP, Neeper R, et al. Functional
ELECTROCORTICOGRAPHY brain imaging in neuropsychiatric disorders of childhood.
J Child Neurol 1999;14:207.
While iVEEG via implanted intracranial electrode arrays 6. Friederici AD. The neural basis of language development
can capture multiple ictal events, intraoperative ictal fo- and its impairment. Neuron 2006;52:941.
cus mapping by ECoG is limited to interictal data un- 7. Berger MS. Functional mapping-guided resection of low-
der general anesthesia, which significantly diminishes grade gliomas. Clin Neurosurg 1995;42:437.
spike frequency.135 Technically, this procedure is sim- 8. Berger MS, Kincaid J, Ojemann GA, et al. Brain mapping
ilar in adults and children. Electrodes are placed over techniques to maximize resection, safety, and seizure con-
the area of interest and electrical activity is recorded. It is trol in children with brain tumors. Neurosurg 1989;25:786.
generally reliable if an interictal spike frequency reaches 9. Berger MS, Ojemann GA. Intraoperative brain-mapping
techniques in neuro-oncology. Stereotact Funct Neuro-
at least 1 spike per minute.135 This technique is widely
surg 1992;58:153.
applied in children with brain tumors and refractory 10. Keles GE, Berger MS. Advances in the neurosurgical tech-
epilepsy, where the ictal onset zone is frequently local- nique in the current management of brain tumors. Semin
ized to cortex adjacent to the tumor,19,139 and the resec- Oncol 2004;31:659.
tion of perilesional cortex with abnormal interictal spike 11. Fandino J, Kollias SS, Wieser HG, et al. Intraoperative
activity correlates with long-term seizure freedom.139 validation of functional magnetic resonance imaging and
cortical reorganization patterns in patients with brain tu-
mors involving the primary motor cortex. J Neurosurg
䉴 CONCLUSION 1999;91:238.
12. Hertz-Pannier L, Chiron C, Jambaque I, et al. Late plasticity
Mapping of cortical function and dysfunction in children for language in a child’s non-dominant hemisphere: a pre-
with cerebral pathology remains a challenge. The advent and post-surgery fMRI study. Brain 2002;125:361.
and evolution of a variety of noninvasive techniques 13. Ard J, Doyle W, Bekker A. Awake craniotomy with
including fMRI, MEG, PET, SPECT, and DWI or DTI dexmedetomidine in pediatric patients. J Neurosurg Anes-
provide attractive alternatives to the traditional invasive thesiol 2003;15:263.
14. Ard J, Doyle W, Bekker A. Dexmedetomidine in awake
methods of direct cortical stimulation (invasive monitor-
craniotomy: a technical note. Surg Neurol 2005;63:114.
ing with intracranial electrodes and the Wada test). How- 15. Kotsoni E, Byrd D, Basey BJ. Special considerations for
ever, despite promising data, children provide unique functional magnetic resonance imaging of pediatric pop-
challenges that often limit the sensitivity of noninva- ulations. J Magn Reson Imaging 2006;23:877.
sive tests, including patient cooperation and the require- 16. Schevon CA, Carlson C, Zaroff CM, et al. Pediatric lan-
ment for sedation, and differences in CBF regulation and guage mapping: sensitivity of neurostimulation and wada
cortical maturation, especially in very young children. testing in epilepsy surgery. Epilepsia 2007;48:539.
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 181

17. Jayakar P, Alvarez LA, Duchowny MS, et al. A safe and ef- stimulation in a dose-dependent manner: a fMRI study.
fective paradigm to functionally map the cortex in child- Acta Anaesthesiol Scand 2005;49:784.
hood. J Clin Neurophysiol 1992;9:288. 35. Souweidane MM, Kim KHS, McDowall R, et al. Brain map-
18. Jayakar P, Duchowny MS, Resnick TJ. Subdural monitor- ping in sedated infants and young children with passive-
ing in the evaluation of children for epilepsy surgery. J functional magnetic resonance imaging. Pediatr Neuro-
Child Neurol 1994;9:61. surg 1999;30:86.
19. Ojemann SG, Berger MS, Lettich E, et al. Localization of 36. Chapman PH, Buchbinder BR, Cosgrove GR, et al. Func-
language function in children: results of electrical stimu- tional magnetic resonance imaging for cortical mapping
lation mapping. J Neurosurg 2003;98:465. in pediatric neurosurgery. Pediatr Neurosurg 1995;23:122.
20. Hinz AC, Berger MS, Ojemann GA, et al. The utility of the 37. Stapleton SR, Kiriakopoulos E, Mikulis D, et al. Combined
intracarotid amytal procedure in determining hemispheric utility of functional MRI, cortical mapping, and frameless
speech lateralization in pediatric epilepsy patients under- stereotaxy in the resection of lesions in eloquent areas of
going surgery. Childs Nerv Syst 1994;10:239. brain in children. Pediatr Neurosurg 1997;26:68.
21. Jansen FE, Jennekens-Schinkel A, Van Huffelen AC, et al. 38. Martin E, Marcar VL. Functional MR imaging in pediatrics.
Diagnostic significance of Wada procedure in very young Magn Reson Imaging Clin N Am 2001;9:231.
children and children with developmental delay. Eur J 39. Dymarkowski S, Sunaert S, Van Oostende S, et al. Func-
Paediatr Neurol 2002;6:315. tional MRI of the brain: localisation of eloquent cortex in
22. Bauman JA, Fioli E, Romanelli P, et al. Multistage epilepsy focal brain lesion therapy. Eur Radiol 1998;8:1573.
surgery: safety, efficacy, and utility of a novel approach 40. Liegeois F, Cross JH, Gadian DG, et al. Role of fMRI in the
in pediatric extratemporal epilepsy. Neurosurg 2005;56: decision-making process: epilepsy surgery for children. J
318. Magn Reson Imaging 2006;23:933.
23. Bookheimer SY, Dapretto M, Karmarkar U. Functional 41. Fair DA, Brown TT, Petersen SE, et al. fMRI reveals novel
MRI in children with epilepsy. Dev Neurosci 1999;21:191. functional neuroanatomy in a child with perinatal stroke.
24. Holodny AI, Schulder M, Liu WC, et al. The effect of brain Neurology 2006;67:2246.
tumors on BOLD functional MR imaging activation in the 42. Benedict SL. Diffusion-weighted imaging in diagnosing
adjacent motor cortex: implications for image-guided neu- neurological disorders in children: a pediatric neurolo-
rosurgery. AJNR Am J Neuroradiol 2000;21:1415. gist’s perspective. Pediatr Radiol 2007;37:734.
25. Roessler K, Donat M, Lanzenberger R, et al. Evaluation 43. Robertson RL, Glasier CM. Diffusion-weighted imaging
of preoperative high magnetic field motor functional MRI of the brain in infants and children. Pediatr Radiol
(3 Tesla) in glioma patients by navigated electrocortical 2007;37:749.
stimulation and postoperative outcome. J Neurol Neuro- 44. Ichord RN, Naim M, Pollock AN, et al. Hypoxix-ischemic
surg Psychiatry 2005;76:1152. injury complicates inflicted and accidental traumatic brain
26. Schiffbauer H, Berger MS, Ferrari P, et al. Preoperative injury in young children: the role of diffusion-weighted
magnetic source imaging for brain tumor surgery: a quan- imaging. J Neurotrauma 2007;24:106.
titative comparison with intraoperative sensory and motor 45. Covarrubias DJ, Luetmer PH, Campeau NG. Posterior re-
mapping. J Neurosurg 2002;97:1333. versible encephalopathy syndrome: prognostic utility of
27. Kim SK, Wang KC, Hwang YS, et al. Intractable epilepsy quantitative diffusion-weighted MR images. AJNR Am J
associated with brain tumors in children: surgical modality Neuroradiol 2002;23:1038.
and outcome. Childs Nerv Syst 2001;17:445. 46. Axer H, Ragoschke-Schumm A, Bottcher J, et al. Initial
28. Thomason ME, Burrows BE, Gabrieli JD, et al. Breath DWI and ADC imaging may predict outcome in acute
holding reveals differences in fMRI BOLD signal in chil- disseminated encephalomyelitis: report of two cases of
dren and adults. Neuroimage 2005;25:824. brainstem encephalitis. J Neurol Neurosurg Psychiatry
29. Byars AW, Holland SK, Strawsburg RH, et al. Practi- 2005;76:996.
cal aspects of conducting large-scale functional magnetic 47. O’Brien TJ, David EP, Kilpatrick CJ, et al. Contrast-
resonance imaging studies in children. J Child Neurol enhanced perfusion and diffusion MRI accurately laterati-
2002;17:885. aze temporal lobe epilepsy: A pilot study. J Clin Neurosci
30. Slifer KJ, Cataldo MF, Cataldo MD, et al. Behavior analy- 2007;14:841.
sis of motion control for pediatric neuroimaging. J Appl 48. Wozniak JR, Krach L, Ward E, et al. Neurocognitive and
Behav Anal 1993;26:469. neuroimaging correlates of pediatric traumatic brain in-
31. Wilke M, Lidzba K, Staudt M, et al. Comprehensive lan- jury: a diffusion tensor imaging (DTI) study. Arch Clin
guage mapping in children, using functional magnetic Neuropsychol 2007;22:555.
resonance imaging: what’s missing counts. NeuroReport 49. Albayram S, Melhem ER, Mori S, et al. Holoprosencephaly
2005;16:915. in children: diffusion tensor MR imaging of white mat-
32. Richter W, Richter M. The shape of fMRI BOLD response ter tracts of the brainstem – initial experience. Radiology
in children and adults changes systematically with age. 2002;223:645.
Neuroimage 2003;20:1122. 50. Cascio CJ, Gerig G, Piven J. Diffusion tensor imaging: ap-
33. Gaillard WD, Kopylev L, Weinstein S, et al. Low incidence plication to the study of the developing brain. J Am Acad
of abnormal [18]FDG-PET in children with new-onset par- Child Adolesc Psychiatry 2007;46:213.
tial epilepsy: a prospective study. Neurology 2002;58:717. 51. Rollins NK. Clinical applications of diffusion tensor
34. Dueck MH, Petzke F, Gerbershagen HJ, et al. Propofol imaging and tractography in children. Pediatr Radiol
attenuates responses of the auditory cortex to acoustic 2007;37:769.
182 SECTION I TECHNIQUES

52. Tummala RP, Chu RM, Liu H, et al. Application of diffu- 70. Burneo JG, Kuzniecky RI, Bebin M, et al. Cortical reorga-
sion tensor imaging to magnetic-resonance-guided brain nization in malformations of cortical development: a mag-
tumor resection. Pediatr Neurosurg 2003;39:39. netoencephalographic study. Neurology 2004;63:1818.
53. Huisman TAGM, Sorenson AG. Perfusion-weighted mag- 71. Lutkenhoner B. Magnetoencephalography and its
netic resonance imaging of the brain: techniques and ap- Achilles’ heel. J Physiol Paris 2003;97:641.
plication in children. Eur Radiol 2004;14:59. 72. RamachandranNair R, Otsubo H, Shroff MM, et al.
54. Ashwal S, Babikian, T, Gardner-Nichols J, et al. MEG predicts outcome following surgery for intractable
Susceptibility-weighted imaging and proton magnetic res- epilepsy in children with normal or nonfocal MRI find-
onance spectroscopy in assessment of outcome after pe- ings. Epilepsia 2007;48:149.
diatric traumatic brain injury. Arch Phys Med Rehabil 73. Wheless JW, Willmore LJ, Breier JI, et al. A comparison
2007;87:S50. of magnetoencephalography, MRI, and V-EEG in patients
55. Firsching R, Bondar I, Heinz HJ, et al. Practicability of evaluated for epilepsy surgery. Epilepsia 1999;40:931.
magnetoencephalography-guided neuronavigation. Neu- 74. Otsubo H, Ochi A, Elliott I, et al. MEG predicts epileptic
rosurg Rev 2002;25:73. zone in lesional extrahippocampal epilepsy: 12 pediatric
56. Minassian BA, Otsubo H, Weiss S, et al. Magnetoen- surgery cases. Epilepsia 2001;42:1523.
cephalographic localization in pediatric epilepsy surgery: 75. Papanicolaou AC, Pataraia E, Billingsley-Marshall R, et al.
comparison with invasive intracranial electroencephalog- Toward the substitution of invasive electroencephalog-
raphy. Ann Neurol 1999;46:627. raphy in epilepsy surgery. J Clin Neurophysiol 2005;22:
57. Lee D, Sawrie SM, Simos PG, et al. Reliability of lan- 231-7.
guage mapping with magnetic source imaging in epilepsy 76. Knowlton RC, Elgavish R, Howell J, et al. Magnetic
surgery candidates. Epilepsy Behav 2006;8:742. source imaging versus intracranial electroencephalogram
58. Jansen FE, Huiskamp G, van Huffelen AC, et al. Identifica- in epilepsy surgery: a prospective study. Ann Neurol
tion of the epileptogenic tuber in patients with tuberous 2006;59:835.
sclerosis: a comparison of high-resolution EEG and MEG. 77. Yoshinaga H, Ohtsuka Y, Watanabe Y, et al. Ictal MEG in
Epilepsia 2006;47:108. two children with partial seizures. Brain Dev 2004;26:403.
59. Chuang NA, Otsubo H, Pang EW, et al. Pediatric magne- 78. Oishi M, Kameyama S, Masuda H, et al. Single and multi-
toencephalography and magnetic source imaging. Neu- ple clusters of magnetoencephalographic dipoles in neo-
roimaging Clin N Am 2006;16:193. cortical epilepsy: significance in characterizing the epilep-
60. Forss N. Magnetoencephalography (MEG) in epilepsy togenic zone. Epilepsia 2006;47:355.
surgery. Acta Neurochir Suppl 1997;68:81. 79. Knowlton RC, Laxer KD, Aminoff MJ, et al. Magnetoen-
61. Balakrishnan G, Grover KM, Mason K, et al. A retrospec- cephalography in partial epilepsy: clinical yield and local-
tive analysis of the effect of general anesthetics on the ization accuracy. Ann Neurol 1997;42:622.
successful detection of interictal epileptiform activity in 80. Hashizume A, Iida K, Shirozu H, et al. Gradient magnetic-
magnetoencephalography. Anesth Analg 2007;104:1493. field topography for dynamic changes of epileptic dis-
62. Szmuk P, Kee S, Pivalizza EG, et al. Anaesthesia for charges. Brain Res 2007;1144:175.
magnetoencephalography in children with intractable 81. Ramantani G, Boor R, Paetau R, et al. MEG versus EEG:
seizures. Paediatr Aneaesth 2003;13:811. influence of background activity on interictal spike detec-
63. Paetau R, Hamalainen M, Hari R, et al. Magnetoencephalo- tion. J Clin Neurophysiol 2006;23:498.
graphic evaluation of children and adolescents with in- 82. Wu JY, Sutherling WW, Koh S, et al. Magnetic source imag-
tractable epilepsy. Epilepsia 1994;35:275. ing localizes epileptogenic zone in children with tuberous
64. Xiang J, Holowka S, Qiao H, Sun B, Chuang S. Volumetric sclerosis complex. Neurology 2006;66:1270.
estimation of functional brain regions in small children us- 83. Kamimura T, Tohyama J, Oishi M, et al. Magnetoen-
ing spatially filtered magnetoencephalography: differenti- cephalography in patients with tuberous sclerosis and
ating thumb from middle finger. Neurol Clin Neurophysiol localization-related epilepsy. Epilepsia 2006;47:991.
2004;110. 84. Iida K, Otsubo H, Mohamed IS, et al. Characterizing
65. Bast T, Wright T, Boor R, et al. Combined EEG and MEG magnetoencephalographic spike sources in children with
analysis of early somatosensory evoked activity in chil- tuberous sclerosis complex. Epilepsia 2005;46:1510.
dren and adolescents with focal epilepsies. Clin Neuro- 85. Sobel DF, Aung M, Otsubo H, et al. Magnetoencephalog-
physiol 2007;118:1721. raphy in children with Landau-Kleffner syndrome and
66. Papanicolaou AC, Simos PG, Castillo EM, et al. Magne- acquired epileptic aphasia. AJNR Am J Neuroradiol
toencephalography: a noninvasive alternative to the Wada 2000;21:301.
procedure. J Neurosurg 2004;100:867. 86. Paetau R, Granstrom ML, Blomstedt G, et al. Magnetoen-
67. Papanicolaou AC, Pazo-Alvarez P, Castillo EM, et al. Func- cephalography in presurgical evaluation of children with
tional neuroimaging with MEG: normative language pro- Landau-Kleffner syndrome. Epilepsia 1999;40:326.
files. Neuroimage 2006;33:326. 87. Lin YY, Chang KP, Hsieh JC, et al. Magnetoencephalo-
68. Papanicolaou AC, Simos PG, Breier JI, et al. Magnetoen- graphic analysis of bilaterally synchronous discharges
cephalographic mapping of the language-specific cortex. in benign rolandic epilepsy of childhood. Seizure
J Neurosurg 1999;90:85. 2003;12:448.
69. Rezai AR, Mogilner AY, Cappell J, et al. Integration of func- 88. Hattori H, Yamano T, Tsutada T, et al. Magnetoen-
tional brain mapping in image-guided neurosurgery. Acta cephalography in the detection of focal lesions in West
Neurochir Suppl 1997;68:85. syndrome. Brain Dev 2001;23:528.
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 183

89. Duncan JD, Moss SD, Bandy DJ, et al. Use of positron 104. Lee SK, Lee SY, Kim KK, et al. Surgical outcome and prog-
emission tomography for presurgical localization of elo- nostic factors of cryptogenic neocortical epilepsy. Ann
quent brain areas in children with seizures. Pediatr Neu- Neurol 2005;58:525.
rosurg 1997;26:144. 105. Sood S, Chugani HT. Functional neuroimaging in the
90. Juhasz C, Chugani HT. Imaging the epileptic brain with preoperative evaluation of children with drug-resistant
positron emission tomography. Neuroimag Clin N Am epilepsy. Childs Nerv Syst 2006;22:810.
2003;13:705. 106. Lee JJ, Kang WJ, Lee DS, et al. Diagnostic performance of
91. Pirotte B, Goldman S, Salzberg S, et al. Combined positron 18F-FDG PET and ictal 99mTc-HMPAO SPET in pediatric
emission tomography and magnetic resonance imaging temporal lobe epilepsy: quantitative analysis by statisti-
for the planning of stereotactic brain biopsies in children: cal parametric mapping, statistical probabilistic anatomi-
experience in 9 cases. Pediatr Neurosurg 2003;38:146. cal map, and subtraction ictal SPET. Seizure 2005;14:213.
92. Kaplan AM, Bandy DJ, Manwaring KH, et al. Functional 107. Park CK, Kim SK, Wang KC, et al. Surgical outcome and
brain mapping using positron emission tomography scan- prognostic factors of pediatric epilepsy caused by cortical
ning in preoperative neurosurgical planning for pediatric dysplasia. Childs Nerv Syst 2006;22:586.
brain tumors. J Neurosurg 1999;91:797. 108. Metsahonkala L, Gaily E, Rantala H, et al. Focal and
93. Ruotsalainen U, Suhonen-Polvi H, Eronen E, et al. Esti- global cortical hypometabolism in patients with newly di-
mated radiation dose to the newborn in FDG-PET studies. agnosed infantile spasms. Neurology 2002;58:1646.
J Nucl Med 1996;37:387. 109. Itomi K, Okumura A, Negoro T, et al. Prognostic value of
94. Suhonen-Polvi H, Ruotsalainen U, Kinnala A, et al. FDG- positron emission tomography in cryptogenic West syn-
PET in early infancy: simplified quantification meth- drome. Dev Med Child Neurol 2002;44:107.
ods to measure cerebral glucose utilization. J Nucl Med 110. Chandra PS, Salamon N, Huang J, et al. FDG-PET/MRI
1995;36:1249. coregistration and diffusion-tensor imaging distinguish
95. Juengling FD, Kassubek J, Martens-Le Bouar H, et al. epileptogenic tubers and cortex in patients with tuber-
Cerebral regional hypometabolism caused by propofol- ous sclerosis complex: a preliminary report. Epilepsia
induced sedation in children with severe myoclonic 2006;47:1543.
epilepsy: a study using fluorodeoxyglucose positron emis- 111. Hamano SI, Yoshinari S, Higurashi N, et al. Regional cere-
sion tomography and statistical parametric mapping. Neu- bral blood flow and developmental outcome in crypto-
rosci Lett 2002;335:79. genic West syndrome. Epilepsia 2007;48:114.
96. Korinthenberg R, Bauer-Scheid C, Burkart P, et al. 112. Maria BL, Drane WE, Mastin ST, et al. Comparative value
[18]FDG-PET in epilepsies of infantile onset with phar- of thallium and glucose SPECT imaging in childhood brain
macoresistant generalised tonic-clonic seizures. Epilepsy tumors. Pediatr Neurol 1998;19:351.
Res 2004;60:53. 113. Barai S, Bandopadhayaya GP, Julka PK, et al. Evaluation
97. Utriainen M, Metsahonkala L, Salmi TT, et al. Metabolic of Tc99m-glucoheptonate for SPECT functional imaging
characterization of childhood brain tumors: comparison of medulloblastoma. J Clin Neurosci 2005;12:36.
of 18F-fluorodeoxyglucose and [11C]-methionine positron 114. Van Paesschen W. Ictal SPECT. Epilepsia 2004;45:35.
emission tomography. Cancer 2002;95:1376. 115. Lee DS, Lee SK, Kim YK, et al. Superiority of HMPAO ictal
98. Borgwardt L, Hojgaard L, Carstensen H, et al. Increased SPECT to ECD ictal SPECT in localizing the epileptogenic
fluorine-18 2-fluoro-2-deoxy-D-glucose (FDG) uptake in zone. Epilepsia 2002;43:263.
childhood CNS tumors is correlated with malignancy 116. So Y, Lee HY, Kim SK, et al. Prediction of the clinical
grade: a study with FDG positron emission tomogra- outcome of pediatric Moya-moya disease with postopera-
phy/magnetic resonance imaging coregistration and im- tive basal/acetazolamide stress brain perfusion SPECT af-
age fusion. J Clin Oncol 2005;23:3030. ter revascularization surgery. Stroke 2005;36:1485.
99. Pirotte B, Acerbi F, Lubansu A, et al. PET imaging in the 117. Acay AP, Kaya GC, Emiroglu NI, et al. Effects of long-term
surgical management of pediatric brain tumors. Childs methylphenidate treatment: a pilot follow-up clinical and
Nerv Syst 2007;23:739. SPECT study. Prog Neuropsychopharmacol Biol Psychia-
100. Gururangan S, Hwang E, Herndon JE 2nd, et al. try 2006;30:1219.
[18 F]fluorodeoxyglucose-positron emission tomography in 118. Chiu Wong SB, Chapman SB, Cook LG, et al. A SPECT
patients with medulloblastoma. Neurosurgery 2004;55: study of language and brain reorganization three years
1280. after pediatric brain injury. Prog Brain Res 2006;157:173.
101. Ohta Y, Nariai T, Ishii K, et al. Meningio-angiomatosis in a 119. Kirton A, Kloiber R, Rigel J, et al. Evaluation of pediatric
patient with focal epilepsy: value of PET in diagnoses and CNS malignancies with 99mTc-methoxyisobutylisonitrile
preoperative planning of surgery. Acta Neurochir (Wien) SPECT. J Nucl Med 2002;43:1438.
2003;145:587. 120. Barai S, Bandopadhayaya GP, Julka PK, et al. Evalua-
102. Pirotte B, Levivier M, Morelli D, et al. Positron emission tion of single photon emission computerised tomography
tomography for the early postsurgical evaluation of pedi- (SPECT) using Tc99m-tetrofosmin as a diagnostic modal-
atric brain tumors. Childs Nerv Syst 2005;21:294. ity for recurrent posterior fossa tumors. J Postgrad Med
103. Pirotte B, Goldman S, van Bogaert P, et al. Integration 2003;49:316.
of [11C]methionine-positron emission tomographic and 121. Buchhalter JR, So EL. Advances in computer-assisted
magnetic resonance imaging for image-guided surgical re- single-photon emission computed tomography (SPECT)
section of infiltrative low-grade brain tumors in children. for epilepsy surgery in children. Acta Paediatr Suppl
Neurosurgery 2005;57:ONS128. 2004;93:32.
184 SECTION I TECHNIQUES

122. Chugani HT. What can we learn from functional neu- 141. Ikeda A, Miyamoto S, Shibasaki H. Cortical motor
roimaging in children? Indian Pediatr 2006;43:203. mapping in epilepsy patients: information from subdu-
123. Ahnlide JA, Rosen I, Linden-Mickelsson Tech P, et al. ral electrodes in presurgical evaluation. Epilepsia 2002;
Does SISCOM contribute to favorable seizure outcome 43:56.
after epilepsy surgery? Epilepsia 2007;48:479. 142. Onal C, Otsubo H, Araki T, et al. Complications of inva-
124. Lee SK, Lee SY, Yun CH, et al. Ictal SPECT in neocor- sive subdural grid monitoring in children with epilepsy. J
tical epilepsies: clinical usefulness and factors affecting Neurosurg 2003;98:1017.
the pattern of hyperperfusion. Neuroradiology 2006;48: 143. Schiller Y, Cascino GD, Sharbrough FW. Chronic in-
678. tracranial EEG monitoring for localizing the epileptogenic
125. Fukuda M, Masuda H, Honma J, et al. Ictal SPECT an- zone: ana electroclinical correlation. Epilepsia 1998;39:
alyzed by three-dimensional stereotactic surface projec- 1302.
tion in frontal lobe epilepsy patients. Epilepsy Res 2006; 144. Bruce DA, Bizzi JWJ. Surgical technique for the insertion
68:95. of grids and strips for invasive monitoring in children with
126. Masters LT, Perrine K, Devinsky O, et al. Wada testing in intractable epilepsy. Childs Nerv Syst 2000;16:724.
pediatric patients by use of propofol anesthesia. AJNR Am 145. Jayakar P. Invasive EEG monitoring in children: when,
J Neuroradiol 2000;21:1302. where, and what? J Clin Neurophysiol 1999;16:408.
127. Szabo CA, Wyllie E. Intracarotid amobarbital testing for 146. Hamer HM, Morris HH, Mascha EJ, et al. Complications
language and memory dominance in children. Epilepsy of invasive video-EEG monitoring with subdural grid elec-
Res 1993;15:239. trodes. Neurology 2002;58:97.
128. Branco DM, Coelho TM, Branco BM, et al. Functional vari- 147. Johnston JM Jr, Mangano FT, Ojemann JG, et al. Complica-
ability of the human cortical motor map: electrical stim- tions of invasive subdural electrode monitoring at St. Louis
ulation findings in perirolandic epilepsy surgery. J Clin Children’s Hospital, 1994–2005. J Neurosurg 2006;105:
Neurophysiol 2003;20:17. 343.
129. Lee GP, Park YD, Westerveld M, et al. Wada memory per- 148. Rydenhag B, Silander HC. Complications of epilepsy
formance predicts seizure outcome after epilepsy surgery surgery after 654 procedures in Sweden, September 1990–
in children. Epilepsia 2003;44:936. 1995: a multicenter study based on the Swedish National
130. Klimek M, Verbugge SJ, Roubos S, et al. Awake cran- Epilepsy Surgery Register. Neurosurg 2001;49:51.
iotomy for glioblastoma in a 9-year-old child. Anaesthesia 149. Simon SL, Telfeian A, Duhaime AC. Complications of in-
2004;59:607. vasive monitoring used in intractable pediatric epilepsy.
131. Berger MS, Rostomily RC. Low-grade gliomas: functional Pediatr Neurosurg 2003;38:47.
mapping resection strategies, extent of resection, and out- 150. Swartz BE, Rich JR, Dwan PS, et al. The safety and efficacy
come. J Neuroncol 1997;34:85. of chronically implanted subdural electrodes: a prospec-
132. Sala F, Krzan MJ, Deletis V. Intraoperative neurophysio- tive study. Surg Neurol 1996;46:87.
logical monitoring in pediatric neurosurgery: why, when, 151. Araki T, Otsubo H, Makino Y, et al. Efficacy of dexam-
how? Childs Nerv Syst 2002;18:264. ethasone on cerebral swelling and seizures during sub-
133. Vtiz TW, Marx W, Victor JD, et al. Comparison of con- dural grid EEG recording in children. Epilepsia 2006;47:
scious sedation and general anesthesia for motor map- 176.
ping and resection of tumors located near motor cortex. 152. Stephan CL, Kepes JJ, SantaCruz K, et al. Spectrum
Neurosurg Focus 2003;15:E8. of clinical and histopathologic responses to intracranial
134. Chitoku S, Otsubo H, Harada Y, et al. Extraoperative corti- electrodes: from multifocal aseptic meningitis to mul-
cal stimulation of motor function in children. Pediatr Neu- tifocal hypersensitivity-type meningovasculitis. Epilepsia
rol 2001;24:344. 2001;42:895.
135. Asano E, Muzik O, Shah A, et al. Quantitative visualization 153. Ganslandt O, Buchfelder M, Hastreiter P, et al. Mag-
of ictal subdural EEG changes in children with neocortical netic source imaging supports clinical decision making
focal seizures. Clin Neurophysiol 2004;115:2718. in glioma patients. Clin Neurol Neurosurg 2004;107:20.
136. Hagberg CA, Gollas A, Berry JM. The laryngeal mask air- 154. Haglund MM. Overview of functional imaging. Neurosurg
way for awake craniotomy in the pediatric patient: report Clin N Am 1997;8:287.
of three cases. J Clin Anesth 2004;16:43. 155. Haglund MM, Berger MS, Shamseldin M, et al. Cortical
137. Costello TG, Cormack JR. Anaesthesia for awake cran- localization of temporal lobe language sites in patients
iotomy: a modern approach. J Clin Neurosci 2004;11:16. with gliomas. Neurosurg 1994;34:567.
138. Taylor MD, Berenstein M. Awake craniotomy with brain 156. Krings T, Reinges MH, Erberich S, et al. Functional MRI
mapping as the routine surgical approach to treating pa- for presurgical planning: problems, artefacts, and solution
tients with intraaxial tumors: a prospective trial of 200 strategies. J Neurol Neurosurg Psychiatry 2001;70:749.
cases. J Neurosurg 1999;90:35. 157. Lee CC, Ward HA, Sharbrough FW, et al. Assessment of
139. Pilcher WH, Silbergeld DL, Berger MS, et al. Intraoperative functional MR imaging in neurosurgical planning. AJNR
electrocorticography during tumor resection: impact on Am J Neuroradiol 1999;20:1511.
seizure outcome in patients with gangliogliomas. J Neu- 158. Ruge MI, Victor J, Hosain S, et al. Concordance be-
rosurg 1993;78:891. tween functional magnetic resonance imaging and intra-
140. Tharin S, Golby A. Functional brain mapping and its appli- operative language mapping. Stereotact Funct Neurosurg
cations to neurosurgery. Neurosurgery 2007;60:ONS-185. 1999;72:95.
CHAPTER 13 PEDIATRIC BRAIN MAPPING: SPECIAL CONSIDERATIONS 185

159. Ment LR, Bada HS, Barnes P, et al. Practice parame- 162. Touho H, Karasawa J, Ohnishi H. Preoperative and
ter: neuroimaging of the neonate: report of the Quality postoperative evaluation of cerebral perfusion and va-
Standards Subcommittee of the American Academy of sodilatory capacity with 99mTc-HMPAO SPECT and ac-
Neurology and the Practice Committee of the Child Neu- etazolamide in childhood Moya-moya disease. Stroke
rology Society. Neurology 2002;58:1726. 1996;27:282.
160. Bonekamp D, Nagae LM, Degaonkar M, et al. Diffusion 163. Devous MD, Altuna D, Furl N, et al. Maturation of speech
tensor imaging in children and adolescents: reproducibil- and language functional neuroanatomy in pediatric nor-
ity, hemispheric, and age-related differences. NeuroImage mal controls. J Speech Lang Hear Res 2006;49:856.
2007;34:733. 164. Najm IM, Bingaman WE, Luders HO. The use of subdu-
161. Mori K, Toda Y, Hashimoto T, et al. Patients with West ral grids in the management of focal malformations due
syndrome whose ictal SPECT showed focal cortical hy- to abnormal cortical development. Neurosurg Clin N Am
perperfusion. Brain Dev 2007;29:202. 2002;13:87.
This page intentionally left blank
SECTION II

SYSTEMS

187
This page intentionally left blank
Chapter 14
Mapping of the Sensorimotor Cortex
Roukoz Chamoun1 , Krishna Satyan1 , and Youssef G. Comair 2
1
Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
2
Department of Neurosurgery, American University of Beirut, Beirut, Lebanon

䉴 INTRODUCTION mapping is typically used to maximize the extent of re-


section of the epileptogenic zone, while at the same
Techniques for mapping motor and sensory areas in time minimizing damage to critical areas for somatomo-
the human brain have progressively and dramatically tor and somatosensory function. In epilepsy cases where
evolved in recent years, benefitting from technological patients undergo invasive monitoring with semichroni-
advances in structural and functional brain imaging and cally implanted subdural electrodes in an epilepsy mon-
improved understanding of the anatomical and neuro- itoring unit, invasive mapping may also be carried out
physiological properties of the brain. Each technique has extraoperatively using the implanted electrodes to de-
its own advantages and limitations, and an understand- fine the location of sensiromotor cortex via recordings
ing of the basic principles of these techniques is essential or with direct electrical stimulation, with the same basic
for proper implementation and correct interpretation. methods employed for intraoperative mapping. A rela-
While in the past accurate identification of mo- tively newer clinical application for mapping of sensiro-
tor and sensory areas required invasive methods, ac- motor cortex is in guiding the insertion of an epidural
curate noninvasive localization of sensorimotor cortex implant of electrodes for chronic motor cortex stimula-
is now routinely available. The location of these areas tion for the management of intractable pain. In these
in individuals can be defined accurately and noninva- cases, the sensiromotor cortex is localized to facilitate
sive both by observing specific imaging landmarks with accurate placement of an electrode array.
structural imaging and by observing regional cortical ac-
tivation with functional imaging. This ability has changed
the way clinicians approach the treatment of pathology 䉴 HISTORICAL PERSPECTIVES
within or near functionally critical motor areas; localiza-
tion of Rolandic cortex can be defined well in advance of Despite early observations that unilateral brain injury
a neurosurgical intervention, which permits better treat- can lead to paralysis on the opposite side of the body,
ment planning and better patient counseling regarding for many years the prevailing theory was that the brain
the risks of a proposed intervention. Furthermore, im- functioned as a single unit without discrete localization
ages from preoperatively obtained maps can be inte- of specific functions. Yet, as early as the first part of
grated into image–guidance systems used for navigation the nineteenth century, clinical observations of brain
during surgery to help guide the procedure as it is per- injury and stroke patients intrigued neurologists and
formed. advanced the theory that specific functions, most no-
In spite of dramatic advances in noninvasive map- tably somatomotor and somatosensory are localized to
ping of motor and sensory areas, invasive methods for specific brain areas.1 The early period of the nine-
mapping remain important in optimizing clinical care. teenth century also witnessed the appearance of medical
Intraoperative mapping of sensiromotor cortex with the applications of electricity to activating and identifying
brain exposed is routinely used as an adjunct in the re- somatomotor cortex. In 1802, Aldini reported strong
section of brain tumors and other structural brain lesions muscle contractions when applying electrical stimulation
that lie within or near the primary sensiromotor cortex. to the dura and deep substance of the brain of an ox.2
This allows surgeons to perform a maximal safe resec- In 1829, Luigi Rolando described the central vertical fis-
tion without damaging nearby critical motor and sensory sure and central area, composed of the precentral and
areas defined by mapping. Invasive mapping of sensiro- postcentral gyri, that still carries his name: the Rolandic
motor cortex is also commonly used in epilepsy surgery, cortex. 3 The acceptance of the modern notion of local-
where the epileptic zone to be resected is often defined ized motor function in the brain was further buttressed
by a physiological signature rather than an anatomically by John Hughlings Jackson, who observed the progres-
identifiable structural lesion. In epilepsy surgery cases, sive spread of motor seizures across the hemibody and

189
190 SECTION II SYSTEMS

theorized that the convolutions of the brain contain or- plosion in mapping of human motor and sensory areas,
derly neural representations of movements.4 In 1870, particularly in the clinical setting, with validation by clas-
Fritsch and Hitzig conducted the first experimentally sical invasive methods.
controlled direct stimulation of mammalian cerebral cor- Further experience in cortical stimulation and
tex, and for the first time, there was an experimen- surgery in the central area in patients, as well as analysis
tal proof of a causal connection between activation of of motor and sensory responses in noninvasive exper-
Rolandic cortex and muscular contractions.5 The first re- iments in normal human subjects and invasive studies
ported case of electric stimulation of the cortex of a hu- in nonhuman primates, has revealed that sensorimotor
man patient was in 1874.6 Robert Bartholow was treat- cortex is more complex in its organization and cannot
ing a patient with an “epithelioma of the scalp” that had be simply reduced to the Penfield homunculus. Many
eroded the skull, and when he applied electric stimula- questions about this organization remained unanswered,
tion to the patient’s exposed cortex, a motor response and our understanding of this region of the brain con-
occurred on the opposite side of the body. tinues to evolve as a number of authors raise questions
The elegant experimental cortical stimulation stud- regarding interindividual variability, mosaicism, redun-
ies of Ferrier in a number of animal species demon- dant representations.14
strated more clearly than ever before that motor
responses were elicited by stimulation of the Rolandic
䉴 ANATOMY
cortex. He also found that electric stimulation at specific
anatomic locations gives rise to predictable movements
The central sulcus is a deep sulcus extending through-
of specific different parts of the body.7 At this time, a
out the convexity and is the key to anatomically iden-
general consensus evolved that the primary motor cor-
tifying the primary sensorimotor cortex. It begins to
tex is located along the central sulcus in the both anterior
develop embryologically at about 26-week gestation,
and posterior banks. In 1901, Grunbaum and Sherring-
about the same time as the cingulate and superior tem-
ton used finer stimulation techniques and found that the
poral sulcus. Unlike the development of the central sul-
primary motor area is largely restricted to the gyrus im-
cus in other primates, the human Rolandic fissure does
mediately anterior to the central sulcus.8
not appear to arise from two separate anlages that unite
William Keen was the first American neurosurgeon
to form a single sulcus. This may explain why it is in-
to perform cortical stimulation in a human patient during
terrupted in only 1% of human brains.15,16 By the 28th
craniotomy.9 Later, Charles Frazier of Philadelphia pub-
week of gestation, the central sulcus covers the entire
lished a map of human brain function that was partly
convexity as a relatively straight line.
based on experience with cortical stimulation during
As per Symington and Crymble (1913)17 , the central
craniotomies.10 Fedor Krause, working in Berlin, per-
sulcus has three genu. The superior and inferior genu
formed stimulation of the central area to map function
are convex anteriorly, while the middle genu is an inter-
and treat epilepsy. He described one of the first detailed
vening concave bend. Broca described three annectant
maps of the motor cortex in humans.11 Otfrid Foerster,
gyri connecting the pre- and postcentral gyri: the “pli de
who began his medical career as a neurologist in Ger-
passage frontoparietal superieur” at the interhemispheric
many, started to treat patients surgically during World
fissure at the level of the paracentral lobule, the “pli
War I, and then became a leading European neurosur-
de passage frontoparietal inferieur” separating the cen-
geon. He performed surgeries under local anesthesia and
tral sulcus from the sylvian fissure at the level of the
used cortical stimulation to localize function. He pro-
subcentral gyrus, and the “pli de passage moyen (PPM)”
duced a detailed map of the brain from the results of
that is mostly hidden within the central sulcus and found
his stimulation work that included sensory responses as
at the level of the superior frontal sulcus.18
well as functions outside the central area.12 Wilder Pen-
The precentral gyrus, or the primary motor cortex,
field spent 6 months with Foerster, and then moved to
consists of four segments:
Montreal and established the Montreal Neurological In-
stitute. Thereafter, the extensive work on human cor- (1) The inferior segment is anteriorly convex, and
tical stimulation by Penfield and Theodore Rasmussen commonly communicates with the postcentral
led to further refinement of our understanding of the gyrus via the pli de passage frontoparietal, giv-
sensorimotor cortex, including the classic description of ing rise to the central operculum.
the somatotopic representation of the motor and sensory (2) The middle segment of the precentral gyrus is
representations along the Rolandic cortex as an “upside- anteriorly concave, but usually does not have a
down man.”13 The more recent emergence of nonin- well-defined anterior border because it blends
vasive methods such as positron emission tomography with the middle frontal gyrus. The middle seg-
(PET), functional magnetic resonance imaging (fMRI), ment is characterized by the PPM, an annectant
and magnetoencephalography (MEG) has led to an ex- gyral structure deep within the central sulcus.
CHAPTER 14 MAPPING OF THE SENSORIMOTOR CORTEX 191

The PPM elevates the floor of the central sul- terior most extent of the genu of the corpus callosum
cus and forms a characteristic omega sign on MRI (Talairach and Bancaud, 1966).23 However, no studies
bulging posteriorly from the precentral gyrus.19,20 have accounted for the variation in the gyral convo-
Recently, Boling et al.21 demonstrated that the lutions of the superior frontal gyrus, and the anterior
whole-hand motor and sensory cortical regions boundary of the SMA remains ill defined.
are consistently localized to the PPM. Unlike the primary motor and sensory cortices, the
(3) Following the somewhat sigmoid pattern of the SMA has significant communication with its contralat-
central region, the superior segment of the pre- eral counterpart via the corpus callosum. A large portion
central gyrus is convex anteriorly. Near its junc- of the corpus callosum is occupied by these crossing
tion with the middle segment, the superior seg- SMA fibers, explaining the rapid spread of a stimulus
ment is sharply distinct from the premotor area, from these regions and difficulties in determining lat-
but as it reaches the interhemispheric fissure, erality. The SMA provides direct contributions to the
these boundaries blur. Consequently, the supe- corticospinal tract, which end in the gray matter of the
rior segment of the motor cortex commonly com- anterior horn of the spinal cord at different levels. These
municates with the supplementary motor area direct projections are mostly involved in proximal limb
(SMA). control.
(4) The paracentral segment of the precentral gyrus
is the fourth and final segment. This segment de-
fines the anterior aspect of the paracentral lobule,
and has no sharp border anteriorly. The paracen- 䉴 STRUCTURAL LANDMARKS FOR
tral segment blends with the postcentral gyrus LOCALIZATION OF
at its inferomedial aspect, providing a significant SENSORIMOTOR CORTEX
communication with the primary sensory area.
The central sulcus provides a posterior bound- Given the functional importance of the central region to
ary for a short distance of the paracentral seg- motor and sensory function, localization of the central
ment, until the aforementioned communication sulcus is a key component to many surgeries. An indirect
is encountered. approach to its identification relies on skull landmarks,
and, subsequently, on brain reference coordinates. Two
As with the precentral gyrus, the postcentral gyrus widely used indirect methods are the Talairach method,
can also be divided into four segments, which closely which is based on the AC-PC reference plane24,25 , and
parallel the course of the precentral gyrus’ segments. Of the Olivier method, which is based on the callosal refer-
note, the inferior, or opercular, region of the postcentral ence plane.26,27 Both provide a crude regional location
gyrus is wider and thicker than in the precentral gyrus, for the central sulcus, but are insufficiently precise for
while the middle and superior segments are thinner in its true identification.
the primary sensory cortex compared with the Rolandic With the advent of modern imaging techniques,
motor cortex. more sophisticated and direct methods are now avail-
The premotor cortex is a transitional area located able for the localization of the central sulcus with nonin-
between the polar aspect of the frontal lobe and the vasive imaging. Using well-validated anatomofunctional
primary motor cortex. The boundaries of the premotor correlations, the clinician can often quickly determine
cortex in humans are nebulous and highly variable. An whether a certain lesion is in close proximity to eloquent
imaginary line joining the anterior border of the SMA areas or not and decide about the need for additional
with the cortical eye fields is the supposed anterior bor- functional imaging and/or intraoperative mapping. Fur-
der of the premotor cortex, while posteriorly it extends thermore, this localization with imaging can be used in
to the depths of the precentral sulcus. the operating room by correlating the anatomy of the
Anterior to the Rolandic cortex and on the medial exposed brain to anatomy on preoperative images us-
aspect of the brain, there are additional somatomotor ar- ing standard surgical navigation systems.
eas that have been extensively studied and are of clinical The direct methods for identifying Rolandic cortex
importance. These include the SMA, immediately ante- on imaging studies rely on landmarks that are visible
rior to medial Rolandic cortex, and the pre-SMA, which on axial and sagittal CT and MR scans.28 These land-
lies immediately anterior to the SMA. Penfield and Welch marks proved to be reliable and usually easily identified
(1951) described the SMA as located in the mesial aspect on structural brain imaging. Yousri et al. showed that
of the superior frontal gyrus, superior to the cingulate the motor hand area is represented on the cortex in a
sulcus and anterior to the primary motor cortex.22 The specific segment of the precentral gyrus that has a char-
anterior boundary of the SMA has been described as a acteristic shape on brain imaging. It is knob-like in the
line perpendicular to the AC-PC line passing by the an- form of an inverted omega in the axial plane (Fig. 14–1)
192 SECTION II SYSTEMS

Figure 14–1. Axial T1-weighted MRI of brain with contrast. On the left side, the
inverted omega shape can be clearly recognized that allows the identification of the
central sulcus and the motor hand area. On the right hemisphere, there is an
enhancing lesion occupying the motor hand area on that side.

and appears as a posteriorly directed hook in the tral sulcus. It connects the pre- and postcentral gyri at
sagittal plane.20 Berger at al. retrospectively correlated the level of the middle bend of the central sulcus. The
brain maps, derived from intraoperative mapping in pa- precentral component of this structure corresponds to
tients with brain tumors, with preoperative MR images the motor hand, while the postcentral part corresponds
in the axial, sagittal, and coronal planes. They found to the sensory hand area.19 Similarly, the tongue sensory
the Rolandic cortex was accurately identified on T2- area was described as a triangular-shaped cortical region
weighted axial images at the most rostral vertex by look- of the postcentral gyrus sitting directly above the sylvian
ing for the central sulcus with its distinctive transverse fissure31 , and the sensory area of the face and lips is in
orientation. The central sulcus was more difficult to iden- the narrow region of the postcentral gyrus located at the
tify on sagittal imaging, but the postcentral sulcus, imme- apex of the triangular tongue area.32
diately posterior to the postcentral gyrus, can readily be Overall, identification of central sulcus by using
identified as the posterior–superior terminal extension these anatomic landmarks on imaging studies is quite
of cingulate sulcus, also known as the marginal ramus reliable in normal human subjects, but is much less accu-
of the cingulate sulcus.29 rate or consistent in the clinical setting, where anatomy
While MRI landmarks for identifying the central may be deformed by regional pathology or where devel-
sulcus are readily correlated with the exposed brain opment anomalies may have led an atypical functional
at surgery through standard neurosurgical navigation organization. Identification of Rolandic cortex based
systems, it can also be useful to attempt to identify on surface anatomy of the exposed brain is even less
the Rolandic cortex by direct visual inspection of the reliable.
anatomy of the exposed brain. However, identification
of eloquent cortical areas based on anatomic landmarks
has been found to be unreliable in 16% of healthy 䉴 DIRECT CORTICAL STIMULATION
patients, and even more unreliable in patients with
pathology—35% due to distortions in anatomy and func- Despite advances in functional neuroimaging, direct cor-
tional organization.30 Still, simple inspection of the cere- tical electrical stimulation, very similar to that carried
bral convolutions during surgery offers valuable clues out by Penfield, may still be considered as the “gold
to the location of the sensiromotor area. Boling and standard” technique for mapping somatomotor and so-
Olivier19 found that the sensory hand area corresponds matosensory area. It entails the direct application of a
to the postcentral component of the “PPM.” The “PPM” low-level electrical current to the cortex in order to
is a prominent gyral fold elevating the floor of the cen- activate local cortical circuits. This results in the focal
CHAPTER 14 MAPPING OF THE SENSORIMOTOR CORTEX 193

muscular contraction when current is applied to pri- the absence of a functional response is not simply a re-
mary motor cortex and the perception of a localized flection of inadequate electrical stimulation, but instead
somatosensory percept when it is applied to primary more likely reflects the absence of an elicitable function
sensory cortex. This cortical stimulation can be applied in the region of cortex that was stimulated.
intraoperatively during craniotomy, or extraoperatively The interpretation of the cortical stimulation criti-
with a previously implanted subdural electrode array. cally relies on the observation and documentation of the
The technique can also be applied intraoperatively to the patient’s response. Mapping of the motor cortex can be
subcortical white matter tracts that project from Rolandic done in awake as well as in anesthetized patients. In ei-
cortex into the internal capsule to identify critical motor ther case, an examiner observes and documents patient’s
and sensory pathways. movements as a result of stimulation. Clonic movements
The application of the electric stimulus can be done typically result from stimulation of the primary motor
through a monopolar or a bipolar method. In bipolar area, while tonic or more complex movements can re-
stimulation, there is depolarization of the membrane at sult from stimulation of the premotor cortex. Movements
the cathode (the negative electrode) and hyperpolariza- can also sometimes be subtle or at a distant site and go
tion at the level of the anode (the positive electrode). If unnoticed, particularly when low current levels are em-
the membrane potential reaches a certain threshold, then ployed. For this reason, continuous electromyographic
an action potential is generated.33 In the case of bipo- monitoring was proposed as an adjunct to detect motor
lar stimulation, only the structures located between the responses.39 Mapping of the sensory cortex with direct
two electrodes are stimulated; many surgeons and neu- electrical stimulation can only be done in awake pa-
rophysiologists prefer bipolar stimulation because it may tients, as it is necessary to have patient cooperation in
offer slightly lesser risk of diffusion of current, allow- reporting subject percepts of somatosensory activation.
ing greater precision.34 In the case of monopolar stim- It is important to explain the intraoperative procedure to
ulation, there is one active electrode and one reference the patient before surgery and make sure that the patient
electrode that is located at a distance, the current in this understands the concept and purpose of the mapping
case is distributed circumferentially around the active and is comfortable with the unfamiliar environment and
electrode. sensations (Fig. 14–2).
The intensity of the depolarization resulting from It has long been recognized that stimulations of the
cortical stimulation depends on the parameters of stim- SMA evoke more complex responses than those result-
ulation and the characteristics of the membrane.33 ing from primary sensorimotor cortex stimulation. Re-
Consequently, the result of stimulation depends on the sults are classically reported as complex motor synergies,
electrode design (monopolar vs. bipolar), stimulation sensory responses, autonomic changes, vocalization,
parameters (intensity, duration, etc.), and tissue charac- and speech arrest.22,23,40,41 Using subdural electrodes
teristics (such as impedance). The impedance and ex-
citability of the brain can be significantly affected by
medications (such as general anesthesia) and by the
presence of a pathological process (such as a tumor or
gliosis). The impedance was also found to be higher
in white matter compared with gray matter.35 Moreover,
the electric stimulation of the brain can induce seizures;
this is why many authors recommend using intraop-
erative electrocorticographic recording for detection of
“after-discharge potentials.”36 In these cases, the use of
ice cold solutions was found to be effective in aborting
seizures.37
The value of intraoperative electrocorticography ex-
tends beyond simply minimizing the risk of an intraop-
erative seizure. It also allows for confirmation that an ad-
equate level of electrical stimulation was delivered. This
is particularly important in cases where a functional re-
sponse to electrical stimulation is not observed, which
may often occur when a small exposure is employed and
Figure 14–2. Exposed brain surface (right
an obviously functional response is not exposed (min-
hemisphere) during awake craniotomy. After direct
imal exposure, or minimap craniotomy).38 If intraoper- cortical stimulation, the areas of the primary
ative electrocorticography demonstrates that the current sensorimotor cortex were identified. 1, sensory face;
used meets or exceeds the threshold for generating after- 2, motor face; 3, sensory hand; 4, motor hand; 5,
discharge potentials, one can be more confident that motor leg.
194 SECTION II SYSTEMS

in patients evaluated for intractable epilepsy, Fried


et al.42 performed a detailed electric stimulation mapping
of the SMA with currents below the threshold of after-
discharges. They classified the motor responses in three
categories: simple (discrete movement of one joint), re-
gional (several joints confined to one extremity or re-
gion), and complex (responses involving several body
regions). Overall, 34.2% of the responses were simple,
41.9% were regional, and 23.9% were complex. While
most responses were contralateral, some of them were
ipsilateral or bilateral. Sensory responses were less fre-
quent than motor and they were of three types: (1) sensa-
tion of tingling and numbness, (2) subjective experience
of movement in the absence of actual movement activity,
and (3) subjective urge to perform a movement. In ad-
dition, stimulation of the dominant hemisphere elicited
speech-related responses (vocalization, speech arrest, Figure 14–3. A strip of electrodes is placed on the
and slowing or hesitancy). The authors were able to de- cortical surface for recording. The electrodes array is
tect a somatotopic organization (from posterior to ante- positioned with its long-axis perpendicular to that of
rior: lower extremity, trunk, upper extremity, face, and the central sulcus.
most anteriorly in the dominant hemisphere: speech).
Although a clear somatotopic organization of the SMA is
a matter of controversy, the stimulation results by Fried based on the recording of a large amplitude, biphasic,
at al.42 appear to be consistent with a number of animal negative–positive response from the postcentral gyrus,
experiments43 and clinical studies correlating the clinical and a corresponding lower amplitude, mirror (positive–
deficit with the extent of resection of the SMA.44 negative) waveform from the precentral gyrus. The la-
tency of this initial deflection of the waveform usually
occurs between 18 and 24 ms, and reflects the N20-P20
phase reversal (at 20 ms after each cutaneous stimulus
䉴 SOMATOSENSORY-EVOKED is delivered). To obtain a clean waveform and average
POTENTIAL PHASE REVERSAL out the noise, multiple trials are collected and averaged
over a few minutes.47 (Figs. 14–3 and 14–4)
The use of the morphology of somatosensory-evoked The success rate of this method in defining the loca-
potentials (SSEPs) recorded from the cortical surface to tion of the central sulcus is usually better than 90% (91%
localize the central sulcus was developed by Goldring in the series reported by King et al.48 and Cedzich et al.49 ,
in the late 1970s.45,46 This was based on the principle 94% for Wood et al.50 and 97% for Kombos et al.51 ). Rom-
that when a somatosensory potential is simultaneously stock et al.47 found in a large series of patients that the
recorded from both the precentral and postcentral gyri, somatosensory evoked potential phase reversal method
responses of reversed polarity are observed across the can be significantly affected by the presence of a tumor
central sulcus. Therefore, the detection of this “phase located in or around the central area. The pathophys-
reversal” allows the determination of the location of the iology of this phenomenon is possibly related to local
central sulcus. Practically, a somatosensory stimulus is mass effect or to desynchronization of the propagated
delivered by transcutaneously, most often by stimulat- afferent signals along the thalamocortical pathways by
ing the median nerve contralateral to the tested hemi- the tumor. Although the phase reversal technique is usu-
sphere with an electrical current. Attempts at using the ally considered as simple, fast, safe, and reliable, it has
tibial and trigeminal nerve to find the phase reversal at a number of important drawbacks. In around 10% of
the leg and face representation respectively appear to be the cases, the phase reversal may be questionable or
less successful and are still not well validated.47 A strip even absent. When successful, this method can allow
or grid of electrodes is placed on the cortical surface for determination of the central sulcus at the level of the
recording. The electrodes array is generally positioned hand area, but extrapolation has to be made to trace the
with its long axis in the anteroposterior direction in order sulcus proximally and distally. It does not allow for dis-
to have the long axis of the array perpendicular to the tinct localization of the somatotopic representation of the
long axis of the central sulcus. When median nerve stim- hemibody, as can be done with direct electrical stimula-
ulation is used, the electrodes have to be placed over the tion; this may be important in cases where the surgeon
suspected hand representation of the sensorimotor cor- is willing to sacrifice the cortical face motor representa-
tex. The localization of the central sulcus is then made tion (which is generally recovers well over time and is
CHAPTER 14 MAPPING OF THE SENSORIMOTOR CORTEX 195

Figure 14–4. Detection of phase reversal allowing the localization of the central
sulcus.

well tolerated by patients), but wishes to spare the hand fMRI has a number of advantages over other map-
representation. It also does not allow for repeated online ping techniques. It is a safe and noninvasive technique.
testing of the integrity of the motor pathways, which can It can be repeated to study more functions, or to follow
also be done with repeated direct electrical stimulation. a specific function in a certain patient over time to study
the impact of the progression of disease for example.
Compared with the gold standard (direct cortical stimu-
䉴 FUNCTIONAL MRI lation), fMRI has the advantages of examining a much
wider area. While cortical stimulation is limited by the
The last decade has witnessed remarkable advances in surgical exposure, fMRI can sample the whole brain. It
functional neuroimaging allowing the noninvasive ex- can also localize function in the depth of cortical sulci
amination and localization of brain functions. Blood that lie below the cortical surface. Compared with other
Oxygenation Level Dependant (BOLD) fMRI allows functional imaging, such as PET, fMRI has a higher spa-
mapping based on local physiological and metabolic tial resolution. The typical spatial resolution for fMRI in
consequences that accompany neuronal activities. In- the clinical arena is estimated to be in the order of 2–5
creased neuronal activity is associated with an increased mm.54 Advanced MRI techniques, including higher field
use of energy due to neuronal depolarization and ac- strength, can potentially provide an even better spatial
tion potential generation. Consequently, an increase in resolution. The main disadvantages of fMRI include (1)
the local blood flow occurs, affecting the ratio of oxyhe- motion-related artifact, this can result in image distor-
moglobin (oxyHb) to deoxyhemoglobin (deoxyHb) in tion in the case of some motor tasks for example, and
the region of neuronal activity. BOLD fMRI imaging is (2) the technique can be affected by large blood vessels,
generated based on the different magnetic properties of making the interpretation of results mostly problematic
oxyHb and deoxyHb.52 BOLD fMRI images are based in the case of an arteriovenous malformation or highly
on relative signal intensity changes related to different vascular tumor.
cognitive states during a single session: usually a block Several clinical studies have been conducted in or-
design is used in order to compare the brain responses der to validate the fMRI findings in mapping the sen-
in different states. In this design, there will be alternat- sorimotor cortex against the “gold standard” technique
ing periods between two states: a task or a stimulus is of direct cortical stimulation. Such a validation process
applied in one state, the other state serving as baseline has been conducted in patients harboring a brain le-
or control.53 sion in the vicinity of the central area. Typically, an fMRI
196 SECTION II SYSTEMS

is done preoperatively, and then direct cortical stimula- during the performance of a task. Blood flow under a
tion is performed during surgery. The comparison be- control condition is then subtracted from that measured
tween the results of the two mapping techniques was during task performance in order to detect regions with
thus used to validate the use of fMRI as a reliable and increased flow. These changes can be used as an indi-
accurate mapping method. Although fMRI is also widely rect method for detection of neuronal activity. On the
used in healthy volunteers, validation against mapping other hand, FDG PET measures cerebral metabolism.
is not possible in these cases for obvious reasons. Ma- FDG PET can also be used for mapping; the areas with
jos et al.55 in a study involving 33 patients with brain increased metabolism compared with the control con-
tumors in the vicinity of the central sulcus found that dition are used for identification of increased neuronal
the agreement in the location of motor centers between activity during a certain task.61
fMRI and cortical stimulation was 84%. The agreement PET can be used to study a wide range of behav-
for sensory centers was 83%. In similar studies, Fandino ioral tasks; however, the technique has a number of
et al.56 , Roux et al.57 , and Lehericy et al.58 found an significant disadvantages. Compared with fMRI, it has a
agreement of 82%, 87%, and 92%, respectively. Using poor signal-to-noise ratio and spatial resolution. PET is
3-T fMRI, Roessler et al.59 found 100% correlation with also considered invasive because of the administration
cortical stimulation data within 1 cm. In most of these of a radioactive tracer that limits the number of studies
studies, the area activated on fMRI is usually larger than that can be done for one patient. With the widespread
that identified by direct stimulation. And, depending on availability and lower degree of invasiveness of BOLD
the task used, there may be many sites of BOLD signal fMRI, PET is now rarely used for clinical mapping of
well beyond the primary motor cortex. This is a reflec- sensiromotor cortex. But it did play an important role
tion of the fundamental nature of BOLD fMRI for motor in the development of noninvasive functional mapping
mapping: it is a technique that measures activity dur- of patients.
ing a motor task, and, therefore, will reveal activity in Bittar et al.62 studied 11 patients with tumors around
brain areas that are activated during a motor task, such the central sulcus. All of them underwent H2 15 O PET
as prefrontal cortex, but are not directly essential or a and fMRI before surgery for mapping of the somatomo-
proximate cause of motor function. tor and somatosensory areas. The average distance be-
In general, BOLD fMRI is very dependent on the tween activation peaks obtained using fMRI and those
task performed during data acquisition. Mapping each obtained using PET imaging was 7.9 ± 4.8 mm, with 96%
muscle group along the entire primary motor cortex, for of the peaks being located on either the same (52%) or
example, would be very difficult because it would re- adjacent sulci and gyri (44%). Overlapping of voxels ac-
quire a large number of tasks and a very long scan time. tivated by each modality occurred in 92% of the studies.
For this reason, only a few selected motor tasks typ- In another study, the same group validated PET mapping
ically are performed in order to delineate key areas of of the sensory cortex against intraoperative cortical stim-
the primary motor cortex. Various motor tasks have been ulation. They found that in 22 (95.6%) of 23 statistically
used, including finger tapping, hand clenching, and el- significant PET activation foci, spatially concordant sites
bow and shoulder movements. Berntsen et al.60 con- on cortical stimulation were also observed. Intraopera-
ducted a study to determine a robust set of motor tasks tive cortical stimulation was positive in 40% of the PET
that could be used to delineate the motor cortex in a pre- activation studies that did not result in statistically signif-
cise and accurate fashion. They found that tasks related icant activation. All PET activation foci with a t-statistic
to finger, toe, and tongue movements were the most ef- greater than 4.75 were associated with spatially concor-
fective and associated with the highest success rates. dant sites of positive intraoperative cortical stimulation
(ICS). All PET activation foci with a t-statistic smaller
than 3.2 were associated with negative ICS.63 Schreck-
䉴 POSITRON EMISSION enberger et al. conducted a study involving 20 patients
TOMOGRAPHY with tumors of the central region to validate FDG-PET
mapping of the motor cortex. Using direct cortical stim-
Brain mapping with PET relies administration of a ra- ulation as the “gold standard,” they found that PET has
dioactive tracer compound labeled with a positron- a 94% sensitivity and 95% specificity for identification of
emitting isotope to localize areas that are metabolically primary motor areas.64
active during a task. Scintillation detectors are used dur-
ing scanning in order to determine the position of the
isotope, and therefore, the portion of the brain acti- 䉴 MAGNETOENCEPHALOGRAPHY
vated by the task. For functional brain mapping, PET
is usually performed using 15 O or fluorodeoxyglucose MEG is another noninvasive technique that measures
(FDG).15 O PET is used to measure cerebral blood flow brain activity through measurement of magnetic fields.
CHAPTER 14 MAPPING OF THE SENSORIMOTOR CORTEX 197

In a sense, it is similar to electroencephalography (EEG), neuronavigation workstation. Awake craniotomies were


but measures magnetic field changes instead of volt- done in 54.3% of the cases. Somatosensory mapping us-
age changes to detect neuronal activity. Ion currents ac- ing MEG was technically successful in 97% for digits
companying neuronal activity generate magnetic field and hand, 90% for lips and 82% for toes. The three-
changes that can be detected using a biomagnetometer, dimensional distance between somatosensory sites on
and used to reconstruct an image of the neural activi- MSI and somatosensory sites on intraoperative cortical
ties. When MEG is used for mapping purposes, a task stimulation ranged between 1.5 and 42.2 mm (mean:
is performed multiple times, and then neuromagnetic 20.5 mm). The mean two-dimensional distance was
signals are averaged over multiple trials in order to de- 12.5 mm.
lineate the signals produced by focal active areas from
the background activity. MEG is mainly used for map-
ping of the somatosensory cortex. Motor mapping can 䉴 TRANSCRANIAL MAGNETIC
also be done but motor data are generally not as re- STIMULATION
producible or precisely located as sensory mapping.65
Magnetic source imaging (MSI) is the coregistration of Transcranial magnetic stimulation (TMS) consists on the
MEG data to a structural image to facilitate the anatomo- noninvasive application of a magnetic field on the scalp
functional correlations. in order to stimulate neuronal electric activity. An elec-
Like any other technique, MEG has both advantages tromagnetic coil is held over the cranium, creating a
and disadvantages. Compared with EEG, MEG has a sim- magnetic field that induces a perpendicular electric field.
ilar excellent temporal resolution, but better spatial res- In 1985, the technique was introduced as a novel, non-
olution (of approximately 2 mm), and better signal-to- invasive, and painless application for mapping of the
noise ratio. Moreover, MEG signals are not attenuated human motor cortex.69 Subsequent studies used TMS
by the cranium and scalp.66 However, MEG scanners re- in conjunction with frameless neuronavigation systems
quire dedicated personnel and specially equipped and and tried to validate the findings against direct cortical
shielded rooms. They are also very sensitive to environ- stimulation. Krings et al.70 applied this technique in two
mental magnetic noise. MEG scanners are currently very patients. TMS was well tolerated with no reported side
expensive with limited availability. effects. They also found good correlation between TMS
Gallen et al.67 conducted a multicenter study to val- and direct stimulation. In locations of more than 2.5 cm
idate MEG data for mapping of the primary sensory distant from the site of direct cortical stimulation, com-
cortex against intraoperative cortical stimulation. They pound muscle action potentials were not elicited in ei-
found that MSI results correctly identified the postcen- ther patient. TMS responses that were 75% of maximum
tral gyrus in all of their six patients. The calculated dis- motor evoked potential amplitude or larger were elicited
crepancy between MSI-determined central sulcus and over cortical regions that were within 1 cm of the corti-
the one determined by phase reversal ranged between cal region where direct stimulation elicited movements
0.37 and 1.28 cm (with an average of 0.7 cm). In a similar of the same muscles. No responses smaller than 50% of
study, Rezai et al.68 performed a preoperative mapping the maximum motor evoked potential (MEP) amplitude
of the sensory and motor cortex using MEG in 50 pa- were elicited within 0.5 cm of the cortical areas of direct
tients with lesions located in close proximity to the cen- cortical stimulation. The same group conducted another
tral sulcus. In ten of these patients, an integration of the study to correlate the findings of TMS with fMRI. 71 They
MEG data with an image-guided stereotactic system was found strong correlation between the two techniques
performed and used during surgery. This allowed an ac- in three volunteers and two patients. Peak MEP ampli-
curate comparison between the functional data provided tudes consistently coincided with peak fMRI activation,
by MEG and the intraoperative direct stimulation. They although the area of TMS responses was larger than the
found that preoperative MEG localization of the sensori- area of fMRI activation. TMS sites further than 2 cm from
motor cortex was successful in all patients. This allowed the cortical surface projection of the fMRI produced no
aggressive but safe removal of the lesions. All patients MEPs. Neggers et al.72 applied a frameless stereotactic
had gross total resections, eight of them with no change device using structural and fMRI data to guide TMS coil
in their neurological exam, and two with transient wors- placement. The distance between the MEP responses
ening of a preoperative weakness. Schiffbauer et al.65 and the maximum fMRI activation areas was on average
treated a total of 224 patients who underwent preoper- less than 5 mm. Kantelhardt et al.73 conducted a pilot
ative MSI. The noninvasive mapping of the sensorimo- clinical study on the use of robot-assisted, image-guided
tor cortex using MEG was done 2 days before surgery TMS for mapping the motor cortex. The areas of maxi-
to allow time for discussion of results with the patient. mal MEP response localized within the “finger tapping”
The MEG data were registered to a high-resolution MRI activated areas by fMRI in all of their six patients, and re-
to form the MSI, the images were then transferred to a peated TMS measurement showed high reproducibility.
198 SECTION II SYSTEMS

䉴 CLINICAL APPLICATIONS Sensory hand resections led to a proprioceptive deficit


that significantly improved with time. Sensory leg resec-
RESECTIONS WITHIN THE tions lead to persistent numbness in the foot. A transient
ROLANDIC CORTEX weakness following face resections was noted, recovery
occurred in 2–3 weeks.
Sir Victor Horsley was perhaps the first person to suc-
cessfully perform surgical resections within the primary
sensorimotor cortex in order to control abnormal in- PRESERVATION OF ROLANDIC
voluntary movements (epilepsy).74 His pioneering work CORTEX IN SURGERY FOR INTRINSIC
was instrumental in the understanding of the central cor- BRAIN TUMORS
tex. Thereafter, multiple reports on procedures involv-
ing extirpation of parts of the primary motor cortex ap- Because surgery within Rolandic cortex often results in a
peared between 1886 and 1950, but rarely since then.75 permanent and disabling deficit, mapping is often used
The work of Penfield and Rasmussen showed that large to curtail a resection to avoid this deficit. In general, the
resections in the precentral gyrus produce complete, im- aim of surgery in the treatment of intrinsic brain tumors
mediate paralysis followed by spasticity. Long-term re- is to maximize the extent of resection while minimizing
covery led to significant improvement in proximal limb the risk of postoperative neurological deficit. When tu-
function but poor recovery of the distal parts. In the mors are located in close vicinity to the central sulcus, it
modern era of neurosurgery, resections of parts of the is of utmost importance to determine the exact location
primary sensorimotor cortex have been largely avoided, of the sensorimotor cortex and its relation to the tumor.
because of concerns regarding a permanent neurolog- Preoperative studies including high-resolution MRI and
ical deficit. As a consequence, residual tumors as well functional imaging help in the identification of the elo-
as epileptogenic foci are commonly left in this area, quent cortex and its relation to the lesion, thus allowing
at the expense of suboptimal seizure control and tu- a clear discussion with the patient about the feasibility of
mor extent of resection. However, in carefully selected gross total resection and the risks of surgery. The data
cases, particularly in surgery for intractable epilepsy, se- gathered from the preoperative work-up is then used
lective resections within the sensorimotor cortex can during surgery typically in conjunction with intraopera-
be made with an acceptable deficit. Patients’ counsel- tive mapping techniques that are still considered as “gold
ing is of paramount importance in these cases; the ex- standard”.
pected postoperative deficit as well as realistic expec- Although surgical resection constitutes a crucial el-
tations regarding recovery should be clearly explained ement in the management of intrinsic brain tumors, the
to them based on the best available data to date. Duf- efficacy of extent of resection in improving patients’ out-
fau et al.76 reported their experience in resections of come remains a matter of debate.38 With the exception
low-grade glioma in eloquent brain, eight of their cases of World Health Organization grade I tumors, where
involved the primary sensory cortex and three of them cure is the goal of surgery, the vast majority of patients
the primary motor face area. In sensory resections, all with glioma are expected to have tumor recurrence at
patients had a severe postoperative sensory loss, but re- some point. The median survival rates are in the order
covered within 3 months, with mild residual dysesthesias of 12–18 months for glioblastoma78 , and 41 months for
in two cases. In the case of motor face area resection, all anaplastic astrocytoma.79 The 5-year survival for low-
three patients had a central facial palsy that recovered grade glioma varies between 40% and 90%.80−82 In the
completely within a month. The transient deficit after re- case of low-grade gliomas, there is mounting evidence
sections of the sensorimotor face area is also consistent suggesting that a more extensive resection is associated
with the results reported by Lehman et al., thus advo- with increased survival. Although class I evidence is
cating resections of epileptogenic foci in this region.77 lacking, multiple studies that included volumetric anal-
These findings are also consistent with the experience of ysis showed that the extent of resection is a significant
the senior author (YC). Between 1997 and 2005, Youssef predictor of 5-year survival and 5-year progression-free
Comair treated 33 patients with seizure onset in the pri- survival.83−86 In the case of high-grade gliomas, the im-
mary sensorimotor cortex (unpublished data). The un- pact of the extent of resection is more controversial.
derlying pathology was glioma in 19 patients, cortical While a number of studies showed that aggressive re-
dysplasia in 10, and vascular lesions in 5. All patients sections were associated with a survival benefit and a
underwent respective surgeries with an overall seizure- delay in tumor progression, an almost equal number of
free outcome rate of 65%. The functional outcome was as studies failed to show such a correlation.79,87−89
follows for hand and/or leg motor resections progressive Intrinsic brain tumors frequently involve eloquent
recovery of function occurred 6-week postoperatively brain areas. The benefits of performing a gross total
and continued to improve over 2–3 years, with return resection in these cases have to be balanced against
of individual finger movements but not toe movements. the risks of inflicting significant neurological deficit with
CHAPTER 14 MAPPING OF THE SENSORIMOTOR CORTEX 199

potentially devastating consequences on the quality of


life and survival. Intraoperative brain mapping allows
the surgeon to identify the eloquent brain regions during
surgery and to preserve them; this will result in maximiz-
ing the extent of resection while minimizing the risk of
deficit. Chang et al.90 recently reported the experience
of the University of California in San Francisco regard-
ing resection of low-grade gliomas in eloquent areas of
the brain. Between 1989 and 2005, they treated 281 pa-
tients; most of them (174 patients) had tumors involv-
ing “presumed” eloquent areas based on preoperative
anatomical MRI, these patients had significantly lower 5-
year overall survival and progression-free survival. How-
ever, when analyzing the role of mapping, they found Figure 14–5. Intraoperative picture of the exposed
that 127 patients with tumors “presumed” to involve elo- surface of the right hemisphere during awake
quent structures underwent intraoperative mapping. In craniotomy for tumor resection. Anatomically the
81 patients, the tumor was indeed found to directly in- tumor occupies the precentral gyrus; however, the
filtrate eloquent structures (“true-eloquent” cases), while functional areas were identified by direct cortical
stimulation: 1, proximal arm motor; 2, face motor;
it spared functional areas in 46 patients, thus allowing a 3, 4, and 5, face sensory; 6, hand sensory; 7, wrist
more aggressive resection (“false-eloquent” cases). The sensory; 8 and 9, hand motor. As a result, the tumor
analysis also showed that the overall survival in the case (the swollen part of the gyrus) was mostly involving
of “false-eloquent” location was not statistically different noneloquent area. However, it invaded the motor
from that of tumors involving noneloquent areas. These face inferiorly. Gross total resection of the tumor was
findings underline the importance of mapping whenever achieved, including removal of the motor face area.
the tumor is suspected to involve eloquent structures, Patient developed facial weakness initially but this
recovered well, as expected.
and show that it can improve the extent of resection
and long-term outcome, because it allows delineating
tumors that truly invade functional structures from those to this end. The intraoperative epidural cortical mapping
that do not. These findings are consistent with the ex- using epidural stimulation and/or SSEPs is widely used.51
perience of other authors who consider that mapping Pirotte et al. used fMRI data registered in a navigation
allowed resection of tumors previously considered un- system to treat 18 patients with intractable pain. They
resectable and that it can improve outcome by virtue of found correspondence between fMRI and intraoperative
maximizing the extent of resection.91 (Fig. 14–5). cortical mapping in 17 of them.94

MOTOR CORTICAL STIMULATION REFERENCES


FOR NEUROPATHIC PAIN
1. Uematsu S, Lesser RP, Gordon B. Localization of sensori-
motor cortex: the influence of Sherrington and Cushing on
Central pain is by definition, damage to or dysfunction,
the modern concept. Neurosurgery 1992;30:904-912; dis-
of the central nervous system resulting in long-lasting cussion 912-903.
pain. A large number of etiologies have been implicated, 2. Boling W, Olivier A, Fabinyi G. Historical contributions to
including stroke, trauma, and tumors. Peripheral nerve the modern understanding of function in the central area.
injury can also be the cause of central pain by secon- Neurosurgery 2002;50:1296-1309, discussion 1309-1210.
darily affecting the central nervous system.92 In view of 3. Rolando L. Della Struttura degli emisferi Cerebrali. Count-
limited success of medical treatment in general, corti- way Library of Medicine, Harvard University, Cambridge,
cal stimulation has emerged as a promising option. The MA: Del Prof, 1929.
presumed mechanism for pain relief is based on the ex- 4. Jackson J. Loss of speech. Clinical Lectures and Reports of
citability modification of neuronal activity, thus cortical London Hospital 1864;1:459.
stimulation is thought to interfere with pain signals re- 5. Fritsch G, Hitzig E. Uber die elektrische erregbarkeit des
Grosshirns. Arch Anat Physiol 1870;37:300-332.
sulting in modulation of abnormal activity. In a recent
6. Bartholow R. Experimental investigations into the functions
systematic review of the literature, the mean responders of the human brain. Am J Med Sci 1874;67:305-313.
rate to epidural motor cortex stimulation was 64%.93 The 7. Ferrier D. The Functions of the Brain. 2nd edition. London:
technique consists of introducing epidural electrodes to Smith, elder, & Co, 1886.
stimulate the motor cortex, the localization of the motor 8. Grunbaum A, Sherrington C. Observations on the physiol-
cortex appears as such to be an integral part of the pro- ogy of the cerebral cortex of some of the higher apes. Proc
cedure. Numerous mapping techniques have been used R Soc Lond 1901;69:206-209.
200 SECTION II SYSTEMS

9. Keen W. Three successful cases of cerebral surgery. Am J 33. Kombos T, Suss O. Neurophysiological basis of direct cor-
Med Sci 1888;96:452-465. tical stimulation and applied neuroanatomy of the motor
10. Mills C, Frazier C. The motor area of the human cerebrum, cortex: a review. Neurosurg Focus 2009;27:E3.
its position and subdivisions, with some discussions of the 34. Haglund MM, Ojemann GA, Blasdel GG. Optical imaging of
surgery of this area. Univ Penn Med Bull 1905;18:135-147. bipolar cortical stimulation. J Neurosurg 1993;78:785-793.
11. Krause F. Chirurgie des Gehirns und Ruckenmarks. Berlin: 35. Geddes LA, Baker LE. The specific resistance of biological
Urban & Schwarzenberg, 1911. material–a compendium of data for the biomedical engi-
12. Foerster O. Motorische Felder und Bahnen. Berlin: Verlag neer and physiologist. Med Biol Eng 1967;5:271-293.
von Julius Springer, 1936. 36. Pouratian N, Cannestra AF, Bookheimer SY, et al. Vari-
13. Penfield W, Rasmussen A. The Cerebral Cortex. New York: ability of intraoperative electrocortical stimulation map-
Macmillan co., 1957. ping parameters across and within individuals. J Neurosurg
14. Farrell DF, Burbank N, Lettich E, Ojemann GA. Individual 2004;101:458-466.
variation in human motor-sensory (rolandic) cortex. J Clin 37. Sartorius CJ, Berger MS. Rapid termination of intraopera-
Neurophysiol 2007;24:286-293. tive stimulation-evoked seizures with application of cold
15. Eberstaller O. Ein Beitrag zur Anatomie der Oberflache des Ringer’s lactate to the cortex. Technical note. J Neurosurg
Grosshirns. Wien: Urban and Schwarzenberg, 1890. 1998;88:349-351.
16. Ono M, Kubik S, Abernathey C. Atlas of the Cerebral Sulci. 38. Sanai N, Berger MS. Intraoperative stimulation techniques
Stuttgart: Thieme, 1990. for functional pathway preservation and glioma resection.
17. Symington J, Crymble PT. The central fissure of the cere- Neurosurg Focus 2010;28:E1.
brum. J Anat Physiol 1913;47:321-339. 39. Yingling CD, Ojemann S, Dodson B, et al. Identification
18. Broca P. Memoires Sur Le Cerveau de L’Homme et des of motor pathways during tumor surgery facilitated by
Primates. Paris: Reinwald, 1888. multichannel electromyographic recording. J Neurosurg
19. Boling WW, Olivier A. Localization of hand sensory func- 1999;91:922-927.
tion to the pli de passage moyen of Broca. J Neurosurg 40. Green JR, Angevine JB, White JC Jr, et al. Significance of
2004;101:278-283. the supplementary motor area in partial seizures and in
20. Yousry TA, Schmid UD, Alkadhi H, et al. Localization of cerebral localization. Neurosurgery 1980;6:66-75.
the motor hand area to a knob on the precentral gyrus. A 41. Foerster O. The motor cortex in man in the light of Hugh-
new landmark. Brain 1997;120(Pt 1):141-157. lings Jackson’s doctrines. Brain 1936;59:135-159.
21. Boling W, Parsons M, Kraszpulski M, et al. Whole-hand sen- 42. Fried I, Katz A, McCarthy G, et al. Functional organization
sorimotor area: cortical stimulation localization and corre- of human supplementary motor cortex studied by electrical
lation with functional magnetic resonance imaging. J Neu- stimulation. J Neurosci 1991;11:3656-3666.
rosurg 2008;108:491-500. 43. Woolsey C, Settlage P, Meyer D, et al. Patterns of local-
22. Penfield W, Welch K. The supplementary motor area of ization in precentral and “supplementary” motor areas and
the cerebral cortex. A clinical and experimental study. Arch their relation to the concept of premotor areas. Res Publ
Neurol Psychiatry 1951;66(3):289-317. Assoc Nerv Ment Dis 1952;30:238-264.
23. Talairach J, Bancaud J. Lesions, irritative zone and epilep- 44. Fontaine D, Capelle L, Duffau H. Somatotopy of the
togenic focus. Confin Neurol 1966;27(1):91-94. supplementary motor area: evidence from correlation of
24. Szikla G, Bouvier G, Hori T. In vivo localization of brain the extent of surgical resection with the clinical pat-
sulci by arteriography: a stereotactic anatomoradiological terns of deficit. Neurosurgery 2002;50:297-303; discussion
study. Brain Res 1975;95(2–3):497-502. 303-295.
25. Talairach J, Tournoux P. Co-Planar Stereotactic Atlas of the 45. Goldring S. A method for surgical management of focal
Human Brain. New York: Thieme Medical Publisher, 1988. epilepsy, especially as it relates to children. J Neurosurg
26. Vanier M, Ethier R, Clark J, et al. Anatomical interpretation 1978;49:344-356.
of MR scans of the brain. Magn Reson Med 1987;4:185-188. 46. Goldring S, Gregorie EM. Surgical management of epilepsy
27. Villemure JG, Marchand E, Peters T, et al. Magnetic res- using epidural recordings to localize the seizure focus. Re-
onance imaging stereotaxy: recognition and utilization of view of 100 cases. J Neurosurg 1984;60:457-466.
the commissures. Appl Neurophysiol 1987;50:57-62. 47. Romstock J, Fahlbusch R, Ganslandt O, et al. Localisation
28. Naidich TP, Valavanis AG, Kubik S. Anatomic relation- of the sensorimotor cortex during surgery for brain tu-
ships along the low-middle convexity: Part I–Normal spec- mours: feasibility and waveform patterns of somatosensory
imens and magnetic resonance imaging. Neurosurgery evoked potentials. J Neurol Neurosurg Psychiatry 2002;72:
1995;36:517-532. 221-229.
29. Berger MS, Cohen WA, Ojemann GA. Correlation of motor 48. King RB, Schell GR. Cortical localization and monitoring
cortex brain mapping data with magnetic resonance imag- during cerebral operations. J Neurosurg 1987;67:210-219.
ing. J Neurosurg 1990;72:383-387. 49. Cedzich C, Taniguchi M, Schafer S, Schramm J. Somatosen-
30. Yetkin FZ, Papke RA, Mark LP, et al. Location of the senso- sory evoked potential phase reversal and direct motor cor-
rimotor cortex: functional and conventional MR compared. tex stimulation during surgery in and around the central
AJNR Am J Neuroradiol 1995;16:2109-2113. region. Neurosurgery 1996;38:962-970.
31. Picard C, Olivier A. Sensory cortical tongue representation 50. Wood CC, Spencer DD, Allison T, et al. Localization of hu-
in man. J Neurosurg 1983;59:781-789. man sensorimotor cortex during surgery by cortical surface
32. Boling W, Reutens DC, Olivier A. Functional topography recording of somatosensory evoked potentials. J Neurosurg
of the low postcentral area. J Neurosurg 2002;97:388-395. 1988;68:99-111.
CHAPTER 14 MAPPING OF THE SENSORIMOTOR CORTEX 201

51. Kombos T, Suess O, Funk T, et al. Intra-operative mapping 68. Rezai AR, Hund M, Kronberg E, et al. The interactive use
of the motor cortex during surgery in and around the motor of magnetoencephalography in stereotactic image-guided
cortex. Acta Neurochir (Wien) 2000;142:263-268. neurosurgery. Neurosurgery 1996;39:92-102.
52. Matthews PM, Jezzard P. Functional magnetic resonance 69. Barker AT, Jalinous R, Freeston IL. Non-invasive magnetic
imaging. J Neurol Neurosurg Psychiatry 2004;75:6-12. stimulation of human motor cortex. Lancet 1985;1:1106-
53. Friston KJ, Zarahn E, Josephs O, et al. Stochastic designs 1107.
in event-related fMRI. Neuroimage 1999;10:607-619. 70. Krings T, Buchbinder BR, Butler WE, et al. Stereotactic tran-
54. Yoo SS, Talos IF, Golby AJ, et al. Evaluating requirements scranial magnetic stimulation: correlation with direct elec-
for spatial resolution of fMRI for neurosurgical planning. trical cortical stimulation. Neurosurgery 1997;41:1319-1325;
Hum Brain Mapp 2004;21:34-43. discussion 1325-1316.
55. Majos A, Tybor K, Stefanczyk L, Goraj B. Cortical mapping 71. Krings T, Buchbinder BR, Butler WE, et al. Functional mag-
by functional magnetic resonance imaging in patients with netic resonance imaging and transcranial magnetic stim-
brain tumors. Eur Radiol 2005;15:1148-1158. ulation: complementary approaches in the evaluation of
56. Fandino J, Kollias SS, Wieser HG, et al. Intraoperative vali- cortical motor function. Neurology 1997;48:1406-1416.
dation of functional magnetic resonance imaging and cor- 72. Neggers SF, Langerak TR, Schutter DJ, et al. A stereotac-
tical reorganization patterns in patients with brain tumors tic method for image-guided transcranial magnetic stimu-
involving the primary motor cortex. J Neurosurg 1999;91: lation validated with fMRI and motor-evoked potentials.
238-250. Neuroimage 2004;21:1805-1817.
57. Roux FE, Ibarrola D, Tremoulet M, et al. Methodological 73. Kantelhardt SR, Fadini T, Finke M, et al. Robot-assisted
and technical issues for integrating functional magnetic res- image-guided transcranial magnetic stimulation for soma-
onance imaging data in a neuronavigational system. Neu- totopic mapping of the motor cortex: a clinical pilot study.
rosurgery 2001;49:1145-1156; discussion 1156-1147. Acta Neurochir (Wien) 2010;152:333-343.
58. Lehericy S, Duffau H, Cornu P, et al. Correspondence be- 74. Vilensky JA, Gilman S. Using extirpations to understand
tween functional magnetic resonance imaging somatotopy the human motor cortex: Horsley, Foerster, and Bucy. Arch
and individual brain anatomy of the central region: com- Neurol 2003;60:446-451.
parison with intraoperative stimulation in patients with 75. Vilensky JA, Gilman S. Motor cortex extirpation (1886-
brain tumors. J Neurosurg 2000;92:589-598. 1950): the influence of Sir Victor Horsley. Neurosurgery
59. Roessler K, Donat M, Lanzenberger R, et al. Evaluation of 2002;51:1484-1487; discussion 1488.
preoperative high magnetic field motor functional MRI (3 76. Duffau H, Capelle L, Denvil D, et al. Functional recovery
Tesla) in glioma patients by navigated electrocortical stimu- after surgical resection of low grade gliomas in eloquent
lation and postoperative outcome. J Neurol Neurosurg Psy- brain: hypothesis of brain compensation. J Neurol Neuro-
chiatry 2005;76:1152-1157. surg Psychiatry 2003;74:901-907.
60. Berntsen EM, Samuelsen P, Lagopoulos J, et al. Mapping the 77. Lehman R, Andermann F, Olivier A, et al. Seizures with
primary motor cortex in healthy subjects and patients with onset in the sensorimotor face area: clinical patterns
peri-rolandic brain lesions before neurosurgery. Neurol Res and results of surgical treatment in 20 patients. Epilepsia
2008;30:968-973. 1994;35:1117-1124.
61. Tai YF, Piccini P. Applications of positron emission tomog- 78. Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing
raphy (PET) in neurology. J Neurol Neurosurg Psychiatry and benefit from temozolomide in glioblastoma. N Engl J
2004;75:669-676. Med 2005;352:997-1003.
62. Bittar RG, Olivier A, Sadikot AF, et al. Presurgical motor and 79. Keles GE, Chang EF, Lamborn KR, et al. Volumetric ex-
somatosensory cortex mapping with functional magnetic tent of resection and residual contrast enhancement on
resonance imaging and positron emission tomography. initial surgery as predictors of outcome in adult pa-
J Neurosurg 1999;91:915-921. tients with hemispheric anaplastic astrocytoma. J Neuro-
63. Bittar RG, Olivier A, Sadikot AF, et al. Localization of so- surg 2006;105:34-40.
matosensory function by using positron emission tomog- 80. Nakamura M, Konishi N, Tsunoda S, et al. Analysis of prog-
raphy scanning: a comparison with intraoperative cortical nostic and survival factors related to treatment of low-grade
stimulation. J Neurosurg 1999;90:478-483. astrocytomas in adults. Oncology 2000;58:108-116.
64. Schreckenberger M, Spetzger U, Sabri O, et al. Locali- 81. Shaw E, Arusell R, Scheithauer B, et al. Prospective ran-
sation of motor areas in brain tumour patients: a com- domized trial of low- versus high-dose radiation therapy
parison of preoperative [18F]FDG-PET and intraoperative in adults with supratentorial low-grade glioma: initial re-
cortical electrostimulation. Eur J Nucl Med 2001;28:1394- port of a North Central Cancer Treatment Group/Radiation
1403. Therapy Oncology Group/Eastern Cooperative Oncology
65. Schiffbauer H, Berger MS, Ferrari P, et al. Preoperative mag- Group study. J Clin Oncol 2002;20:2267-2276.
netic source imaging for brain tumor surgery: a quantitative 82. Yeh SA, Ho JT, Lui CC, et al. Treatment outcomes and
comparison with intraoperative sensory and motor map- prognostic factors in patients with supratentorial low-grade
ping. J Neurosurg 2002;97:1333-1342. gliomas. Br J Radiol 2005;78:230-235.
66. Pizzella V, Romani G. Principles of Magnetoencephalogra- 83. Claus EB, Horlacher A, Hsu L, et al. Survival rates in pa-
phy. New York: Raven Press, 1990, pp. 1-9. tients with low-grade glioma after intraoperative magnetic
67. Gallen CC, Schwartz BJ, Bucholz RD, et al. Presurgical lo- resonance image guidance. Cancer 2005;103:1227-1233.
calization of functional cortex using magnetic source imag- 84. Karim AB, Maat B, Hatlevoll R, et al. A randomized trial on
ing. J Neurosurg 1995;82:988-994. dose-response in radiation therapy of low-grade cerebral
202 SECTION II SYSTEMS

glioma: European Organization for Research and Treatment 90. Chang EF, Clark A, Smith JS, et al. Functional mapping-
of Cancer (EORTC) Study 22844. Int J Radiat Oncol Biol guided resection of low-grade gliomas in eloquent areas of
Phys 1996;36:549-556. the brain: improvement of long-term survival. J Neurosurg
85. Sanai N, Polley MY, Berger MS. Insular glioma resection: 2011;114(3):566-573.
assessment of patient morbidity, survival, and tumor pro- 91. Duffau H. Surgery of low-grade gliomas: towards a
gression. J Neurosurg 2010;112:1-9. ‘functional neurooncology’. Curr Opin Oncol 2009;21:
86. Smith JS, Chang EF, Lamborn KR, et al. Role of extent of re- 543-549.
section in the long-term outcome of low-grade hemispheric 92. Zaghi S, Heine N, Fregni F. Brain stimulation for the treat-
gliomas. J Clin Oncol 2008;26:1338-1345. ment of pain: a review of costs, clinical effects, and mech-
87. Pope WB, Sayre J, Perlina A, et al. MR imaging correlates anisms of treatment for three different central neuromod-
of survival in patients with high-grade gliomas. AJNR Am J ulatory approaches. J Pain Manag 2009;2:339-352.
Neuroradiol 2005;26:2466-2474. 93. Lima MC, Fregni F. Motor cortex stimulation for chronic
88. Keles GE, Anderson B, Berger MS. The effect of extent of pain: systematic review and meta-analysis of the literature.
resection on time to tumor progression and survival in pa- Neurology 2008;70:2329-2337.
tients with glioblastoma multiforme of the cerebral hemi- 94. Pirotte B, Voordecker P, Neugroschl C, et al. Combina-
sphere. Surg Neurol 1999;52:371-379. tion of functional magnetic resonance imaging-guided neu-
89. Lacroix M, Abi-Said D, Fourney DR, et al. A multivari- ronavigation and intraoperative cortical brain mapping
ate analysis of 416 patients with glioblastoma multiforme: improves targeting of motor cortex stimulation in neu-
prognosis, extent of resection, and survival. J Neurosurg ropathic pain. Neurosurgery 2005;56:344-359; discussion
2001;95:190-198. 344-359.
Chapter 15
Mapping of Human Language
Nitin Tandon
Department of Neurosurgery, University of Texas Medical School, Houston, Texas

䉴 INTRODUCTION AND interference with language output—including speech ar-


HISTORICAL BACKGROUND rest, alexia, agraphia, paraphasia, and anomia.13,12 Even
more detailed and comprehensive language maps were
Language is a uniquely developed human skill, essential generated by Ojemann15 who systematically compiled
for the normal functioning of an individual in society. data relating to language localization in large numbers
It is the inveterate symbolic or spoken representation of patients. Intraoperative mapping techniques devel-
of abstract concepts, consistent across a group within oped for the management of patients with epilepsy,
a species. While certain bees,1 birds,2 simians,3 and were then also broadly applied to the resection of glial
cetaceans4 are able to communicate information vocally neoplasms.16 Population data compiled in this way15,17,18
or symbolically, the human ability to produce and com- were represented as two-dimensional representations of
prehend symbolic and spoken language is unique. The language loci overlaid on schematic outlines of the brain
limitations of interclass homology (confounding the rel- and major sulci.19 Such maps are well suited for individ-
evance of insect and avian data), coupled with practi- ual patient management, but are untenable for accurate,
cal and ethical considerations in the invasive research unbiased comparisons between patients, or with func-
of marine mammals or free ranging simians have pre- tional imaging data. Despite these and other limitations
cluded the development of animal models of language (see later), these maps have been vital to the develop-
acquisition and production that could then be applied ment of modern concepts regarding the organization of
to humans. human language, and are broadly accepted as the cur-
The first spatially specific representations of lan- rent clinical standard for language localization.
guage came from careful studies of cerebral lesions
produced in humans by “experiments of nature.” Paul
Broca5 and Carl Wernicke6,7 used these to develop a
model that represented specific language processes. The 䉴 LATERALIZATION VERSUS
next advance after lesion localization came from the use LOCALIZATION
of focal currents for language localization. This tech-
nique of intraoperative language mapping with elec- The goal of language localization cannot be reason-
tricity was first reliably accomplished in Germany by ably accomplished without definitive a priori knowl-
Otfrid Foerster, a neurologist turned surgeon, who edge of which hemisphere is dominant for language.
treated soldiers with epilepsy following penetrating cere- Even though the exact proportion of normal humans in
bral trauma in World War I. Foerster pioneered the the general population who have left, right, or bilateral
use of intraoperative electrocorticography (ECoG)8 to language dominance is not known, about 9% of right-
localize seizure foci and developed the earliest maps handed patients scheduled for epilepsy surgery have
of language using this technique.9 These techniques atypical language dominance, while as many as 46% of
were later adapted by Wilder Penfield10 at the Mon- ambidextrous and 69% of strongly left-handed epilepsy
treal Neurological Institute, to make more precise and patients have atypical cerebral dominance.20 However,
detailed maps of cortical localization.11−13 His work12,14 atypical language lateralization is likely less common in
established that language could be localized to discrete normal humans and those with lesions acquired after
cortical areas, quite a bit smaller than the larger re- the first decade of life, by which time the ability of the
gions described by Broca and Wernicke. These areas previously nondominant hemisphere to take over major
were subsequently designated as “eloquent” and cortical language function is limited. A study of healthy adults,
stimulation mapping (CSM) with electrical currents has using repetitive transcranial magnetic stimulation (rTMS)
since been the mainstay for the preresection localiza- to temporarily disrupt language revealed atypical domi-
tion of language function. Penfield’s work established nance in only 4.2% of right handers and in 10.5% of left
that stimulation of the inferior frontal gyrus leads to handers.21 In addition, the vast majority of adult brain

203
204 SECTION II SYSTEMS

tumor patients with proven right hemispheric language


organization are left-handed.22
A pragmatic approach to decide when language lat-
eralization should precede localization is as follows. The
threshold to formally assess language lateralization in
patients undergoing resections for epilepsy should be
lower than in patients with tumors, especially in cases
where epilepsy involves the dominant hemisphere and
the age of onset of seizures is in the first decade of life.
In adult patients with gliomas, handedness, the clini-
cal history, and the bedside exam are critical metrics of
the need for formal measures of language lateralization.
Subtle disfluency, dysnomia, or altered comprehension
consequent to peritumoral edema, or to seizures at pre-
sentation are powerful indicators of laterality. In cases Figure 15–1. The major language pathways in the
where these are not noted, left-handedness should gen- dominant hemisphere—the arcuate fasciculus (blue),
erally be a trigger for testing to lateralizing language prior the inferior fronto-occipital fasciculus (orange), the
uncinate fasciculus (yellow ), and the inferior
to proceeding further.
longitudinal fasciculus (purple)
The stated “gold standard” for determining language
laterality is the Wada procedure.23 An intracarotid injec-
tion of sodium amobarbital (or methohexital or propofol nant hemisphere developed early in life (arteriovenous
or etomidate)24 is carried out into each carotid artery. malformations (AVMs) or epilepsy). In these situations,
After injection, the patient’s responses to a battery of functional imaging modalities can be viewed as screen-
expressive and receptive language tests and memory ing tests. Patients in whom activation is not definitively
function are measured.25 A temporary aphasia is usu- lateralized can then be studied with the Wada test. In
ally induced when the dominant hemisphere is injected. addition, combined use of Wada testing and functional
However, Wada testing is invasive, costly, and carries imaging may be useful in cases where both hemispheres
some risk.26,27 This prompted the utilization of nonin- support language function.35,36
vasive functional assessments of laterality—functional A newer, task independent measure of language
magnetic resonance imaging (fMRI),28−31 magnetoen- laterality relies on the evaluation of the white matter
cephalography (MEG)32,33 —and other less commonly pathways subserving language function. The arcuate fas-
used measures (positron emission tomography (PET), ciculus (AF) connects cortical areas in the frontal lobe
functional transcranial Doppler’s and TMS) to assess important for articulation to areas in parietal and tempo-
hemispheric dominance for language. Of these, fMRI ral lobes important for comprehension7,37−39 and can be
is the most widely used. The strongest correlation be- identified using diffusion tensor imaging (DTI).40 Given
tween Wada and fMRI determined laterality is obtained the rapid rates at which language is perceived and pro-
by comparing activity in Broca’s area with its contralat- duced, it is reasonable to expect the presence of highly
eral homolog.29 anisotropic fiber pathways in the AF of the dominant
It is important to recognize that the classical meth- hemisphere to mediate the rapid flow of information.41,42
ods of functional localization, the Wada procedure and Comparative estimates of the numbers of the compo-
electrical CSM, produce transient “lesions” of the brain. nent fibers of the AF and their anisotropy may allow for
On the other hand, functional imaging techniques reveal the lateralization of the language dominant hemisphere43
regions involved in, but not necessarily essential for the (Figs. 15–1 and 15–2). The broad clinical applicability of
task being performed. In addition, enormous variability this new strategy awaits further evaluation.
exists across centers in how functional imaging studies
are performed, the paradigms used, the scanner type,
and the thresholds used for the purpose of laterality es- 䉴 CORTICAL AND SUBCORTICAL
timation. Lastly, this testing needs a nonsedated, coop- SUBSTRATES OF LANGUAGE
erative patient able to reliably perform the tasks in the
confines of a scanner. Patients with even minor degrees Language can be considered as a disseminated cogni-
of claustrophobia or cognitive/language limitations can- tive process reliant upon interdependent nodes essen-
not be easily evaluated. While many centers have moved tial for subfunctions such as phonology, semantics, and
away from routinely performing the Wada procedure syntax.44 From a clinical standpoint, a broad understand-
for language lateralization in patients with epilepsy,34 ing of the language system is essential, as it allows the
it remains relevant in patients with unusual handedness surgeon to integrate data from invasive and noninva-
and in those where the lesion in the presumed domi- sive mapping strategies into devising a plan for a safe
CHAPTER 15 MAPPING OF HUMAN LANGUAGE 205

The classic anatomical model of language pro-


cessing proposed by Lichtheim48 was derived from le-
sional studies of Broca5 and Wernicke.6 In this model,
language expression is localized to the left posterior
inferior frontal lobe (Broca’s area) and language com-
prehension to the posterior superior temporal lobe
(an area with vaguely defined boundaries—Wernicke’s
area). The classical model also describes the AF,49,50
connecting these two areas. This classical model is
an underdeveloped predecessor that does not include
other anatomical substrates of language.51−53 In addi-
tion, terms such as comprehensive and expressive, used
to describe posterior and anterior language sites are per-
haps simplistic, as semantic and phonological processes
occur in both Broca’s and Wernicke’s areas. A more con-
temporary view of the functional roles of distinct brain
regions is listed in Table 15–1. These regions have been
A
incorporated into a plethora of models of language that
seek to depict information flow between them.
We now briefly review four contemporary mod-
els of language, which deal with three major language
processes. These models integrate data about cortical
and subcortical language sites. Levelt’s model of lexi-
cal access54,55 breaks down the process of naming into
an overtly serial process in which a core conceptual-
syntactic entity called the “lemma” is selected in the
anterior middle temporal gyrus (MTG), followed by mor-
phophonological code selection in posterior MTG and
Wernicke’s area, and syllabification in Broca’s area, fol-
lowed by eventual articulation using primary sensorimo-
tor cortex (Fig. 15–3).
A model integrating written and spoken language
comprehension,53 derived using functional imaging and
B neuropsychological data proposes that auditory word
analysis occurs in the superior temporal cortex and vi-
Figure 15–2. Representation of three major language sual analysis occurs in the posterior inferior temporal
pathways—the arcuate fasciculus (AF—green), the gyrus (visual word form area) and in temporo-occipital
inferior longitudinal fasciculus (ILF—yellow ), and the cortex. This model holds that semantic processes involve
uncinate fasciculus (UF—blue) in a given patient (Fig.
a network including the angular gyrus and the anterior
15–2B) and the laterality index computed using a - b/a
+ b method for each fiber pathway (Fig. 15–2A) at
inferior temporal cortex and that lexical retrieval occurs
increasing thresholds for the mean fractional via two routes: a semantic route through posterior infe-
anisotropy along each fiber pathway. The AF is the rior temporal cortex, and a nonsemantic route through
best pathway among these three for lateralizing the posterior superior temporal cortex (Wernicke’s area).
language function (From Ellmore T M, Beauchamp Friderici’s model56 of sentence processing invokes
M S, Breier J I, et al. Temporal lobe white matter a temporofrontal network that identifies and integrates
asymmetry and language laterality in epilepsy
syntactic and semantic information. According to this
patients. Neuroimage 2010;49:2033–2044.)
model, syntactic processing occurs before semantic pro-
cessing with integration of the two only in the later stages
of language processing.
resection. Traditionally, the focus of mapping has been Hickok and Poeppel57 modeled speech perception
to localize cortical substrates. However, it is now quite by proposing that that two asymmetric bidirectional au-
apparent that critical language pathways that mediate in- ditory streams originate from each superior temporal
formation flow also need to be localized and preserved gyrus. The ventral semantic stream projects to the pos-
to prevent language impairment.45,46 Newer models of terior middle temporal gyrus, and interfaces between
the language system should therefore also incorporate phonological representations of speech in the supe-
interregional connectivity of cortical substrates.47 rior temporal gyrus and widely distributed conceptual
206 SECTION II SYSTEMS

䉴 TABLE 15–1. CORTICAL SUBREGIONS THAT ARE COMPONENTS OF THE LANGUAGE SYSTEM

Eponym Location Function

Broca’s area Pars opercularis and pars triangularis Phonological assembly and encoding
of inferior frontal gyrus Semantic decoding
Wernicke’s area Posterior superior temporal gyrus Phonological decoding and repetition
Posterior middle temporal gyrus (MTG) Posterior MTG Lexical and semantic processing
Geschwind’s territory Angular and supramarginal gyri Semantic and lexical processing
Visual word form area Posterior inferior temporal gyrus Lexical decoding
Parahippocampal place area Posterior parahippocampal gyrus Place recognition
Fusiform face area Posterior occipitotemporal gyrus Face recognition
Anterior insula Anterior insula Articulatory planning
Premotor cortex Caudal middle frontal gyrus Articulatory planning
Supplementary motor area Posterior superior frontal gyrus Articulatory planning
Dorsolateral prefrontal cortex Rostral middle frontal gyrus Verbal working memory
Primary sensorimotor cortex Inferior precentral gyrus Articulation

Note: The commonly used descriptor is in column 1, the anatomic locus in column 2, and the functional role most commonly ascribed
to the region is in the last column.

representations. The dorsal phonological stream projects In addition to these four “corticocentric” models of
to the inferior parietal lobe and ultimately to frontal re- language, others have details of the roles of subcortical
gions, and maps sound to its articulatory representations. pathways mediating information flow in the language
This network provides a mechanism for the develop- system.51,47 There is increasing recognition that the ar-
ment and maintenance of “parity” between auditory and cuate AF40,58,59 and the inferior fronto-occipital fasciculi
motor representations of speech. (IFOF)60 play crucial roles in fluent language production.

Figure 15–3. Schematic representation of a meta-analysis of functional imaging tasks


during word production. Colors in the two panels indicate relations between regions
and functional processing components. The numbers indicate the time windows
(in milliseconds) during which the regions are activated in picture naming. The right
panel is the time course of the picture naming process. (From Indefrey P, Levelt W J.
The spatial and temporal signatures of word production components. Cognition
2004;92:101–144.)
CHAPTER 15 MAPPING OF HUMAN LANGUAGE 207

Figure 15–4. Corepresentation of schematic representation of the AF and cortical


language regions. Distinct subregions of the AF connect anterior versus posterior
components of Broca’s area with the middle and superior temporal gyri respectively.
These cortical–subcortical units are postulated to mediate specific language
processes. (From Glasser M F, Rilling J K. DTI tractography of the human brain’s
language pathways. Cereb Cortex 2008;18:2471–2482.)

At the current time, the existing data propose a partici- cal electrical currents—or CSM is carried out in one of
patory, noncritical role for the uncinate fasciculus (UF)61 two clinical settings. The first, historically older method,
and the inferior longitudinal fasciculus (ILF)62 in nam- involves the use of a bipolar probe to stimulate a small
ing and fluent speech production. Glasser and Rilling51 volume of cortex and is carried out in an operating room
have related the Hickok and Poeppel57 model and the with the cortex exposed by a craniotomy. It is char-
Price model to the connectivity of anterior and posterior acterized as intraoperative CSM (iCSM). The other, ex-
language systems by the components of the AF (Fig. traoperative CSM (eCSM),63 involves passing controlled
15–4). Such models integrate information about corti- currents into semichronically implanted subdural elec-
cal and subcortical substrates, and are likely to provide trodes (SDEs) and is typically carried out in the epilepsy
more ecologically valid and clinically relevant predic- monitoring unit, with ongoing video-electroencepha-
tions about the impact of planned cerebral lesions dur- lographic monitoring. The choice between iCSM and
ing neurosurgical procedures. eCSM is largely dictated by the patients’ diagnosis, and
their ability to participate in an awake craniotomy. In
patients undergoing SDE placement to localize seizure
䉴 MAPPING METHODS onset sites, eCSM is generally performed. Rarely SDEs
are implanted purely for purposes of eCSM, such as in
This sections outlines methods used to localize language. patients too young, too claustrophobic, or with very vas-
Details about methodological aspects not in this portion cular lesions (AVMs) that cannot be safely mapped using
of the text can be found in the references listed and iCSM. While maps created using eCSM and iCSM are gen-
in chapters on the relevant method elsewhere in this erally similar, there are some relative pros and cons of
book. each method. The amount of cortical surface covered by
iCSM is generally smaller than that for eCSM, as regions
beyond the craniotomy are not stimulated. Also, there is
DIRECT CORTICAL STIMULATION a limited amount of time available for sophisticated cog-
nitive or sensorimotor testing in the operating room, im-
This is the classical, time-tested method for localizing posing limits on data acquisition using iCSM. However,
language function using the application of artificial cur- the sites of stimulation in eCSM are fixed, while they
rents directly to the cortex. Language mapping using fo- can be dynamically varied for iCSM to accommodate
208 SECTION II SYSTEMS

t Object naming in an overt manner, using pictorial


1–15 mA stimuli from the Boston naming test71 or other
similar sources.72−74
t Repetition of spoken words or phrases.
t Auditory comprehension using the token test or
200–500 μS
one/two step commands.75
50 Hz t Spontaneous speech—counting, reciting the al-
phabet, nursery rhymes, etc.—useful in mapping
Figure 15–5. Characteristics of the electrical current anterior language sites and in mapping patients
typically used for both intraoperative and
with prominent verbal deficits.
extraoperative cortical stimulation mapping. The t
balanced square wave current minimizes charge Verb generation from pictures of objects, actions,
deposition. or from written nouns.
t Reading sentences out aloud.
t Auditory naming—naming driven by a phrase or
for sulcal anatomy and to tailor resection margins pre- a sentence (e.g., question—what a king wears on
cisely. Also, the iCSM stimulating probe has electrodes his head—answer—crown).
5 mm apart, which provides a higher resolution spatial
map than accomplished with traditional SDEs that have Caveats
10 mm interelectrode spacing.
The neural substrates involved in language tasks may
vary based on the degree to which the same task is
Methods overlearned.76 If CSM is performed using a small group
of stimuli, a region may be nonessential only for an over-
In either case, mapping is carried out with the patient
learned task.77 To minimize such confounds, it is useful
on anticonvulsants after he or she is familiarized with
to have a large battery of stimuli per task and to minimize
the testing process, using a balanced faradic current,
repetition of individual stimuli. Electrodes or stimula-
delivered at 50 Hz. The duration of each set of waves
tion sites where the task results in a delay, paraphrasias,
varies from 200 to 500 microseconds, with longer du-
anomia, or speech arrest are noted, as are those locations
ration waves used for mapping in children. A train of
where stimulation produces overt phenomenology. CSM
these waves is delivered for 3–5 seconds during task
carries a small risk of producing seizures. A seizure dur-
performance, via two contacts located on the corti-
ing stimulation may severely curtail the mapping process
cal surface (Fig. 15–5). Stimulation currents range from
or lead to a postponement of the planned resection. To
1 to 15 mA, and mapping is performed at 15 mA or at
minimize this possibility, patients should be on thera-
1 mA below the potential that results in persistent after-
peutic doses of anticonvulsants prior to mapping. It is
discharges (AD),64 overt phenomenology or discomfort,
also useful to administer an intravenous loading dose
whichever is lowest. It is preferable to maximize currents
of phenytoin or fosphenytoin before commencing map-
at each cortical site regardless of adjacent AD threshold
ping. Despite these measures, stimulation of perilesional
rather than to map the entire cortex at a single current
cortex can result in seizure induction. In the setting of
level.65,66 Concurrent ECoG is essential to monitor for
iCSM, this can be controlled by irrigation of the brain
ADs and seizures. It also provides visible evidence of
surface with ice-cold saline.78 In the context of eCSM,
completeness of the electrical circuit, by the presence of
the seizure can sometimes be aborted with a second
artifacts in the recording. The passage of currents meet-
pulse of stimulation79,80 applied at the same electrodes at
ing these parameters has been shown to be generally
the peak of the negativity of the AD. These CSM tech-
safe and does not appear to cause neuronal injury.67 The
niques have been successfully used to map language in
pulse generators commonly used for mapping are the
patients as young as 4 years and as old as 80 years of
Ojemann stimulator OCS-1 (Integra LifeSciences Corp,
age.81
Plainsboro, New Jersey), a portable, constant current,
battery-powered unit that generates biphasic rectangular
Overview of Published Series
waveforms with 5–100 Hz, 0.1–2 milliseconds duration,
0–20 mA and is powered by four 9-volt alkaline batteries, Several compilations of CSM data resulting from maps
and the S88 Grass stimulator and variants thereof (Grass of large numbers of patients have been reported. The
technologies, West Warwick, Rhode Island), which can best known of these is George Ojemann’s seminal
generate one or two trains of biphasic rectangular waves publication82 that is based on intraoperative awake map-
with greater variations of range (0.01–1000 Hz, 0.01– ping of 117 left hemispheric dominant patients. This
10,000 milliseconds duration, 0–1000 mA). revealed that language sites are diffusely organized
The tasks used for CSM vary across institutions, but all around the left perisylvian region, with additional
the ones commonly used are68−70 : language sites in the middle frontal gyrus and less
CHAPTER 15 MAPPING OF HUMAN LANGUAGE 209

commonly in the superior frontal gyrus. The middle and lation maps or surface anatomic landmarks. A compari-
inferior gyri of the anterior temporal lobe were relatively son of a large series of standard right versus left tempo-
bereft of language function, and no sites associated with ral lobectomies, in whom typical language lateralization
anomia were found in the inferior temporal gyrus. Sim- had been shown by Wada testing revealed that in the
ilar findings have been noted in a larger series of 250 absence of language mapping, about 7% of patients did
patients undergoing awake language mapping for the have a significant deterioration in naming ability at 6
resection of gliomas,83 suggesting that it is safe to carry months postoperatively.84
out an aggressive resection of a neoplasm in the absence
of any positive stimulation sites. Both these large series Limitations
suggest that significant variability exists in the organiza-
CSM is a technology that has been used in an almost
tion of language sites between individual humans. This
unchanged form for over a century. Its limitations are
variability is much greater in the location of posterior
therefore well known and understood. Testing in a given
temporal and temporoparietal language sites15 than in
individual may be difficult or inconsistent.66 The method
the location of frontal lobar language sites. Language
is less than ideally sensitive—perhaps due to the fact that
does not always reside in the typical confines of classi-
only one-third of cortex is on the surface, and therefore
cal language sites. In Ojemann’s work, no language sites
testable—no language sites are found in Wernicke’s area
were found in the vicinity of Wernicke’s area in 36% of
in 36% and none in Broca’s area in 21%.82,85 Depend-
subjects and none were found in Broca’s area in 21%.82
ing on the tasks used for mapping, it may not consis-
Other work revealed that no posterior language areas
tently prevent postoperative language deficits.70 Lastly,
were identifiable in 10% and no anterior language site
three pragmatic considerations impact upon the utility
could be identified in 9% of patients.81 CSM reveals in-
of CSM—the induction of seizures, the need for an ad-
teresting characteristics about “essential” language sites.
ditional procedure (SDE placement for eCSM) or oper-
Their cortical surface area is rather small: less than or
ative time (for iCSM), and constraints on the extent of
equal to 6 cm2 in 85% of individuals, especially so in
language assessment imposed by the time available.
individuals with a high verbal fluency. There is a strik-
Older studies have suggested that the local spread
ing subspecialization of cortical regions with regards to
of cortical currents and hence the range of the dis-
different language functions, with distinct loci in many
ruptive effect of stimulation on cognitive processing is
instances for naming, reading, phoneme identification,
localized.86,87 However, remote changes in local field po-
and so forth. More than half of all sites localized by
tentials (LFPs) occur following even a single suprathresh-
stimulation mapping are involved in only one aspect of
old electrical pulse lasting less than 0.3 milliseconds, as
language.15
evidenced by the recordings of cortico-cortical evoked
There is some diversity in opinion in regards to
potentials (CCEPs)88,89 (Fig. 15–6). It may well be use-
the extent of interpatient variability of language sites.
ful to view CSM as a process that disrupts information
A study of 45 patients with implanted SDEs,17 with ex-
flow in a broad network, rather than just at a specific site
haustive extraoperative language testing, and a specific
(Fig. 15–6).
evaluation of “negative” motor areas, revealed language
sites in and around classical Broca’s and Wernicke’s
Summary
locations and in the basal temporal lobe only. How-
ever, most awake mapping (iCSM) studies suggest that CSM strategies have provided unique insights into
otherwise. This disparity may be attributable either to the biology of human language and have served as
the statistical threshold used in the awake mapping the standard used to evaluate some models of language
techniques.82,83 These iCSM techniques make an incom- organization.90 Ojemann’s work, revealed the parallel
plete distinction between sites where a dysnomia is activation of multiple function specific modules during
always produced, versus those where it is produced language processing,85 a model that has stood in distinc-
above a certain statistical threshold. This raises ques- tion to traditional models of hierarchical serial processes
tions about whether these sites are truly essential for lan- starting with decoding in the posterior temporal lobe and
guage. These data may also be affected by the inherent eventual phonation by the inferior frontal lobe.91 This
limitations of making population language maps based parallel processing model is widely accepted today.92
solely on cortical surface anatomy. However, the multi- Surgical techniques that specifically target the resec-
tude of functional imaging studies and lesional analysis tion of mesial temporal lobe structures while minimizing
studies of language all suggest a significant amount of neocortical resections might appear to be a useful strat-
intersubject variability in language sites. Taken together, egy to avoid the need for mapping in cases of epilepsy
these findings suggest that language sites need to be lo- originating due to mesial temporal lobe sclerosis in the
calized for each individual and for each function that dominant hemisphere. However, no improvement in
is considered important. Their locations cannot be de- outcome has been noted with a selective transsylvian
rived reliably using heuristic strategies based on popu- approach.93 The reason for this may be the disregard for
210 SECTION II SYSTEMS

Figure 15–6. Corepresentation of diffusion tensor imaging pathways and cortico-


cortical evoked potentials (CCEPs) from stimulation of Broca’s area in a single
subject. Stimulation was carried out over Broca’s area (black electrodes) at 1 Hz,
10 mA, using pulses of 500-microsecond width. Subsequent N1 peak from the CCEP
response measured at all other electrodes represented in terms of amplitude (radius
of electrode) and latency (color scale). Electrodes without CCEP response within
40 milliseconds are depicted as white spheres. All pathways passing 1 cm of the
stimulating pair are shown as green streamlines. A clear relationship between the
tracts and the elicited CCEPs is noted.

subcortical damage, especially to important white matter have all been used to localize regions involved in the
fibers in the temporal stem, including the UF, the IFOF, component processes of speech production and lan-
and the cholinergic outflow from the nucleus basalis of guage perception. Of these, fMRI-mapping techniques
Meynert91 A subtemporal approach that spares most of possess the best spatial resolution, and several prior
the temporal neocortex may minimize the decline in comparative estimates of maps generated with BOLD-
verbal memory94 ; but this approach needs to be sub- fMRI and CSM have been made.
stantiated in languages that use syllabograms rather than
morphograms, and its long-term impact on temporal Comparisons with CSM
lobe epilepsy needs to be qualified.
What matters ultimately is the specificity and sensi- While efforts to supplant CSM for localizing regions in-
tivity of mapping results as related to the language out- volved in motor function with NIMS have been gen-
come following focal resections. In this regard, Ojemann erally successful,100 functional imaging methods have
and others have pointed out that carrying the resection had limited applicability for presurgical localization of
to within 1 cm of a site that leads to consistent anomia or language sites.101 This is especially true for temporal
speech arrest is likely to result in a permanent long-term and temporoparietal language regions,102−104 and less
verbal deficit.15,95 No such outcomes data are available so for frontal language sites.105 This limitation of func-
for other mapping strategies (which are discussed later). tional imaging is likely due to several reasons. The first
Though there is some plasticity96,97 in the language sys- is that these techniques provide a map of the network
tem in adults, it remains preferable to not have to rely involved in, but not of the nodes of that network essen-
upon it for a good long-term outcome following resec- tial to, language generation. This problem worsens with
tions. increasing sensitivity of the mapping technique used—
as more functional areas are detected by lowering the
statistical threshold used to create the language map—
FUNCTIONAL IMAGING the specificity of the map becomes lower. The second
reason is that the cognitive “load” of “simple” language
Modalities
tasks such as object naming that are typically used for
The concerns regarding the spatial specificity and sensi- CSM are relatively low—such that the neuronal activa-
tivity of CSM listed previously, along with the need for tion or its hemodynamic correlate are not easily dis-
additional procedures (SDE placement for eCSM) or op- cernible from background activity. This has led imagers
erative time (for iCSM), have led to efforts to supplant to use different tasks for generating the functional acti-
CSM for localizing language function with functional vation map, as compared to the tasks used during CSM,
imaging techniques. Positron emission tomography,98 leading to inherent mismatches between such compar-
fMRI,55 event-related potentials (ERPs), and MEG99 — isons. A third, and relatively tractable limitation relates to
CHAPTER 15 MAPPING OF HUMAN LANGUAGE 211

technical issues: spatial resolution of the imaging tech- DTI Tractography


nique; mismatches in co-registration of functional data
DTI uses the relative anisotropy induced by the pres-
sets with high-resolution MRI scan data used for intra-
ence of axonal cell membranes and myelin on the diffu-
operative stereotactic guidance; and problems with pa-
sion of water in the brain, to deduce the orientation of
tients’ abilities to comprehend the task or cooperate with
white matter tracts in each imaging voxel. As discussed
scanning.106 Lastly, speech production in both MEG107
previously and shown in Figs. 15–1 and 15–2, cortical
and clinical fMRI environments is generally covert (ex-
language sites are extensively interconnected. The AF
cept in event-related fMRI—which has worse signal to
connects Broca’s area with Wernicke’s area and is well
noise characteristics), as movement associated with ar-
known from fiber dissection techniques.114 DTI tractog-
ticulation produces artifacts and signal dropout. Covert
raphy has led to the identification of other components
speech does not activate some of the frontal opercular
of the AF of which both connect Geschwind’s area (in-
regions that are essential for articulation, limiting the ap-
ferior parietal lobe) with Broca’s area and Wernicke’s
plicability of these techniques for mapping these frontal
area.40 The AF mediates comprehension, repetition, and
language sites.105 Importantly though, functional imag-
semantic processes.7,115,116,117 The IFOF connects pos-
ing maps of language have been compared with CSM
terior occipitotemporal regions to anterior temporal re-
maps, but not to the patient’s functional state following
gions, linking disseminated components involved in
a resection.
object, face, and written words recognition62,116,118,119
and mediates semantic processes and naming. The UF
Summary connects the anterior temporal lobe with the medial and
Taken together, the published literature on functional lateral orbitofrontal cortex and is related to semantic
imaging suggests that while useful in lateralizing the processes and to confrontation naming.120−122 Electrical
language dominant hemisphere, functional imaging stimulation of these pathways—the AF,123,124 the ILF,62
techniques are largely unsuccessful in demonstrating and the IFOF—60 has been shown to interfere with lan-
absolute and reliable concordance between CSM guage production.
and functional imaging derived maps.106,108 Hence, A clue that the anatomical asymmetry in hemi-
stimulation-based mapping remains the prevailing stan- spheric connectivity may support different functional
dard for functional localization of language regions at roles was first evident, when it was noted that the direct
the current time.109 Maps derived using fMRI are of some segment of the AF linking Broca’s to Wernicke’s area,40
value in guiding CSM and in providing a better under- did not exist in the right hemisphere. Instead, in the
standing of language organization in patients with atyp- presumed nondominant hemisphere, only the two indi-
ical results following an intracarotid amytal procedure. rect segments that connect Broca’s and Wernicke’s areas
separately to Geschwind’s area exist. Evaluations of the
fractional anisotropy of the AF in each hemisphere reveal
STRUCTURAL IMAGING that differences in these measures may be sufficient in
some cases to categorize language dominance.43 The AF
Broca’s Morphology can also be used to predict the location of language sites.
Upto 80% of all essential language sites (ELS) are situated
As opposed to other functional regions that bear a
in intimate proximity to the terminations of the AF.58
strict relationship with cortical surface landmarks, (e.g.,
Intuitively, it stands to reason that ELS should exhibit
the central sulcus, the pli de passage moyen, or the
robust long-range connectivity, therefore a greater
calcarine fissure), most language regions do not bear
understanding of the relationship between cortical and
a constant relationship with the surface landmarks. In
subcortical language sites may allow for newer ways
the case of Broca’s area, the relationship between func-
of functional localization that are independent of both
tion and anatomy is relatively better defined, but not
the need to produce transient lesions and functional
invariant. CSM of the pars opercularis is often associated
activation.
with speech arrest, and CSM of pars triangularis can lead
to paraphrasic errors, errors in auditory cued naming and
disfluency. The component regions of the posterior in-
ferior frontal gyrus can be well visualized using sagittal LOCAL FIELD POTENTIAL
MRI images.110 Pars opercularis and triangularis in the RECORDINGS FROM INTRACRANIAL
language dominant hemisphere are generally accepted ELECTRODES
to constitute Broca’s area. Localizing these subregions
in relation to the planned resection, allows for guidance Functional imaging, CSM, and DTI methods taken to-
regarding CSM. In addition, the relative volumes of pars gether map a disseminated language network that in-
triangularis and111 pars opercularis may help predict lan- cludes regions outside traditional Broca’s, Wernicke’s,
guage lateralization.112,113 and Geschwind’s areas—for example the ventral
212 SECTION II SYSTEMS

temporo-occipital region, anterior lateral temporal lobe, imprecise in certain cases. The Wada test remains essen-
premotor cortex, and insula.125,55 However, those meth- tial to confirm language laterality in selected cases. In ad-
ods do not readily explain the dynamics of informa- dition to the methodological and pragmatic concerns dis-
tion processing within the disseminated network. ECoG cussed previously, there are special situations in regards
recordings from SDEs, while the patient is engaged in to language mapping, which should be considered.
task processing reflect the activity of small cortical neu-
ronal assemblies coherently activated during language
processes, and are characterized as LFPs.126 SDEs make it LANGUAGE MAPPING IN
possible to observe interactions in large cell assemblies, YOUNG CHILDREN
spanning distributed cortical regions, by sampling both
low- and high-frequency signal components such as SDEs may sometimes need to be implanted purely for
those in the gamma-frequency range, which are damped functional mapping in children, as some of them are
by the media around the brain.127 The local nature of LFP unable to participate in iCSM.90 Mapping language in
sampling greatly minimizes the “inverse problem” (com- children with CSM can be difficult, both due to poorly
puting the dynamic spatial locus as well as the com- developed language skills at an early age as well as due
ponent frequencies of an averaged electric or magnetic to the fact that it is generally difficult to disrupt cortical
signal) that confounds such measures made with MEG processing in children using electrical stimuli. Stimula-
and ERP. tion parameters that work well in adults often do not
Such recordings provide an unparalleled opportu- work well in children, possibly because partly myeli-
nity to study the dynamics of information flow during nated or unmyelinated nerve fibers need longer pulse
language processes. A localized desynchronization in durations to be activated.135 This difficulty may be over-
the alpha-beta 10–20 Hz range,82 greatest between 700 come by lengthening the pulse durations beyond the
and 1200 milliseconds after stimulus presentation, with usual 300 microseconds (upto 3500 microseconds in an
persisting gamma 30–200 Hz activity, occurs at most incremental fashion), coupled with stepwise progression
temporoparietal naming sites.126 Such response is not in the intensity, to arrive at the appropriate chronaxy136
typically associated with frontal naming sites. Other early for each individual child. If these measures are not taken,
work revealed that slow potential shifts at the onset of it is possible to obtain one or more false-negative sites
naming in frontal naming sites as well in motor cortex126 of stimulation in children.
and that changes in electrical activity in the frontal and
temporoparietal language sites occur in parallel and not
sequentially.81 LANGUAGE ORGANIZATION IN
More recent work has focused on studying corre- MULTILINGUAL PATIENTS
lated activity between language regions, at higher fre-
quency ranges.128 Activity in the high-gamma range Lesion studies in aphasic polyglots tell us little about
70–150 Hz, has been shown to be spatially correlated the organization of multiple languages in the cortex.
with CSM-derived language maps.129−132 At this time, the Plasticity and the difficulty in estimating premorbid lin-
degree of such correlation is poor, 40% specific and 84% guistic competence in each language, prevent definitive
sensitive.133 Optimization of the criteria used to define conclusions. In addition, the limited spatial resolution
sites of “significant” change, particularly in regards to the of functional neuroimaging techniques prevents precise
frequencies chosen to designate significance, may lead delineation of adjoining cortical regions that perform
to this technique becoming clinically meaningful for lan- similar functions in different languages.137 Hence, elec-
guage localization.134 trical mapping techniques possess unique characteristics
suited for the study of multilingualism.157 CSM maps re-
veal that naming areas in languages acquired later in life
(L2) cover larger cortical regions than the language that
䉴 CLINICAL APPLICATIONS AND is acquired earlier in life (L1). As proficiency in a lan-
CAVEATS OF MAPPING guage develops, the ELS related to it tend to become
STRATEGIES more compactly organized.81 CSM of bilingual patients,
often, though not always, reveals that sites responsible
As outlined previously the dominant and most proven for naming and reading in two different languages are
method for language localization in the current era re- distinct from each other,138,139 and that L2 specific sites
mains CSM. Newer technologies such as DTI and ECoG tend to be located exclusively in the posterior tempo-
may provide alternatives in the future. Functional imag- ral and parietal regions.145 This corresponds well to data
ing techniques are often adequate to confirm the lateral- obtained by functional imaging of individuals who learnt
ization of language suspected on clinical grounds, but it L2 late in life.137 On the basis of the spatial disparity of
must be remembered that all of these measures may be language sites in polyglots, mapping in each language
CHAPTER 15 MAPPING OF HUMAN LANGUAGE 213

that the individual is proficient in should be carried out, that use morphograms. Whether this actually results
whenever practicable, to minimize the possibility of a in improved verbal fluency without compromising
monolingual decline in verbal fluency.139 This is espe- seizure-free outcomes is unproven so far.
cially true if the proposed resection is proximate to the
posterior language sites.
FUNCTIONAL LOCALIZATION BASED
ON POPULATION MAPS AND THE
BASAL TEMPORAL LANGUAGE NOTION OF DEGENERACY

On the basis of clinicopathological data, Mills and Mar- The ability to co-register imaging datasets to each
tin postulated141 the existence of a “naming center” in other, with adjustments made for individual anatomic
the basal temporal lobe. The region has since been differences, allows for the generation of population
well described by CSM142 and functional neuroimaging maps of cerebral anatomy and functional activation. The
techniques.140 CSM studies have called the region the generation of high-resolution three-dimensional (3D)
basal temporal language area (BTLA) and stimulation cytoarchitectonic maps113 or spatial probability maps
here during naming, produces a specific deficit in con- from meta-analyses of functional imaging data156 that
frontation naming. In some instances, auditory and vi- are can be co-registered onto a patient’s 3D structural
sual comprehension may also be impaired.143,144,146,147 MRI provide novel ways for functional localization.
Both fMRI and CSM have shown the region to con- The four functions of interest in the neurosurgical
tain subdivisions, which preferentially process particular setting are language, motor control, vision, and mem-
visual stimuli, such as written words (visual word form ory. The cortical regions without which these functions
area),140 faces (fusiform face area),148 and places such as would no longer be considered to be normal are des-
buildings (parahippocampal place area). The represen- ignated as “eloquent.” This designation does not imply
tation of particular categories of objects or faces appears that other neural functions are not essential or that there
to not be sharply delimited, but do display a spatial to- are necessarily noneloquent brain regions that can be
pography across the ventral temporal region.149,150 resected with impunity.158 Instead, it is a categorization
The term BTLA is sometimes loosely used to de- of cortical regions based on the lasting effects of le-
scribe language regions in the entire ventral surface of sions and the impacts of these lesions upon the activ-
the temporal lobe, from the temporal tip to the temporo- ities of daily living. The term “eloquent” has been used
occipital junction, and including the inferior tempo- here, to allude to cortical regions without which lan-
ral gyrus. Comprehensive characterization of the region guage function could not be discerned as normal.5,6 It
by Lüders and colleagues,146 and others143,147,151 have is clear from multiple lesional studies that a reorganiza-
found that this language region involves the parahip- tion of language function can occur even in the adult
pocampal and fusiform gyri, extending from 11 to 75 brain.159 Recently, it has been shown that sites localized
mm caudal to the temporal tip. In most cases, however, by CSM demonstrate spatial plasticity both acutely160 and
the BTLA is restricted to a zone that has a 2–4 cm extent as slow-growing lesions encroach upon them.161 While
in the rostrocaudal axis.144 By varying the onset of elec- resection of language sites generally does result in an
trical interference relative to a behavioral task (so-called acute language impediment, significant and sometimes
time slicing) it appears that processing of verbal object complete recovery of function can occur, over a pe-
meaning in the occipitotemporal gyrus occurs 450–750 riod of 3–6 months.96,97 Lasting deficits are more likely
milliseconds after stimulus presentation.152 when the subcortical substrates of function are dam-
Deficits seen after dominant temporal lobectomy, aged in some way.7 The best level of recovery is gen-
comparing patients who undergo a resection of the erally observed, when compensatory recruitments take
BTLA is resected with those where it is not, involve only place in the perilesional area, along the rim of the in-
confrontation naming. Deficits in nonvisual language jured tissue, rather than in the homologous region of the
processing are generally not noted.153,154 Recent studies nondominant, contra-lateral hemisphere.162−164 These
in Japanese subjects who speak using Kanji and Kana findings, along with the fact that large amounts of domi-
characters have shown that the BTLA may play an nant temporal neocortex can be resected with relatively
important role in connecting visual semantic decoders few deficits support the concept of an implicit degener-
(Kanji characters, like Cantonese, uses morphograms; acy in language systems. The term “degenerate” is used
Kana characters, like English, are syllabograms) with to describe structurally discrete neural elements that per-
phonological processors.154 To this end, there is a spe- form the same function.165 This approach has recently
cial interest in designing surgical strategies that would been used to create a map of “truly essential” brain re-
spare as much of the fusiform gyrus as possible by using gions, and such approaches may have a bearing on the
a subtemporal-transventricular-transchoroidal approach need for as well as the interpretation of cortical mapping
for hippocampectomy,155 for speakers of languages studies.166 Holistic approaches that integrate the results
214 SECTION II SYSTEMS

of functional imaging, lesional analysis, CSM, and the 20. Isaacs KL, Barr WB, Nelson PK, Devinsky O. De-
potential for plasticity (based on age, site of planned re- gree of handedness and cerebral dominance. Neurology
section) would probably provide a more precise way of 2006;66:1855-1858.
determining which cortical regions are truly eloquent. 21. Khedr EM, Hamed E, Said A, Basahi J. Handedness
and language cerebral lateralization. Eur J Appl Physiol
2002;87:469-473.
22. Chang EF, Wang DD, Perry DW, Barbaro NM, Berger
MS. Homotopic organization of essential language sites
REFERENCES in right and bilateral cerebral hemispheric dominance. J
Neurosurg 2011;114:893-902.
1. Von Frisch K. The Dance Language and Orientation of 23. Wada J, Rasmussen T. Intracarotid injection of sodium
Bees. Harvard University Press, Cambridge, MA 1967. amytal for the lateralization of cerebral speech domi-
2. Pepperberg IM. The Alex Studies: Cognitive and Com- nance. 1960. J Neurosurg 2007;106:1117-1133.
municative Abilities of Grey Parrots. Harvard University 24. Jones-Gotman M, Sziklas V, Djordjevic J, et al. Etomidate
Press, Cambridge, MA 2000. speech and memory test (eSAM): a new drug and im-
3. Arnold K, Zuberbuhler K. Language evolution: semantic proved intracarotid procedure. Neurology 2005;65:1723-
combinations in primate calls. Nature 2006;441:303. 1729.
4. Herman LM, Richards DG, Wolz JP. Comprehen- 25. Breier JI, Simos PG, Zouridakis G, et al. Language dom-
sion of sentences by bottlenosed dolphins. Cognition inance determined by magnetic source imaging: a com-
1984;16:129-219. parison with the Wada procedure. Neurology 1999;53:938-
5. Broca P. Remarques sur le sie‘ge de la faculte’ du langage 945.
articule’; suivies d’une observation d’aphemie. Bulletin de 26. Kemp S, Wilkinson K, Caswell H, Reynders H, Baker
la Societe Anatomique 1861;2:330-357. G. The base rate of Wada test failure. Epilepsy Behav
6. Wernicke C. Der Aphasische Symtomen Komplex. Bres- 2008;13:630-633.
lau, Poland: Max Cohn & Weigert, 1874. 27. Loddenkemper T, Morris HH, Moddel G. Complications
7. Dronkers NF, Plaisant O, Iba-Zizen MT, Cabanis EA. Paul during the Wada test. Epilepsy Behav 2008;13:551-553.
Broca’s historic cases: high resolution MR imaging of the 28. Binder JR, Swanson SJ, Hammeke TA, et al. Determination
brains of Leborgne and Lelong. Brain 2007;130:1432-1441. of language dominance using functional MRI: a compari-
8. Foerster O, Altenburger H. Electrobiologische Vorgänge son with the Wada test. Neurology 1996;46:978-984.
an der menschlichen Hirnrinde. Dtsch Z Nervenheilkd 29. Chlebus P, Mikl M, Brazdil M, Pazourkova M, Krupa P,
1934;135:277-288. Rektor I. FMRI evaluation of hemispheric language dom-
9. Foerster O. The motor cortex in man in light of Hughlings inance using various methods of laterality index calcula-
Jackson’s doctrines. Brain 1936;59(2):10-159. tion. Exp Brain Res 2007;179:365-374.
10. Foerster O, Penfield W. The structural basis of traumatic 30. Binder JR, Frost JA, Hammeke TA, Cox RW, Rao SM, Prieto
epilepsy and results of radical operation. Brain 1930;53:8- T. Human brain language areas identified by functional
119. magnetic resonance imaging. J Neurosci 1997;17:353-362.
11. Penfield W, Boldrey E. Somatic motor and sensory rep- 31. Benke T, Koylu B, Visani P, et al. Language lateralization
resentation in the cerebral cortex of man as studied by in temporal lobe epilepsy: a comparison between fMRI
electrical stimulation. Brain 1937;60:389-443. and the Wada Test. Epilepsia 2006;47:1308-1319.
12. Penfield W, Roberts L. Speech and Brain Mechanisms. 32. Papanicolaou AC, Simos PG, Castillo EM, et al. Magne-
Princeton, New Jersey: Princeton University Press, 1959. tocephalography: a noninvasive alternative to the Wada
13. Penfield W, Jasper H. Epilepsy and the Functional procedure. J Neurosurg 2004;100:867-876.
Anatomy of the Human Brain. Boston: Little Brown, 1954. 33. Hirata M, Kato A, Taniguchi M, et al. Determination of lan-
14. Penfield W, Rasmussen A. The Cerebral Cortex of Man. guage dominance with synthetic aperture magnetometry:
New York: Macmillan, 1957, pp. 11-65. comparison with the Wada test. Neuroimage 2004;23:46-
15. Ojemann GA. Individual variability in cortical localization 53.
of language. J Neurosurg 1979;50:164-169. 34. Binder JR. Functional MRI is a valid noninvasive alterna-
16. Berger MS, Kincaid J, Ojemann GA, Lettich E. Brain tive to Wada testing. Epilepsy Behav 2011;20:214-222.
mapping techniques to maximize resection, safety, and 35. Moddel G, Lineweaver T, Schuele SU, Reinholz J, Lod-
seizure control in children with brain tumors. Neuro- denkemper T. Atypical language lateralization in epilepsy
surgery 1989;25:786-792. patients. Epilepsia 2009;50:1505-1516.
17. Schaffler L, Luders HO, Beck GJ. Quantitative comparison 36. Lee D, Swanson SJ, Sabsevitz DS, et al. Functional MRI and
of language deficits produced by extraoperative electri- Wada studies in patients with interhemispheric dissocia-
cal stimulation of Broca’s, Wernicke’s, and basal temporal tion of language functions. Epilepsy Behav 2008;13:350-
language areas. Epilepsia 1996;37:463-475. 356.
18. Schwartz TH, Devinsky O, Doyle W, Perrine K. Preop- 37. Dronkers N, Ogar J. Brain areas involved in speech pro-
erative predictors of anterior temporal language areas. J duction. Brain 2004;127:1461-1462.
Neurosurg 1998;89:962-970. 38. Makris N, Kennedy DN, McInerney S, et al. Segmenta-
19. Whitaker HA, Ojemann GA. Graded localisation of nam- tion of subcomponents within the superior longitudinal
ing from electrical stimulation mapping of left cerebral fascicle in humans: a quantitative, in vivo, DT-MRI study.
cortex. Nature 1977;270:50-51. Cereb Cortex 2005;15:854-869.
CHAPTER 15 MAPPING OF HUMAN LANGUAGE 215

39. Schmahmann J, Pandya D. Fiber Pathways of the Brain. 60. Martino J, Vergani F, Robles SG, Duffau H. New insights
New York: Oxford University Press, Oxford, UK 2006. into the anatomic dissection of the temporal stem with
40. Catani M, Jones DK, ffytche DH. Perisylvian language net- special emphasis on the inferior fronto-occipital fascicu-
works of the human brain. Ann Neurol 2005;57:8-16. lus: implications in surgical approach to left mesiotempo-
41. Behrens TE, Berg HJ, Jbabdi S, Rushworth MF, Wool- ral and temporoinsular structures. Neurosurgery 2010;66:
rich MW. Probabilistic diffusion tractography with mul- 4-12.
tiple fibre orientations: what can we gain? Neuroimage 61. Duffau H, Gatignol P, Moritz-Gasser S, Mandonnet E. Is
2007;34:144-155. the left uncinate fasciculus essential for language? A cere-
42. Tandon N, O’Neill T, Beauchamp M, Ellmore T. Rapid bral stimulation study. J Neurol 2009;256:382-389.
identification of the arcuate fasciculus using an FA thresh- 62. Lesser RP, Luders H, Klem G, et al. Extraoperative corti-
olding technique. Epilepsia 2007;48(1):161. cal functional localization in patients with epilepsy. J Clin
43. Ellmore TM, Beauchamp MS, Breier JI, et al. Temporal Neurophysiol 1987;4:27-53.
lobe white matter asymmetry and language laterality in 63. Mandonnet E, Nouet A, Gatignol P, Capelle L, Duffau H.
epilepsy patients. Neuroimage 2010;49:2033-2044. Does the left inferior longitudinal fasciculus play a role in
44. Pulvermuller F, Fadiga L. Active perception: sensorimotor language? A brain stimulation study. Brain 2007;130:623-
circuits as a cortical basis for language. Nat Rev Neurosci 629.
2010;11:351-360. 64. Gordon B, Lesser RP, Rance NE, Hart J Jr, Webber R, Ue-
45. Duffau H. The anatomo-functional connectivity of lan- matsu S, Fisher RS. Parameters for direct cortical electrical
guage revisited. New insights provided by electrostimu- stimulation in the human: histopathologic confirmation.
lation and tractography. Neuropsychologia 2008;46:927- Electroencephalogr Clin Neurophysiol 1990;75:371-377.
934. 65. Sederberg PB, Kahana MJ, Howard MW, Donner EJ, Mad-
46. Duffau H, Leroy M, Gatignol P. Cortico-subcortical organi- sen JR. Theta and gamma oscillations during encod-
zation of language networks in the right hemisphere: an ing predict subsequent recall. J Neurosci 2003;23:10809-
electrostimulation study in left-handers. Neuropsycholo- 10814.
gia 2008;46:3197-3209. 66. Lesser RP, Lueders H, Dinner DS, Hahn J, Cohen L. The
47. Turken AU, Dronkers NF. The neural architecture of the location of speech and writing functions in the frontal lan-
language comprehension network: converging evidence guage area. Results of extraoperative cortical stimulation.
from lesion and connectivity analyses. Front Syst Neurosci Brain 1984;107:275-291.
2011;5:1. 67. Pouratian N, Cannestra AF, Bookheimer SY, Martin NA,
48. Lichtheim L. On Aphasia. Brain 1885;7:433-484. Toga AW. Variability of intraoperative electrocortical stim-
49. Geschwind N. Disconnexion syndromes in animals and ulation mapping parameters across and within individu-
man. I. Brain 1965;88:237-294. als. J Neurosurg 2004;101:458-466.
50. Geschwind N. Disconnexion syndromes in animals and 68. Lesser R, Gordon B, Uematsu S. Electrical stimulation and
man. II. Brain 1965;88:585-644. language. J Clin Neurophysiol 1994;11:191-204.
51. Glasser MF, Rilling JK. DTI tractography of the human 69. Hamberger MJ, Goodman RR, Perrine K, Tamny T.
brain’s language pathways. Cereb Cortex 2008;18:2471- Anatomic dissociation of auditory and visual naming in
2482. the lateral temporal cortex. Neurology 2001;56:56-61.
52. Binder JR, Desai RH, Graves WW, Conant LL. Where is 70. Hamberger MJ, Seidel WT, McKhann GM 2nd, Perrine K,
the semantic system? A critical review and meta-analysis Goodman RR. Brain stimulation reveals critical auditory
of 120 functional neuroimaging studies. Cereb Cortex naming cortex. Brain 2005;128:2742-2749.
2009;19:2767-2796. 71. Goodglass H, Kaplan E, Weintraub S. Boston Naming Test
53. Price CJ. The anatomy of language: contributions from (Experimental Edition). Boston University, Boston, MA
functional neuroimaging. J Anat 2000;197(Pt 3):335-359. 1976.
54. Levelt WJ, Roelofs A, Meyer AS. A theory of lexical ac- 72. Dunn LM, Hottel JV. Peabody picture vocabulary test per-
cess in speech production. Behav Brain Sci 1999;22:1–38; formance of trainable mentally retarded children. Am J
discussion 38-75. Ment Defic 1961;65:448-452.
55. Indefrey P, Levelt WJ. The spatial and temporal signatures 73. Dunn L. Peabody Picture Vocabulary Test – Revised. Circle
of word production components. Cognition 2004;92:101- Pines, MN: American Guidance Service, 1981.
144. 74. Snodgrass JG, Vanderwart M. A standardized set of 260
56. Friederici AD. Towards a neural basis of auditory sentence pictures: norms for name agreement, image agreement,
processing. Trends Cogn Sci 2002;6:78-84. familiarity, and visual complexity. J Exp Psychol Hum
57. Hickok G, Poeppel D. Dorsal and ventral streams: a Learn 1980;6:174-215.
framework for understanding aspects of the functional 75. Schaffler L, Luders HO, Dinner DS, Lesser RP, Chelune GJ.
anatomy of language. Cognition 2004;92:67-99. Comprehension deficits elicited by electrical stimulation
58. Ellmore TM, Beauchamp MS, O’Neill TJ, Dreyer S, Tandon of Broca’s area. Brain 1993;116:695-715.
N. Relationships between essential cortical language sites 76. Petersen SE, van Mier H, Fiez JA, Raichle ME. The
and subcortical pathways. J Neurosurg 2009;111:755-766. effects of practice on the functional anatomy of task
59. Rilling JK, Glasser MF, Preuss TM, Ma X, Zhao T, Hu X, performance. Proc Natl Acad Sci U S A 1998;95:853-
Behrens TE. The evolution of the arcuate fasciculus re- 860.
vealed with comparative DTI. Nat Neurosci 2008;11:426- 77. Ojemann JG, Ojemann GA, Lettich E. Cortical stimulation
428. mapping of language cortex by using a verb generation
216 SECTION II SYSTEMS

task: effects of learning and comparison to mapping based 96. Duffau H, Denvil D, Capelle L. Long term reshaping of
on object naming. J Neurosurg 2002;97:33-38. language, sensory, and motor maps after glioma resection:
78. Sartorius CJ, Berger MS. Rapid termination of intraopera- a new parameter to integrate in the surgical strategy. J
tive stimulation-evoked seizures with application of cold Neurol Neurosurg Psychiatry 2002;72:511-516.
Ringer’s lactate to the cortex. Technical note. J Neurosurg 97. Duffau H, Capelle L, Denvil D, et al. Functional recovery
1998;88:349-351. after surgical resection of low grade gliomas in eloquent
79. Motamedi GK, Lesser RP, Miglioretti DL, et al. Optimiz- brain: hypothesis of brain compensation. J Neurol Neuro-
ing parameters for terminating cortical afterdischarges surg Psychiatry 2003;74:901-907.
with pulse stimulation. [erratum appears in Epilepsia 2002 98. Petersen SE, Fox PT, Posner MI, Mintun M, Raichle ME.
Nov;43(11):1441]. Epilepsia 2002;43:836-846. Positron emission tomographic studies of the cortical
80. Lesser RP, Kim SH, Beyderman L, et al. Brief bursts of anatomy of single-word processing. Nature 1988;331:585-
pulse stimulation terminate afterdischarges caused by cor- 589.
tical stimulation. Neurology 1999;53:2073-2081. 99. Billingsley-Marshall RL, Clear T, Mencl WE, et al. A com-
81. Ojemann G. Organizations of language cortex derived parison of functional MRI and magnetoencephalogra-
from investigations during neurosurgery. Semin Neuro- phy for receptive language mapping. J Neurosci Methods
sciences 1990;2:297-305. 2007;161:306-313.
82. Ojemann G, Ojemann J, Lettich E, Berger M. Cortical lan- 100. Schiffbauer H, Berger MS, Ferrari P, Freudenstein D, Row-
guage localization in left, dominant hemisphere. An elec- ley HA, Roberts TP. Preoperative magnetic source imaging
trical stimulation mapping investigation in 117 patients. J for brain tumor surgery: a quantitative comparison with
Neurosurg 1989;71:316-326. intraoperative sensory and motor mapping. J Neurosurg
83. Sanai N, Mirzadeh Z, Berger MS. Functional outcome after 2002;97:1333-1342.
language mapping for glioma resection. N Engl J Med 101. Roux FE, Boulanouar K, Lotterie JA, Mejdoubi M, LeSage
2008;358:18-27. JP, Berry I. Language functional magnetic resonance imag-
84. Gleissner U, Helmstaedter C, Schramm J, Elger CE. Mem- ing in preoperative assessment of language areas: cor-
ory outcome after selective amygdalohippocampectomy relation with direct cortical stimulation. Neurosurgery
in patients with temporal lobe epilepsy: one-year follow- 2003;52:1335-1345; discussion 1345-1347.
up. Epilepsia 2004;45:960-962. 102. Pouratian N, Bookheimer SY, Rex DE, Martin NA, Toga
85. Ojemann GA. Cortical organization of language. J Neu- AW. Utility of preoperative functional magnetic resonance
rosci 1991;11:2281-2287. imaging for identifying language cortices in patients with
86. Ojemann GA, Whitaker HA. Language localization and vascular malformations. J Neurosurg 2002;97:21-32.
variability. Brain Lang 1978;6:239-260. 103. Rutten GJ, Ramsey NF, van Rijen PC, Noordmans HJ, van
87. Haglund MM, Ojemann GA, Blasdel GG. Optical imaging Veelen CW. Development of a functional magnetic res-
of bipolar cortical stimulation. J Neurosurg 1993;78:785- onance imaging protocol for intraoperative localization
793. of critical temporoparietal language areas. Ann Neurol
88. Matsumoto R, Nair DR, LaPresto E, et al. Functional con- 2002;51:350-360.
nectivity in the human language system: a cortico-cortical 104. Brannen JH, Badie B, Moritz CH, Quigley M, Meyerand
evoked potential study. Brain 2004;127:2316-2330. ME, Haughton VM. Reliability of functional MR imag-
89. Matsumoto R, Nair DR, Lapresto E, Bingaman W, Shibasaki ing with word-generation tasks for mapping Broca’s area.
H, Luders HO. Functional connectivity in human cortical AJNR Am J Neuroradiol 2001;22:1711-1718.
motor system: a cortico-cortical evoked potential study. 105. Petrovich N, Holodny AI, Tabar V, et al. Discordance
Brain 2007;130(Pt 1):181-97. between functional magnetic resonance imaging during
90. Ojemann SG, Berger MS, Lettich E, Ojemann GA. Localiza- silent speech tasks and intraoperative speech arrest. J
tion of language function in children: results of electrical Neurosurg 2005;103:267-274.
stimulation mapping. J Neurosurg 2003;98:465-470. 106. Schlosser MJ, Luby M, Spencer DD, Awad IA, McCarthy G.
91. Geschwind N. The organization of language and the Comparative localization of auditory comprehension by
brain. Science 1970;170:940-944. using functional magnetic resonance imaging and cortical
92. Vigneau M, Beaucousin V, Herve PY, et al. Meta-analyzing stimulation. J Neurosurg 1999;91:626-635.
left hemisphere language areas: phonology, semantics, 107. Simos PG, Papanicolaou AC, Breier JI, et al. Localiza-
and sentence processing. Neuroimage 2006;30:1414-1432. tion of language-specific cortex by using magnetic source
93. Gleissner U, Helmstaedter C, Schramm J, Elger CE. Mem- imaging and electrical stimulation mapping. J Neurosurg
ory outcome after selective amygdalohippocampectomy: 1999;91:787-796.
a study in 140 patients with temporal lobe epilepsy. 108. Giussani C, Roux FE, Ojemann J, Sganzerla EP, Pirillo
Epilepsia 2002;43:87-95. D, Papagno C. Is preoperative functional magnetic res-
94. Hori T, Yamane F, Ochiai T, et al. Selective subtemporal onance imaging reliable for language areas mapping in
amygdalohippocampectomy for refractory temporal lobe brain tumor surgery? Review of language functional mag-
epilepsy: operative and neuropsychological outcomes. netic resonance imaging and direct cortical stimulation
J Neurosurg 2007;106:134-141. correlation studies. Neurosurgery 2010;66:113-120.
95. Haglund MM, Berger MS, Shamseldin M, Lettich E, Oje- 109. Billingsley-Marshall RL, Simos PG, Papanicolaou AC. Re-
mann GA. Cortical localization of temporal lobe language liability and validity of functional neuroimaging tech-
sites in patients with gliomas. Neurosurgery 1994;34:567- niques for identifying language-critical areas in children
576; discussion 576. and adults. Dev Neuropsychol 2004;26:541-563.
CHAPTER 15 MAPPING OF HUMAN LANGUAGE 217

110. Naidich TP, Valavanis AG, Kubik S. Anatomic relationships word production in spoken and sign language. Neurology
along the low-middle convexity: Part I–Normal specimens 2001;57:2045-2053.
and magnetic resonance imaging. Neurosurgery 1995;36: 129. Sinai A, Bowers CW, Crainiceanu CM, et al. Electrocortico-
517-532. graphic high gamma activity versus electrical cortical stim-
111. Foundas AL, Leonard CM, Gilmore RL, Fennell EB, Heil- ulation mapping of naming. Brain 2005;128:1556-1570.
man KM. Pars triangularis asymmetry and language dom- 130. Crone NE, Miglioretti DL, Gordon B, et al. Functional
inance. Proc Natl Acad Sci U S A 1996;93:719-722. mapping of human sensorimotor cortex with electro-
112. Dorsaint-Pierre R, Penhune VB, Watkins KE, et al. Asym- corticographic spectral analysis. I. Alpha and beta
metries of the planum temporale and Heschl’s gyrus: re- event-related desynchronization. Brain 1998;121(Pt 12):
lationship to language lateralization. Brain 2006;129:1164- 2271-2299.
1176. 131. Crone NE, Miglioretti DL, Gordon B, Lesser RP. Functional
113. Amunts K, Schleicher A, Burgel U, Mohlberg H, Uylings mapping of human sensorimotor cortex with electrocor-
HB, Zilles K. Broca’s region revisited: cytoarchitecture and ticographic spectral analysis. II. Event-related synchro-
intersubject variability. J Comp Neurol 1999;412:319-341. nization in the gamma band. Brain 1998;121(Pt 12):
114. Ture U, Yasargil MG, Friedman AH, Al-Mefty O. Fiber 2301-2315.
dissection technique: lateral aspect of the brain. Neuro- 132. Tanji K, Suzuki K, Delorme A, Shamoto H, Nakasato N.
surgery 2000;47:417-426; discussion 426-427. High-frequency gamma-band activity in the basal tem-
115. Catani M, Allin MP, Husain M, et al. Symmetries in human poral cortex during picture-naming and lexical-decision
brain language pathways correlate with verbal recall. Proc tasks. J Neurosci 2005;25:3287-3293.
Natl Acad Sci U S A 2007;104:17163-17168. 133. Wu M, Wisneski K, Schalk G, et al. Electrocorticographic
116. Catani M, Mesulam M. The arcuate fasciculus and the dis- frequency alteration mapping for extraoperative local-
connection theme in language and aphasia: history and ization of speech cortex. Neurosurgery 2010;66:E407-
current state. Cortex 2008;44:953-961. E409.
117. Rauschecker AM, Deutsch GK, Ben-Shachar M, Schwartz- 134. Brunner P, Ritaccio AL, Lynch TM, et al. A practical proce-
man A, Perry LM, Dougherty RF. Reading impairment in a dure for real-time functional mapping of eloquent cortex
patient with missing arcuate fasciculus. Neuropsychologia using electrocorticographic signals in humans. Epilepsy
2009;47:180-194. Behav 2009;15:278-286.
118. Bauer RM, Trobe JD. Visual memory and perceptual 135. Ranck JB Jr. Which elements are excited in electrical stim-
impairments in prosopagnosia. J Clin Neuroophthalmol ulation of mammalian central nervous system: a review.
1984;4:39-46. Brain Res 1975;98:417-440.
119. Epelbaum S, Pinel P, Gaillard R, et al. Pure alexia as a 136. Jayakar P, Alvarez LA, Duchowny MS, Resnick TJ. A safe
disconnection syndrome: new diffusion imaging evidence and effective paradigm to functionally map the cortex in
for an old concept. Cortex 2008;44:962-974. childhood. J Clin Neurophysiol 1992;9:288-293.
120. Lu LH, Crosson B, Nadeau SE, et al. Category-specific 137. Perani D, Abutalebi J. The neural basis of first and second
naming deficits for objects and actions: semantic at- language processing. Curr Opin Neurobiol 2005;15:202-
tribute and grammatical role hypotheses. Neuropsycholo- 206.
gia 2002;40:1608-1621. 138. Roux FE, Tremoulet M. Organization of language areas in
121. Govindan RM, Makki MI, Sundaram SK, Juhasz C, Chugani bilingual patients: a cortical stimulation study. J Neurosurg
HT. Diffusion tensor analysis of temporal and extra- 2002;97:857-864.
temporal lobe tracts in temporal lobe epilepsy. Epilepsy 139. Bello L, Acerbi F, Giussani C, et al. Intraoperative lan-
Res 2008;80:30-41. guage localization in multilingual patients with gliomas.
122. Rodrigo S, Oppenheim C, Chassoux F, et al. Uncinate fas- Neurosurgery 2006;59:115-125; discussion 115-125.
ciculus fiber tracking in mesial temporal lobe epilepsy. 140. Cohen L, Dehaene S. Specialization within the ventral
Initial findings. Eur Radiol 2007;17:1663-1668. stream: the case for the visual word form area. Neuroim-
123. Duffau H, Gatignol P, Mandonnet E, et al. New insights age 2004;22:466-476.
into the anatomo-functional connectivity of the semantic 141. Mills C, Martin E. Aphasia and agraphia in some practical
system: a study using cortico-subcortical electrostimula- surgical relationships. JAMA 1912;59:1513-1518.
tions. Brain 2005;128:797-810. 142. Huettel SA, McKeown MJ, Song AW, Hart S, Spencer DD,
124. Duffau H, Gatignol P, Denvil D, Lopes M, Capelle L. The Allison T, McCarthy G. Linking hemodynamic and elec-
articulatory loop: study of the subcortical connectivity by trophysiological measures of brain activity: evidence from
electrostimulation. Neuroreport 2003;14:2005-2008. functional MRI and intracranial field potentials. Cereb Cor-
125. Wise RJ, Greene J, Buchel C, Scott SK. Brain regions in- tex 2004;14:165-173.
volved in articulation. Lancet 1999;353:1057-1061. 143. Burnstine TH, Lesser RP, Hart J Jr, et al. Characterization
126. Fried I, Ojemann GA, Fetz EE. Language-related potentials of the basal temporal language area in patients with left
specific to human language cortex. Science 1981;212:353- temporal lobe epilepsy. Neurology 1990;40:966-970.
356. 144. Krauss GL, Fisher R, Plate C, Hart J, Uematsu S, Gordon
127. Canolty RT, Edwards E, Dalal SS, et al. High gamma power B, Lesser RP. Cognitive effects of resecting basal temporal
is phase-locked to theta oscillations in human neocortex. language areas. Epilepsia 1996;37:476-483.
Science 2006;313:1626-1628. 145. Lucas TH 2nd, McKhann GM 2nd, Ojemann GA. Func-
128. Crone NE, Hao L, Hart BJ, Boatman D, Lesser RP, Irizarry tional separation of languages in the bilingual brain: a
R, Gordon B. Electrocorticographic gamma activity during comparison of electrical stimulation language mapping
218 SECTION II SYSTEMS

in 25 bilingual patients and 117 monolingual control pa- area for intractable mesial temporal lobe epilepsy: pre-
tients. J Neurosurg 2004;101:449-457. liminary results. Epilepsia 2006;47:1347-1353.
146. Luders H, Lesser RP, Hahn J, Dinner DS, Morris H, Resor S, 156. Fox PT, Huang A, Parsons LM, et al. Location-probability
Harrison M. Basal temporal language area demonstrated profiles for the mouth region of human primary motor-
by electrical stimulation. Neurology 1986;36:505-510. sensory cortex: model and validation. Neuroimage
147. Luders H, Lesser RP, Hahn J, Dinner DS, Morris HH, 2001;13:196-209.
Wyllie E, Godoy J. Basal temporal language area. Brain 157. Ojemann GA, Whitaker HA. The bilingual brain. Arch
1991;11(Pt 2):743-754. Neurol 1978;35:409-412.
148. Haxby JV, Gobbini MI, Furey ML, Ishai A, Schouten JL, 158. Fried I. The myth of eloquent cortex, or what is non-
Pietrini P. Distributed and overlapping representations eloquent cortex? J Neurosurg 1993;78:1009-1010.
of faces and objects in ventral temporal cortex. Science 159. Rasmussen T, Milner B. The role of early left-brain injury
2001;293:2425-2430. in determining lateralization of cerebral speech functions.
149. Ishai A, Ungerleider LG, Martin A, Schouten JL, Haxby JV. Ann N Y Acad Sci 1977;299:355-369.
Distributed representation of objects in the human ventral 160. Duffau H. Acute functional reorganisation of the human
visual pathway. Proc Natl Acad Sci U S A 1999;96:9379- motor cortex during resection of central lesions: a study
9384. using intraoperative brain mapping. J Neurol Neurosurg
150. Gauthier I, Skudlarski P, Gore JC, Anderson AW. Expertise Psychiatry 2001;70:506-513.
for cars and birds recruits brain areas involved in face 161. Desmurget M, Bonnetblanc F, Duffau H. Contrasting acute
recognition. Nat Neurosci 2000;3:191-197. and slow-growing lesions: a new door to brain plasticity.
151. Schaffler L, Luders HO, Morris HH 3rd, Wyllie E. Anatomic Brain 2007;130:898-914.
distribution of cortical language sites in the basal temporal 162. Heiss WD, Kessler J, Thiel A, Ghaemi M, Karbe H. Dif-
language area in patients with left temporal lobe epilepsy. ferential capacity of left and right hemispheric areas for
Epilepsia 1994;35:525-528. compensation of poststroke aphasia. Ann Neurol 1999;45:
152. Hart J Jr, Crone NE, Lesser RP, et al. Temporal dynamics 430-438.
of verbal object comprehension. Proc Natl Acad Sci U S A 163. Heiss WD, Thiel A. A proposed regional hierarchy in re-
1998;95:6498-6503. covery of post-stroke aphasia. Brain Lang 2006;98:118-
153. Ishitobi M, Nakasato N, Suzuki K, Nagamatsu K, Shamoto 123.
H, Yoshimoto T. Remote discharges in the posterior lan- 164. Price CJ, Crinion J. The latest on functional imaging stud-
guage area during basal temporal stimulation. Neurore- ies of aphasic stroke. Curr Opin Neurol 2005;18:429-434.
port 2000;11:2997-3000. 165. Price CJ, Friston KJ. Degeneracy and cognitive anatomy.
154. Usui K, Ikeda A, Takayama M, et al. Conversion of Trends Cogn Sci 2002;6:416-421.
semantic information into phonological representation: 166. Ius T, Angelini E, Thiebaut de Schotten M, Mandonnet
a function in left posterior basal temporal area. Brain E, Duffau H. Evidence for potentials and limitations of
2003;126:632-641. brain plasticity using an atlas of functional resectability
155. Mikuni N, Miyamoto S, Ikeda A, et al. Subtemporal hip- of WHO grade II gliomas: towards a “minimal common
pocampectomy preserving the basal temporal language brain.” Neuroimage 2011;56:992-1000.
Chapter 16
Mapping of the Human
Visual System
Muhammad M. Abd-El-Barr 1 , Mario F. Dulay 2 , Paul Richard 3 , William H. Bosking4 ,
and Daniel Yoshor 5
1
Department of Neurosurgery, University of Florida, Gainesville, Florida
2
Department of Neurosurgery, The Methodist Hospital Neurological Institute, Houston, Texas
3
Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
4
Max Planck Florida Institute, Jupiter, Florida
5
Department of Neurosurgery, Baylor College of Medicine, and Neuroscience Center, St. Luke’s Episcopal Hospital,
Houston, Texas

䉴 INTRODUCTION localization of visual cortex may help in choosing a sur-


gical route to minimize functional deficits, and may also
Vision is a complex process in which characteristics of define the optimal limits for resection of some brain
our environment such as color, contrast, location, mo- lesions. Mapping techniques are particularly useful in
tion, and orientation are encoded by functionally spe- planning and guiding the resections of lesions that are
cialized clusters of neurons. A large percentage (∼20%) not anatomically well defined. This includes cases of
of the human cortex is dedicated to the processing of nonlesional epilepsy or infiltrative brain tumors. Map-
visual information, highlighting its importance to hu- ping helps to conceptualize risks associated with surgery
man behavior, and survival. Perhaps the most impor- that can be used in the preoperative counseling of a pa-
tant property of the visual image is its spatial arrange- tient. Mapping visual cortex can also be useful for un-
ment, so it not surprising that spatial representations of derstanding visual abnormalities in patients who present
visual images are found throughout the visual cortex.1 In with visual complaints. Finally, mapping in patients of-
human visual cortex, there are multiple distinct “retino- fers a unique opportunity for study of visual processing
topic” maps that preserve (with varying levels of fidelity) in the human brain.
the spatial content of visual stimuli projected onto the
retina. This is analogous to the multiple somatotopic 䉴 HISTORICAL PERSPECTIVE
maps of the human body that exist for sensory or mo-
tor processing.2,3 Each of these areas differ in terms of Although earlier work had established the basic anatomy
receptive field properties, connectivity, and physiology, of the visual pathways, it was only in the 18th and 19th
and are thought to contribute to visual processing in dis- centuries that we first gained an understanding of how
tinct ways. Collectively, these areas contribute to human visual information from the retina is physically mapped
behaviors such as perception of object shape, color, and in the cerebral cortex. In 1776, Francisco Gennari pro-
motion, object discrimination, visual-motor control, vi- vided the first indication that the cortex was not homo-
sual memory, and visual planning. Mapping of the visual geneous by describing a white stripe, later known as
cortex is a difficult process because of its complexity, the stria of Gennari, that was especially prominent in
but technological advances in invasive and noninvasive the area we now know as primary visual cortex, “striate
structural and functional mapping techniques have led cortex,” or V1.4 Subsequently, evidence from multiple
to a better understanding of the human brain and visual disciplines converged on the conclusion that this area of
processing. occipital cortex was associated with the sense of vision.5
This chapter will review the role of structural and This included anatomical work by Gratiolet (1854) and
functional mapping techniques in providing useful in- later by Flechsig (1886) that identified that the optic ra-
formation in a clinical setting, particularly for addressing diations targeted the same area that had been identi-
clinical problems that involve the visual system. These fied by Gennari, and experiments in animals by Munk
techniques are important for providing optimal care in (1881) and Shafer (1888), which showed a loss of vision
clinical neurological settings. For example, preoperative associated with lesions of this area.5,6 Furthermore, by

219
220 SECTION II SYSTEMS

extensive study of clinical cases, Henschen identi- tomography (PET) technology.24−26 However, this tech-
fied damage to this area as causing homonymous nique requires the injection of radioactive tracers, and
hemianopia.7 Henschen, also introduced the idea of the has limited spatial resolution. The development of func-
“cortical retina” and was able to correctly map the lower tional MRI (fMRI) allowed for the truly noninvasive and
retina (upper visual field) to the lower bank of the cal- accurate mapping of visual cortex in humans.27−30 Imag-
carine fissure, and the upper retina (lower visual field) ing studies in humans and monkeys have revealed many
to the upper bank.7,8 However, he incorrectly located similarities in the organization of visual cortex between
the foveal representation to the anterior portion of the the two species, especially for early visual areas such as
cuneus.7 V1 and V2, but have also highlighted important differ-
A more accurate depiction of how the visual field ences in functional organization of the two species that
is mapped onto the cortex resulted from the work of appear as early as V3 and V3a.27,31−33
Inouye, who studied the visual field deficits produced Intracranial recordings from subdural electrodes
by focal lesions to the cerebral cortex sustained by sol- placed during clinical monitoring of epilepsy patients
diers in the Russo-Japanese war, and then Holmes, who offer a unique opportunity to directly measure electrical
studied similar patients from World War I.8−11 Inouye activity from human visual cortex and have been used
and Holmes corrected Henschen’s mistake by placing to confirm and extend observations made by monkey
the cortical representation of the fovea near the poste- electrophysiology and human fMRI.34,35
rior pole of occipital cortex, and the peripheral represen-
tation further anterior in the cuneus. The Holmes map
was the standard for many years until it was updated 䉴 NEUROANATOMY,
by use of magnetic resonance imaging (MRI) to exam- NEUROPHYSIOLOGY, AND
ine the location of lesions associated with specific visual ORGANIZATIONAL PRINCIPLES
field deficits.12 OF VISUAL CORTEX
Further refinements of our understanding of the
retinotopic organization of primary visual cortex came ANATOMY AND PHYSIOLOGY
from animal experiments that used evoked poten-
tials recorded from contacts placed on the cortical There are a number of pathways in the brain that
surface.13,14 These investigations revealed the large over- incorporate information transmitted by light. Some of
representation of the fovea in primary visual cortex, and these pathways involve the control of circadian rhythm
were the first to formally introduce the idea of the corti- or pupillary reactivity, and function relatively indepen-
cal magnification factor, which is a quantitative descrip- dently of conscious awareness. In this chapter, we will
tion of the physical amount of visual cortex that rep- only consider pathways directly relevant to perceptual
resents one degree of the visual field. Later work by processing of visual objects, although the other path-
Hubel and Wiesel revealed that a columnar organiza- ways may be important in clinical settings.36,37 Much
tion of cells with similar response properties existed in of our understanding of the perceptual processing of
primary visual cortex,15 as had been demonstrated in visual objects originates from studying nonhuman pri-
the somatosensory cortex,16 and that not only the loca- mates; while, many findings in visual cortex of nonhu-
tion of an object in visual space, but other features such man primates may be generalized to humans, important
as the orientation of a stimulus are precisely mapped differences do exist and should be considered.38
across the cortical surface.17 The ability of activity in lo- Visual processing begins when information from
calized portions of human primary visual cortex to actu- the environment reaches the receptive field of photore-
ally support the perception of a simple visual sensation, ceptors in the retina (see Fig. 16–1).39 These receptors
or phosphene, in a corresponding part of visual space (which consist mostly of rods and cones) transmit their
was uncovered with electrical stimulation studies con- responses to retinal ganglion cells (RGCs) through spe-
ducted by Penfield,18 Brindley and Lewin,19 and Dobelle cialized interneurons (bipolar cells). Axons of the RGCs
and Mladejovsky.20 form the optic nerve and travel to the optic chiasm where
Later, the demonstration that visual space is mapped inputs from the nasal retina cross over to the contralat-
not once but many times in the cerebral cortex initially eral hemisphere, while inputs from the temporal retina
came from electrophysiological investigations in nonhu- continue ipsilaterally. After this partial decussation at
man primates. The response properties of cells in these the optic chiasm, the visual fibers then travel via the
areas, and the connections between areas, have been optic tract to synapse in (1) the lateral geniculate nu-
used to sort these visually responsive areas into path- cleus (LGN) of the thalamus, and (2) the superior col-
ways and hierarchies.21−23 liculi of the midbrain. The latter projections to the col-
The ability to demonstrate the complete map of liculi are important in controlling saccades, or fast eye
visual space using noninvasive techniques in humans movements, and are not thought to be directly involved
emerged with the development of positron emission in visual perception.40 From the LGN, magnocellular
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 221

Right visual field

Left visual field

Temporal

Nasal
Temporal

Optic
chiasm

Amygdala

Pulvinar nucleus

Lateral geniculate
nucleus
Superior colliculus

Optic radiation

Primary visual
cortex

Figure 16–1. Visual pathways from retina to primary visual cortex. Light from the right
visual field (shown in blue) is projected onto the nasal hemiretina of the right eye and
the temporal hemiretina of the left eye. Axons of the retinal ganglion cells (RGCs) form
the optic nerve. Those from the nasal retinal ganglion cells, cross the midline at the
level of the optic chiasm, while those from the ganglion cells of the temporal retina
remain ipsilateral. After the optic chiasm, the axons of the contralateral nasal RGCs
and the ipsilateral temporal RGCs form the optic tract. The majority of these axons will
synapse in the lateral geniculate nucleus (LGN), while some will synapse in the
superior colliculus (shown as dotted lines). The superior colliculus is thought to play
roles in saccades and papillary control. After forming synapses in the LGN, the
postsynaptic cells will send axons (which form the optic radiations) to the ipsilateral
primary visual cortex. The optic radiations are split into a superior and inferior divisions
(not shown), which are important in considering clinical resections. (Reproduced with
permission of WM Norton & Company, from Gazzaniga MS, Ivry RB, Mangun GR.
Cognitive Neuroscience: The Biology of the Mind. Vol 1. New York: Norton, 2002,
(various pagings).

(M-cells, specialized for high-contrast and noncolored to portions of extrastriate cortex.42,43 Before terminat-
stimuli) and parvocellular (P-cells, specialized for low- ing in the occipital lobe, the optic radiations split into
contrast and color stimuli) cells send projections through a superior division that forms the retrolenticular limb of
the retrolenticular fibers of internal capsule and then, the internal capsule and ends in the upper bank of the
via the optic radiations, to layer four of the primary calcarine sulcus (cuneus), and an inferior division that
visual cortex, located along the banks of the calcarine travels through the temporal lobe (Meyer’s loop) and
fissure in the occipital lobe.41 A third pathway originat- ends in the lower bank of the calcarine sulcus (lingual
ing from koniocellular cells of the LGN also projects gyrus). Following V1, processing continues in a large ar-
via the optic radiations and appears to carry specific ray of other visual cortical areas.44,45 Figure 16–2 gives an
color information to the superficial layers of V1, and indication of the complexity of the primate visual system,
222 SECTION II SYSTEMS

Figure 16–2. Hierarchy of visual areas in the macaque monkey. In addition to the
retina and the lateral geniculate nucleus, over 30 areas of the cerebral cortex are
thought to play a role in visual information processing. Most of the connections shown
have been shown to be bidirectional. The order of each area within the visual
hierarchy was established by an analysis of the laminar specificity of feed-forward,
feedback, and lateral connections. (Reproduced with the permission of Oxford
University Press, from Felleman DJ, Van Essen DC. Distributed hierarchical processing
in the primate cerebral cortex. Cereb Cortex 1991;1(1):1–47.)

as demonstrated by the number of reciprocal subcorti- or ventral aspects of the cerebral cortex. This physical
cal and cortical anatomical connections among the more and functional segregation appears to be well conserved
than 30 visual areas that have been identified in the between nonhuman primates and humans.38 Fig. 16–3
macaque monkey.46 provides a schematic illustration of this model of cortical
Beginning early on in the hierarchy of visual cortex, organization.47
there is an organized segregation of information into two In humans, the ventral stream lies in the occipi-
functionally specialized streams, or pathways, that deal totemporal areas of the cerebral cortex. This pathway
with different aspects of visual processing.23 Following is commonly referred to as the “what” pathway because
primary visual cortex, these two pathways diverge and it is involved in processing of object color and form, and
follow routes that proceed toward either more dorsal in object recognition. From V1 at the occipital pole, the
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 223

magno-, parvo-, and koniocellular is created in the V1


that is then fed into these two streams in specific ways.52

ORGANIZATIONAL PRINCIPLES

In addition to the division of visual processing into dorsal


and ventral streams (where and what pathways), there
are other important principles that underlie the organi-
zation of visual cortex, and which are useful for map-
ping visual areas. Retinotopic organization is a recurrent
motif, and is perhaps the most important principle in
identifying and delineating discrete visual areas. Visual
areas are said to be retinotopically organized, if there
is a structured correspondence between points on the
retina (and hence the visual field) and points in the cor-
tex. For example, in V1, as already noted, the upper
bank of the calcarine sulcus responds strongly to stimu-
lation in the lower half of the visual field, and the lower
bank of the calcarine sulcus responds to stimulation of
Figure 16–3. Parallel processing in the visual system. the upper half of visual field. In general, within each
This highly simplified schematic indicates the main
visual area, we expect to find only one map of visual
flow of information through two processing streams
that are thought to contribute to the perception of space, and we expect most parts of visual space to be
object location (the “where” pathway also known as represented, although not necessarily equally. The most
“dorsal,” or “parietal”), and object shape and color obvious departure from equal representation is the large
(the “what” pathway, also known as “ventral,” or overrepresentation of the central visual field in V1 where
“temporal”). LNG, lateral geniculate nucleus. the central 10◦ of the visual field occupies about half of
(Reproduced with the permission of Lippincott the surface area of V1.12
Williams & Wilkins, from Ozdemir A, Black PM.
Retinotopic mapping techniques use visual stimu-
Mapping of Human Visual Cortex. Neurosurgery
Quarterly 2005;15(2):65–71. lation of a set of different points in the visual field to
reconstruct the representation of the visual field on the
cortex. Since many visual areas contain retinotopic
ventral stream projects to V2, V3, hV4, and on to the maps, this organization can then be used to define and
inferior temporal lobe (IT), in a generally posterior to label the borders of individual visual areas. The vertical
anterior projection. The “h” in hV4 is used to differen- and horizontal meridians of the visual field often define
tiate this human visual area from macaque V4, as there the border between two visual areas. A subset of the to-
is considerable debate about the homology of this area pographically organized visual areas are presented on a
between species.1 three-dimensional model of the occipital–temporal cor-
The dorsal stream, often called the “where” path- tex and on a flat map in Fig. 16–4.53
way, is involved in processing object location, object For individual locations within a visual area, the re-
motion, and control of the eyes and arms, especially ceptive field of that site can be defined as the region of
when visual information is used to guide motor behav- visual space that will drive significant activity from the
iors (saccades or reaching). Beginning at V1, the dorsal site in question when stimulated with a suitable stim-
stream projects to V2 and V3, V5/hMT+ (analogous, but ulus. For example, Fig. 16–5 depicts the determination
not entirely the same as area MT/V5 in the macaque) of the receptive field for a site located on the lateral
in the medial temporal area, and to the inferior parietal surface of occipital cortex of a human subject recorded
lobule.48 with a standard clinical subdural (cortical surface) elec-
The schematic shown in Fig. 16–3 is highly simpli- trode (2.2 mm diameter recording surface) using a small
fied, and there is considerable evidence to suggest that checkerboard stimuli.35
the ventral and dorsal streams are not completely seg- Another important organizational principle is that
regated, or functionally independent, as there are sig- the visual system processes information in a hierarchi-
nificant interactions among the two pathways at multi- cal manner. The strongest evidence that the cortex is
ple stages.22,49−51 Specifically, it would be inappropriate organized in this manner comes from the study of feed-
to think of the dorsal and ventral streams in the cor- forward, feedback, and lateral connections between vi-
tex as simple continuations of the pathways begun in sual areas. These connections terminate in a stereotyped
the retina. Instead, a recombination of input from the fashion in each cortical area, and can be used to trace
224 SECTION II SYSTEMS

B F

A C D E G

Figure 16–4. Functional specialization in human visual cortex. (A) Topographically


organized visual areas shown on a flat map of occipito-temporal cortex in the Visible
Man. (B) The same areas on a medial view of the occipital lobe. (C) The same regions
on a lateral view of the hemisphere. (D) Cortical regions implicated in processing of
color. Green dots denote centers of activation foci, and lighter green denotes cortex
with the 10-mm uncertainty limit. (E) Cortical regions implicated in the processing of
motion (M); form (F); form and color (FC); form and motion (FM); and motion, color,
and spatial relations (MCS). Question marks denote additional regions potentially
involved in processing of color (green), motion (red), and spatial relations (yellow ).
(F) The same functional specializations shown on lateral and ventral 3D views. (G) The
same pattern on extensively smoothed surfaces. (Reproduced with the permission of
the National Academy of Sciences, from Van Essen DC, Drury HA, Joshi S, et al.
Functional and structural mapping of human cerebral cortex: solutions are in the
surfaces. Proc Natl Acad Sci USA 1998;95:788–795.

the main routes of information flow through the cortex.46 a still finer level, parallel processing occurs within each
Corresponding with this anatomically defined hierarchy, retinotopically organized visual area, since columns of
there are a set of functional changes that are observed cells within each part of the area process information
with movement further along the cortical hierarchy. for different parts of the visual field in a largely parallel
These changes include larger receptive fields, longer fashion.
response latencies, more complex response proper- One final principle of functional organization that
ties, and less orderly visual field maps. For example, is important to mention is that there are likely to be
within the ventral pathway, visual processing begins in multiple features that are mapped within each area. For
V1, which has short response latencies, small receptive example, in primary visual cortex, it is not just visual
fields, and which can be activated by relatively simple space that is mapped across the surface, but also ori-
visual stimuli such as lines or gratings. Visual process- entation preference, and ocular dominance. Orientation
ing in later areas of the ventral pathway tends to have preference refers to the fact that each column of cells in
longer latencies, larger receptive fields, and requires V1 tends to respond best to lines or contours placed at
more complex stimuli for activation, such as objects or a particular angle or orientation in the visual field. Oc-
faces. ular dominance refers to the fact that inputs from the
Parallel processing is another guiding organizational contralateral and ipsilateral eye are routed through spe-
principle for understanding the visual system. Parallel cific layers of the LGN and then to partially segregated
processing, or organization, exists at many levels within columns of cells in V1. To fully predict the responses of
the visual cortex. As we have already discussed, at a a given site in V1, it is therefore necessary to know not
coarse level, the visual system processes information si- just the receptive field of that site, but also the preferred
multaneously through multiple pathways.23,48 Further- orientation and ocular preference. Other visual areas are
more, within each pathway, a single visual stimulus typ- likely to contain maps of other stimulus features, and it
ically activates at least several different cortical areas. At appears that mapping of these features dominates over
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 225

C
A

Figure 16–5. Quantitative characterization of receptive fields in human visual cortex


measured with subdural electrodes. (A) Average voltage response (black curves) and
99% confidence intervals (gray shaded regions) for each of the 36 visual field
locations tested for one electrode located on the lateral surface of the occipital cortex
near the occipital pole (red circle on schematic). Each panel shows the time period
from 0 to 200 milliseconds after stimulus onset. Stimulus duration was 200
milliseconds. (B) Root Mean Squared (RMS) voltage calculated for each position using
the time interval between 40 and 200 milliseconds after stimulus onset. One level of
interpolation has been used for presentation of the RMS data resulting in an 11 × 11
grid rather than a 6 × 6 grid. (C) The best fit two-dimensional Gaussian envelope to
the data shown in panel B. The black ellipse indicates the half maximum response
contour. (Reproduced with the permission of Oxford University Press, from Yoshor D,
Bosking WH, Ghose GM, Maunsell JH. Receptive fields in human visual cortex
mapped with surface electrodes. Cereb Cortex 2007;17(10):2293–2302.

accurate mapping of the visual field at later stages in with retinotopic mapping in human cortex (see Wandell
visual processing. The importance of understanding the et al. for a review).1 Each mapping method has relative
underlying fundamental maps in each area of cortex for strengths and weaknesses relating to properties such as
understanding data from fMRI experiments is reviewed the invasiveness of the procedure, the ability to use mul-
by Op de Beeck et al.54 tiple sets of stimuli, the spatial and temporal resolution
of the technique, and the type of signal that is being
measured.
䉴 MAPPING METHODS In this section, we begin with a discussion of
anatomical and structural imaging techniques that are
Technological advances have revolutionized the map- useful for localization of visual areas, briefly describe
ping of visual cortex by providing invaluable invasive functional neuroimaging methods useful for mapping
and noninvasive tools to infer localized brain function in human visual cortex, provide interesting examples from
vivo. For example, with the advent of modern mapping human research with healthy controls in the study of
methods, as many as 16 visual areas have been identified the visual system, and then discuss important issues to
226 SECTION II SYSTEMS

A B

Figure 16–6. Relating structural magnetic resonance imaging (MRI) to functional


visual cortex. (A) Coronal MRI of occipital lobe cortex. (B) Arrangement of V1, V2, and
V3 visual cortices, as well as connections between them. (Reproduced with the
permission of Elsevier, from Wichmann W, Müller-Forell W. Anatomy of the visual
system. Eur J Radiol 2004;49(1):8-30.)

consider when using the these methods to map the vi- to make contributions to our understanding of cortical
sual system in health and disease. function.56
Today, structural imaging methods play an impor-
tant role by providing precisely detailed neuroanatomy
STRUCTURAL IMAGING that can be co-localized with data from functional
studies. Systems such as Analysis of Functional Neu-
Before the era of functional neuroimaging, brain map- roimaging (AFNI) and other software suites allow for
ping researchers correlated behavioral impairments with functional data to be easily fused with anatomical data in
structurally defined lesions to characterize the relation- three-dimensional reconstructions of the brain surface.
ship between visual function and neuroanatomy. As de- Regardless of the functional imaging or recording tech-
scribed earlier, this method was used initially to help nique used, structural MRI and computed tomography
identify the location and organization of primary visual (CT) identified structures provide a common reference
cortex, and more recently it has been used to study frame to estimate neural activity. Structural MRI can now
localization of function within extrastriate areas. For ex- be used to provide detailed anatomical information with
ample, cerebral achromatopsia, a loss of ability to dis- a spatial resolution as small as 1 mm3 ,28 and at a min-
criminate color, was associated with MRI defined le- imum, can be used to identify typical sulci or gyri that
sions to occipital–temporal cortex,55,56 which suggested are located near particular visual areas. In some cases,
the existence of a color center in the human brain.57 high-resolution structural MRI has even been used to re-
Problematically, this method is often imprecise for map- veal differences in cytoarchitectonics, which can help
ping human visual cortex because even relatively focal identify particular visual areas. For example, Fig. 16–6
lesions routinely affect more than one functional area shows typical surface-based landmarks identifiable on
of the visual system. Also, studies that rely on lesions structural MRI for the locations of V1, V2, and V3 within
or inhibition of activity in a particular area can only cortical folding along the calcarine fissure.59 Talairach
be used to show the necessity of that area for a par- and Tournoux coordinates are used with neuroimaging
ticular function, and not sufficiency.58 Nonetheless, le- data to provide standard coordinate system based on
sion studies have made seminal historical contributions gross morphology.60 A limiting factor in combining data
to our understanding of the visual brain, and continue across multiple subjects is that brain topography varies
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 227

among individuals. To overcome this problem, unique


methods such as surface warping have been utilized to
make population averages of cortical areas based on
landmarks such as cortical folds.53,61,62
A relatively novel use of MRI, diffusion tensor imag-
ing (DTI), can be used to model the restricted movement
of water molecules diffusing within a given volume of
neural tissue.63 By subjecting tissue to multiple magnetic
field gradients, a tensor (or matrix) can be formed that
can estimate the amount of diffusion of water in any
arbitrary direction. Since the diffusion of water in neu-
ral tissue is direction-dependent, or anisotropic, being
greatest in the direction of white matter tracts, this tech-
nique can be used to predict the local trajectory of a
given white matter path found within a particular voxel.
By starting at one voxel in the imaged area and then
following the predicted trajectory from voxel to voxel, it
is possible to trace an entire white matter pathway from
one structure to another within the brain using this tech-
nique. One advantage of this methodology is that it can
be easily combined with both structural MRI and fMRI,
and allows for careful visualization of the white matter
tracts, which are not as well delineated using structural
MRI technology63−66 (see Fig. 16–7). This method has
been particularly useful in tracing the optic radiations
(see later), and is readily integrated into surgical naviga-
tion systems for guiding intraoperative dissection.

FUNCTIONAL NEUROIMAGING

Functional neuroimaging techniques are now standard


tools used to study visual cortex, both for research pur-
poses and to guide clinical decision-making. Functional
imaging of visual cortex in healthy subjects continues to
advance our understanding of the organization and func-
tion of various parts of the visual system. These imaging
techniques have been used to identify the location of,
and boundaries between, specialized areas of visual cor-
tex, and to study the specificity of the responses in those
areas. Data from healthy controls also provide a basis
to better understand the impact of clinical problems on Figure 16–7. Tractography and magnetic resonance
brain function. In this section, we review how advanced imaging. (Top) Bihemispheric reconstructions of the
imaging techniques, in particular fMRI, have been used optic nerve and tract (red) as well as of the optic
to map human visual areas in healthy subjects. Many of radiation (yellow ). (Bottom) Dissection of the optic
radiation into Meyer’s loop (yellow ), central bundle
these methods are readily applied to patients as well.
(green), and dorsal bundle (blue). R, right; L, left.
(From Hofer S, Karaus A, Frahm J. Reconstruction
Functional MRI and dissection of the entire human visual pathway
using diffusion tensor MRI. Front Neuroanat
fMRI allows localization of task-related neural activity by
2010;4:15.)
measurement of changes in blood flow and blood oxy-
genation levels (the blood oxygenation level dependent
(BOLD) signal or response).67 Because fMRI is noninva- One of the first uses of fMRI was to identify and
sive, has high spatial resolution, and can be used with differentiate the borders of multiple retinotopically orga-
many different tasks and visual stimuli in the same sub- nized areas with high-spatial precision (e.g., Fig. 16–4).
jects, it has become one of the dominant tools for study The stimuli used in these experiments are typically
of the visual system. small checkerboard or grating stimuli that elicit robust
228 SECTION II SYSTEMS

responses from sites in early visual cortex. By slowly hMT+ on the ventral bank of the occipital extent of the
moving these stimuli to different locations in the visual superior temporal sulcus that responds specifically to bi-
field, and analyzing the time course of the fMRI response ological motion (i.e., the motion of people and animals)
from each voxel, it is possible to reconstruct the region of compared to nonbiological motion.80
the visual field that best stimulates each location within a Perhaps the largest amount of fMRI research has
particular visual area. This procedure, known as “phase- focused on studying the object selectivity of the hu-
encoding,” is an extremely powerful technique since it man visual system.81,82 fMRI has been used to docu-
enables generation of high-resolution visual field maps ment a region within the LOC that responds selectively
simultaneously for multiple visual cortical areas in a rel- to objects.83,84 Ventral to the LOC exist two regions de-
atively short period of time.27−30 Before the advent of scribed by Kanwisher and colleagues, the fusiform face
fMRI and the development of phase-encoding stimu- area (FFA)81 and parahippocampal place area (PPA).85
lation and recording techniques, to create these maps These areas, as their names imply, respond specifically
would have required tens or hundreds of electrode pen- to identifying faces and places, respectively. The FFA is
etrations painstakingly acquired in animals, and simply consistently located at the fusiform gyrus and demon-
would not have been possible in humans. strates a strong foveal bias, while the PPA is closer to
Visual cortical areas contain maps of the visual field the parahippocampal gyrus and has a more peripheral
that are either mirror images of the world, or nonmirror visual field bias. More dorsally, there are also areas that
images. Reversals in the direction of mapping of visual respond to objects located around areas V3a and V7, but
space across the cortical surface, and the location of these areas are poorly understood. Grill-Spector et al.
changes from mirror image to nonmirror image repre- have shown with high-resolution imaging that a patchy
sentations can be used to establish the borders between organization exists in FFA such that both objects and
visual areas.29,30 By using fMRI, and varying both the faces are processed in this area, but that the informa-
nature of the task for the subject, and the type of visual tion may be averaged out only showing the most robust
stimuli used, retinotopic organization has been found “face” signals.57 Haxby et al.82 proposed a distributed
not only in early areas in the occipital cortex such as V1, neural network associated with the processing of faces
V2, and V3, but also areas in the lateral occipital cortex (see Fig. 16–8) that initially incorporates primary visual
(LOC) (LO-1, LO-2, hMT+), the ventral occipital cortex cortex, superior temporal sulcus, and fusiform gyrus, but
(hV4, VO-1, VO-2), dorsal occipital cortex (V3a, V3b), then incorporates areas such as amygdala, anterior tem-
and the posterior parietal cortex (IPS 0–4) (list and ter- poral lobe, and insula based on the requirements of the
minology from Wandell et al.).1 Interestingly, visual areas visual facial stimuli (e.g., processing an angry face incor-
appear to occur in clusters, with shared borders that rep- porates the amygdala).
resent one of the visual meridians, and confluent repre- Methodologically, although fMRI has good spatial
sentations of the fovea.68−70 The high-spatial resolution resolution, on the order of 1 mm, the temporal resolu-
of fMRI also allows researchers to study spatial organiza- tion is limited by changes in cerebral blood flow (CBF)
tion and functional specificity of processing within visual and is on the order of seconds. That is, although fMRI
areas. For example, while the measurement of orienta- data can be collected at 50–100 millisecond intervals, the
tion columns in humans is still slightly beyond the capa- slow changes in blood flow limit the conclusions that can
bility of fMRI, ocular dominance columns in V1, which be made about the temporal characteristics of functional
are approximately 500 microns in width, have been im- visual areas. Event-related fMRI has helped to resolve the
aged using 4T or 7T magnets.71,72 temporal resolution issue, but as with any method based
fMRI has been used extensively to examine how on blood flow or other measures of metabolic demand,
stimulus properties such as color, motion, and object fMRI will always be an indirect measurement of neuronal
category are represented in the visual cortex. In the function. Other advantages of fMRI include that it is non-
case of color, fMRI studies have suggested the existence invasive, does not require isotope injection (e.g., PET),
of several different specialized areas in an organized and affords better spatial resolution and localization than
stream beginning with primary visual cortex and extend- visual evoked potentials (VEPs) or PET. Furthermore, re-
ing through V4,73 V8,74 and LOC.75 For motion, fMRI peating testing with different tasks or stimuli is easily
has demonstrated the existence of an area (or complex possible in each subject.
of related areas) known as hMT+ that is an analogue to
monkey area V5/MT.76,77 Because of the excellent spatial
Positron Emission Tomography
resolution of fMRI, it has been possible to identify subre-
gions within hMT+ that are selective for different types A PET scanner quantifies regional CBF (rCBF) or
of motion, such as radial and circular motion.78 These metabolism during voluntary behavior. Clinically, PET
subdivisions may correspond to area MT and the other imaging is used to identify brain regions with re-
motion sensitive areas that surround MT in the monkey duced metabolism or decreased blood flow, which in
such as MST and FST.79 Grossman and colleagues have turn suggests functional impairment. Fluorine-18 fluo-
described an additional motion sensitive area anterior to rodeoxyglucose (FDG) is the most widely used PET
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 229

Figure 16–8. A model of the distributed human neural system for face perception.
This model consists of a core system, which consists of three regions of the visual
extrastriate cortex, and an extended system, which consists of components
important for other cognitive functions. Note bidirectional relationships of many of the
constituents. (Reproduced with the permission of Elsevier, from Haxby JV, Hoffman
EA, Gobbini MI. The distributed human neural system for face perception. Trends
Cogn Sci 2000;4(6):223–233.

tracer in a clinical setting to evaluate cortical glucose potentials (LFPs), that become synchronized in response
metabolism. Oxygen-15 (O-15) is a PET tracer used to to a given activity.87 However, the spatial resolution of
evaluate CBF that is more commonly used in experi- MEG has traditionally been poor, due to the nonunique-
mental settings because of its short half-life (2 minutes), ness of the inverse problem of trying to ascribe current
which makes it better suited for studies that require rapid dipoles to the magnetic fields measured. To overcome
and repeated presentation of visual stimuli.86 this problem, MEG can be combined with structural MRI
PET methodology has several strengths and weak- in a method called magnetic source imaging.
nesses when localizing functional areas of visual cortex. The main advantage of MEG over other methods
PET spatial resolution, while not as good as fMRI, is on is better temporal precision. Because of this property,
the order of several millimeters, especially with the ad- some have used it to attempt to unravel the temporal
vent of technology that makes it easier to co-localize sequence of neural activation in various brain areas dur-
PET scans with anatomic CT and structural MRI. As with ing visual perception. The waveform elicited by visual
fMRI, PET has poor temporal resolution, which is a ma- stimuli (i.e., the visual evoked magnetic field or VEF)
jor limitation because the visual system processes infor- is used to quantify the location and latencies required
mation quickly. Another limitation is that PET requires in processing the information. There has been consid-
radioactive marker substances to be injected into the erable work trying to assign particular VEF waveforms
subject’s bloodstream. Also like fMRI, PET provides an to certain areas of visual cortex.88−90 One problem with
indirect estimate of neuronal activity based on hemo- MEG is that it can incorrectly assign neuronal activity
dynamic parameters (CBF, blood volume, and oxygen when multiple areas of visual cortex are responding
content). As discussed later, other methods, like magne- simultaneously.91
toencephalography (MEG), VEP, and subdural grids ac-
tually measure neuronal electrical activity. Although PET
was the imaging technique first used to noninvasively in- Visual Evoked (or Event-Related)
vestigate the functional organization of visual cortex, it Potentials
has now largely been replaced by fMRI for this purpose.
VEPs are another noninvasive approach to mapping vi-
sual cortex. The method capitalizes on measuring whole
OTHER NONINVASIVE METHODS brain scalp waveform recordings of EEG data in re-
sponse to a time-locked visual event.92 By studying
Magnetoencephalography
normal controls, the location and latency of several spe-
MEG attempts to localize brain activity by recording mag- cific waveforms from visual cortical areas have been de-
netic fields generated by electrical activity, or local field termined. The timing of different peaks and valleys of
230 SECTION II SYSTEMS

waveforms are thought to provide a signature of the ployed standard clinical DCS techniques, both in the
hierarchical timing (as with MEG technology) of the pro- operating room, or extraoperatively using the standard
cessing of different visual stimuli. While this methodol- clinical electrodes implanted for monitoring of epilepsy
ogy has good temporal resolution, its spatial resolution patients (electrodes with a recording surface diameter
is highly limited. Recently, there has been an interest in of about 2.2 mm). The level of current employed was
trying to differentiate different visual pathways based on also in the standard milliampere range that is routinely
spectral analysis of the VEP waveforms.93 used for DCS mapping in the clinical arena. Several stud-
ies have reported that DCS of specific cortical areas can
inhibit specific visual perceptual abilities. For example,
Transcranial Magnetic Stimulation
Allison et al. (1994) and Mundel et al. (2003) reported
Transcranial magnetic stimulation (TMS) involves place- that DCS of the fusiform gyrus produces a temporary
ment of an electromagnetic coil on the scalp and then inability to name famous or familiar faces.34,101 Further-
high-intensity electrical current is rapidly turned on and more, Blancke et al. reported that DCS of area hMT+/V5,
off in the coil through the discharge of capacitors. The the visual area that responds selectively to visual mo-
goal is to provide focal magnetic stimulation to a specific tion, results in the bias of a patient’s perception of visual
brain region. TMS involves either stimulating local exci- motion.102
tation of neurons, leading to an overt behavior, or local DCS of visual cortex has also been shown to elicit vi-
suppression of neurons leading to cessation of an ongo- sual percepts. For example, Lee et al. reported inducing
ing behavior. Elicitation or suppression of behavior is de- (1) the percepts of geometric shapes, triangles, and dia-
pendent on stimulation parameters (frequency, intensity, monds with DCS to lateral occipital gyrus and fusiform
pulse duration, stimulation site). Effective interference of gyrus, (2) color perception with DCS to lingual gyrus,
stimulation of visual cortex by TMS is often confirmed fusiform gyrus, and interior occipital gyrus, and (3) the
when reaction time is slower when performing a visual perception of movement with DCS to the basal temporal-
task compared to reaction time on the task without stim- occipital junction, lateral posterior temporal cortex and
ulation. TMS of visual cortex has been used to induce occipital pole.103
phosphene perception,94,95 which subjects typically de- While traditionally DCS studies have reported only
scribe as a flash of gray or white light. TMS has also qualitative information about subject percepts follow-
been used to interfere with the visual perception of form ing stimulation, it is possible to design studies to pro-
by stimulation of striate96,97 and extrastriate cortex.98 It vide more quantitative information about the efficacy
has also been used to interfere with the perception of of stimulation at each site tested. Murphey et al. per-
motion99 and with visual memory100 in healthy subjects. formed a systematic study using DCS in subjects with
implanted subdural electrodes undergoing extraopera-
tive monitoring for epilepsy.104 In addition to noting
DIRECT CORTICAL STIMULATION any visual percepts elicited by DCS, they also used
two alternative forced choice psychophysical methods
Mapping with direct cortical stimulation (DCS) typically to quantify the level of current required for the sub-
involves the manually controlled delivery of suprathresh- jects to perform above chance in detecting the electrical
old electrical current to a discrete area of the brain. These stimulation. DCS of early visual areas, such as V1 and
procedures are performed either intraoperatively or from V2, was found to reliably elicit the perception of visual
chronically implanted subdural grid or strip electrodes phosphenes, while similar stimulation of later visual ar-
in patients undergoing extended exploration to better eas, even with higher currents, rarely elicited a verifiable
characterize a seizure focus. As with TMS, DCS involves visual response. However, in the rare examples in which
either the generation of an overt behavior or percept that a percept was elicited by DCS of a later visual area, per-
the patient can report, or the disruption of performance cepts were still generally reported to be phosphenes,
in some sort of task. DCS is considered a gold standard more complex than phosphenes generated in early vi-
for mapping cortical functions when used to stimulate sual areas, but not complex visual objects or scenes. In-
overt behavior. Unlike methods that assess subject per- terestingly, for sites where DCS did elicit visual percepts,
formance following a lesion or inhibition of a particular the amount of current necessary to elicit the percept
brain area, which only proves necessity, DCS stimulation did not vary significantly across the hierarchy of visual
of an overt behavior strongly suggests the sufficiency of cortex.
the stimulated region of cortex, or at least of a small net- Overall, DCS is useful for identifying early visual
work including that region of cortex, for the production cortex, but does not appear to reliably distinguish be-
of that behavior. tween different visual areas. It does remain a useful tech-
DCS with cortical surface electrodes has been ap- nique for studying visual cortex because, unlike other
plied to characterize visual function in the occipital and mapping methods, which only demonstrate correlations
temporal cortex of patients. These studies have em- between visual stimulation and cortical activity, DCS
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 231

stimulation can demonstrate a causal relationship be- cortex in particular, for generation of visual percepts. Ex-
tween brain activity and visual perception. Perhaps in periments using fMRI, subdural LFP recordings, and DCS
the future DCS will prove even more useful in dissecting in humans, as well as single-unit electrophysiology and
extrastriate visual cortex if it is employed while subjects’ lesion work in monkeys, are all in concordance on this
engaged in various controlled visually guided behaviors. general point. But, even for V1, there are discrepancies
when we move from asking about the basic response
properties, or the basic ability of this area of cortex to
LOCAL FIELD POTENTIAL support vision, to more complicated issues such as the
RECORDINGS FROM INTRACRANIAL ability of spatial attention or task demands to modu-
ELECTRODES late activity. For example, fMRI experiments have shown
large effects of changing spatial attention on the activity
LFPs evoked by visual stimulation are readily recorded measured in V1, while single-unit studies in monkeys,
with the subdural electrodes that are used clinically and LFP recordings from subdural electrodes in humans
to characterize epileptic foci in patients. A handful of have shown only very small effects.109
papers have studied LFP responses to map recep- These discrepancies are not surprising given the
tive fields using subdural electrodes of primary visual different type of signals measured by each technique,
cortex35 or to study later visual areas in response to faces and the different spatial and temporal resolution of
and objects.34,105 In a set of classic papers, McCarthy and each technique. It is the intersection of the spatial
associates106−108 were able to show robust event-related temporal pattern of activity across the cortex in re-
potentials at 200 milliseconds latency (N200) in human sponse to a particular stimulus, the type of signal being
ventral temporal cortex in response to face stimuli, and measured, and the spatial and temporal resolution of
were able show specificity and modulation of this signal. the specific technique being used to measure that sig-
In another study, Yoshor et al. were able to precisely nal, which results in a given measured “activity” pattern.
map spatial receptive fields in human visual cortex us- First, consider just the spatial resolution of various
ing small checkerboard stimuli presented while patients recording techniques. Imagine a stimulus that evokes
performed a letter detection task at the fixation point. a strong increase in the firing rate of neurons in some
They showed that receptive fields increased in size when columns of a cortical area of interest, and strong de-
moving from V1 to later visual areas in ventral occipi- creases in firing from other columns of cells in the same
tal cortex, and that there was a concomitant increase in area. In a single fMRI voxel, many thousands of cells
response latency, as would be expected for later visual would be activated in both directions, and the overall
areas.35 Furthermore, small but orderly changes in recep- change in activity might be quite small. This could lead
tive field location could be measured for successive con- to the incorrect conclusion that the stimulus did not ef-
tacts on the same subdural recording strips. These exper- fectively drive cells in that area at all. Recording from
iments suggest that with improvements in the size and single cells or from small groups of cells with electrodes
spacing of electrical contacts on recording grids or strips would reveal a much different story.
it should prove possible to perform detailed mapping in One strategy that has been employed by experi-
visual cortex of these subjects. menters to overcome limits in the spatial resolution of
Finally, recording of LFPs from subdural electrodes fMRI relative to the scale of the local architecture in vi-
has also been used to study the degree to which spatial sual cortex is to use visual adaptation. Visual adaptation
attention can cause modulation of visual responses in refers to the phenomenon wherein repeated presenta-
early visual cortex.109 This is an example of how record- tions of a preferred visual stimulus typically result in a
ing of LFPs from these electrodes can be used as a bridge diminished neural response in visual cortex. Functional
between single-unit electrophysiological studies in mon- imaging studies can take advantage of this property by
keys and fMRI experiments in humans, which had pro- testing the relative effectiveness of several stimuli after
duced conflicting results on this issue in the past. a period of adaptation. If following this adaptation pe-
Thus, although there are a limited number of oppor- riod it can be shown that a particular cortical area now
tunities to record from patients who have electrodes im- shows reduced responses to the adapting stimulus, but
planted over visual cortex, these electrodes can provide does not show reduced responses to another stimulus,
extremely valuable information when they are available. this implies that the area contains cells that are selective
for the first stimulus (a preferred stimulus). This is a use-
ful method for demonstrating visual selectivity that does
CONCORDANCE ACROSS METHODS not require spatial resolution at the scale at which the
preferred stimulus property is encoded. This technique
There is now a huge amount of information from many has recently been used to examine three-dimensional
sources about the general importance of the occipital motion selectivity in human MT110 and orientation se-
cortex for vision, and of the importance of primary visual lectivity in human V1.111
232 SECTION II SYSTEMS

Next, consider some of what we know about the can be carried out without significant functional conse-
signal that is measured by each recording technique. quence. In contrast, for early visual cortex and V1 in
As outlined by a recent review,112 many neuronal cir- particular, resection of the cortex or its projections re-
cuits consist of both excitatory and inhibitory inputs, sults in obvious visual deficits.
feed-forward and feedback systems, and finally output. Similarly, the optic radiations running from the lat-
Though a great oversimplification, one can think of the eral geniculate nuclei to primary visual cortex are also
BOLD response as an overall measure of changing ex- a place where damage can have profound effects on
citatory and inhibitory neuronal inputs and perhaps as- patient outcome. It is well known that lesions may com-
trocytic function,113 while electrophysiological recording promise visual tracts en route to visual cortex and result
of spiking is a direct measurement of neuronal output. in characteristic visual field defects. However, an impor-
LFPs are also a measurement of overall neuronal (and tant clinical application is the avoidance of these path-
perhaps astrocytic) response. Thus, it is not surprising ways in the attempt to resect lesions in the temporal
that there seems to be more of an agreement between lobe or deep within cortex such as in the atrium of the
fMRI and LFPs compared to fMRI and single or multiunit lateral ventricles.114,115 The relative anatomic position of
spike recordings.67 these tracts should be considered in planning surgical
trajectories, such as in temporal lobectomy surgery or
approaches to deep tumors.116 These tracts may be iden-
䉴 CLINICAL APPLICATIONS tified with DTI; the DTI images can be integrated into a
surgical navigation system used at surgery, to help the
Mapping methods predominantly used in neuroscience surgeon avoid damaging these fibers.114
to study the visual system may also play a role in the di-
agnosis and treatment of patients. The efficacy of func-
tional mapping methods in a clinical setting lies in the FUNCTIONAL MRI
ability of these techniques to provide useful information
to spare eloquent areas of visual cortex or optic radia- Several studies have validated fMRI techniques in a
tions, identify the effects of tumors or other structural ab- clinical setting by comparing the results of visual acti-
normalities on vision, or to characterize areas that have vation patterns based on fMRI with those of perimet-
reorganized. In this section, we discuss the application ric evaluation (the gold standard for identifying sco-
of clinical mapping methods in neurological and neuro- tomas) in patients with visual field defects.117,118 The
surgical settings. results of these studies demonstrate high consistency be-
Before reviewing the use of different modalities for tween decreased fMRI activation with visual paradigms
mapping visual areas as an adjunct to clinical care and and visual field defects measured using perimetric eval-
in particular for guiding neurosurgical resections, it is uation. For example, Kollias et al. (1998) used fMRI
important to recognize the functional redundancy, and (with a binocular repetitive photic stimulation task) in
more importantly the bilateral representations, of later the presurgical planning of 10 patients with space-
visual areas that may render patients relatively resistant occupying lesions involving the posterior afferent visual
to adverse effects with unilateral resections. Early vi- system.119 Results showed concordance for nine of the
sual areas, such as V1, are highly retinotopic and are ten patients between asymmetric response (e.g., an ab-
composed of neurons with small spatial receptive fields. sence or minimal fMRI activation) in visual cortex corre-
When a portion of V1 is lesioned or resected, a corre- sponding to the visual field deficits (e.g., homonymous
sponding contralateral visual field deficit or scotoma will hemianopsias).
invariably result. But later visual areas encode a far less Functional imaging has also been used to guide
veridical representation of the visual world and typically surgery in patients with space-occupying lesions to
contain visually responsive neurons with large, even bi- help spare eloquent visual areas.120 For example, Fried
lateral, receptive fields. So, it is not surprising that an et al.120 used fMRI (with a binocular photic stimulation
isolated unilateral surgical resection or stroke involving of 8.3-Hz flashing red lights task) to map visual cortex in
a later visual area such as hMT or LOC does not result a patient who had intractable seizures originating near a
in a readily apparent visual deficit that alters a patient’s temporo-occipital cyst. By using this fMRI to guide a sub-
quality of life. Bilateral lesions of a later visual area, in sequent surgical resection, they were able to resect the
contrast, are likely to result in a functional deficit. Fortu- seizure focus and leave the patient with only a limited
nately, bilateral homotopic surgical resections are almost visual hemifield defect. Similarly, Roux et al.117 reported
never a clinical consideration anyway. In a practical clin- how fMRI was helpful in minimizing postoperative vi-
ical sense, this means that while mapping studies reveal sual field defects in five patients who underwent occip-
large swaths of cortex that are visually responsive, for ital cortex tumor removal. These authors noted, how-
many of these areas, particularly those that fall late in ever, a very important limitation of fMRI data when de-
the hierarchy of visual processing, unilateral resections termining the extent of a resection, which is an absence
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 233

of functional activity does not always imply that neural In an interesting study of patients without OLE,
tissue is not capable of being functional. For example, Wong et al.127 used FDG-PET to study occipital lobe
they presented data on a patient with a right homony- hypometabolism in seven patients who had visual
mous hemianopsia who had no functional activation in field cuts and four patients without field defects post-
the left lingual gyrus (corresponding to the location of a temporal lobectomy. Imaging results indicated occipi-
grade III astrocytoma). The functional importance of the tal lobe hypometabolism corresponding to visual field
area around the tumor could not be discerned because defects in the seven of the patients with defects, and
the visual defect could have resulted from mass effect hypometabolism was found in one of the patients with-
that led to the absence of functional activation, or may out a field defect. The hypometabolism was proposed to
have corresponded to tumor.117 occur due to deafferentation after interruption of optic
DTI, or MR tractography, has also proved useful in pathways because of the temporal lobectomy.127
helping to preserve visual function during neurosurgi- With its ability to measure receptor distributions,
cal procedures. This is particularly true for resections PET may find future use in delineating subtle changes
of tumors or epileptic foci that may neighbor or in- in visual pathways or maps in normal and neuropsycho-
vade the extensive visual pathways from retina to pri- logical diseases. For example, Gerstl et al. (2008) were
mary visual cortex. In fact, there has been a resurgence able to differentiate between V1 and V2 in early human
of interest in studying these tracts using cadavers121,122 cortex based on 5-HT1A receptor distribution and this
and subsequent suggestions for new methods to seems to be corroborated in postmortem studies.129,130
resect lesions that may neighbor these radiations. Pow-
ell et al. (2005) showed that for two patients undergoing
MAGNETOENCEPHALOGRAPHY
anterior temporal lobe resections, one had the stereo-
typical contralateral homonymous hemianopsia and the
MEG is useful in localizing eloquent cortex for surgi-
other patient did not. Comparing pre- and postoper-
cal planning in patients with brain tumors or intractable
ative DTIs, they showed that in the patient who had
epilepsy.131 A distinct advantage over fMRI and PET in
a visual field defect, the optic radiations were in fact
a presurgical epilepsy setting is that MEG may be able
traversed.123 Taking this approach further, Nimsky et al.
to capture ictal spread to better identify seizure foci.132
(2006) showed how DTI can be integrated into stan-
Several studies have also demonstrated the efficacy of
dard intraoperative MRI navigation to map out the optic
MEG in presurgical planning for tailored resection of tu-
radiations.64
mors in visual cortex.133,134 For example, Grover et al.
Interestingly, Govindan et al. (2008) using DTI,
(2006) reported on the efficacy of MEG in helping to
measured water diffusion-related changes in the optic
minimize visual field defects after resection of tumors in
radiations ipsilateral and contralateral to occipital lobe
temporo-parieto-occipital areas.133
resections in ten children who had undergone surgery to
In a unique clinical application, MEG has been
treat intractable epilepsy.124 They validated the method
used to study the cortical reorganization of visual cortex.
by showing a strong relationship between optic radia-
Understanding unexpected change in visual cortex is
tion tract damage and visual field losses. Their data also
important to help characterize irregular localization of
found that contralateral optic radiations underwent sig-
function, reflecting reorganized or displaced cortex. For
nificant changes in anisotropy after resection, suggesting
example, Burneo et al. (2004) demonstrated that visual
that plasticity changes occur elsewhere to compensate
functions are more likely to be reorganized with cortical
for ipsilateral damage.
dysplasia but not polymicrogyria.135 Inoue et al. (2004)
combined three-dimensional anisotropy contrast (e.g.,
anisotropic DWI that shows isotropic water motion) and
POSITRON EMISSION TOMOGRAPHY
MEG VEFs to identify optic radiations and primary vi-
sual cortex in a 26-year-old patient with a tumor along
PET is used for brain mapping in a clinical setting for
the right calcarine fissure.134 They demonstrated that the
many different types of purposes. For example, it is used
techniques could be combined to tailor a resection to
as tool in the diagnosis of Alzheimer’s disease,125 and to
avoid postoperative visual impairments.
differentiate radiation necrosis from tumor recurrence in
patients who had already undergone radiation therapy
to treat the tumor.126 For clinical brain mapping of visual EVENT-RELATED SCALP VISUAL
areas, the largest area of research comes from the presur- EVOKED POTENTIALS
gical evaluation of individuals with epilepsy. FDG-PET
is an integral component of the presurgical evaluation Analogous to motor-evoked potentials and somato-
because it can aid in the confirmation of an epileptic sensory-evoked potentials used in cranial and spinal
seizure focus in patients with medically intractable oc- cases, VEPs can be used intraoperatively to monitor the
cipital lobe epilepsy (OLE).127,128 robustness of the visual pathways.136 Unfortunately, as
234 SECTION II SYSTEMS

VEPs have poor localization, VEPs do not give a priori DIRECT CORTICAL STIMULATION
information to guide resection or procedure, and often MAPPING
times, it is only after damaging a visual tract that one
sees a difference in VEPs. DCS has a long history in clinical neurosurgery, begin-
Often clinical practice provides evidence for ex- ning with the works of Penfield who showed that he
perimental hypotheses. For example, Tobimatsu and could elicit phosphenes during electrical stimulation of
Celesia137 illustrate the usefulness of VEP in an interest- early visual areas.145 As our understanding of the intri-
ing case presentation of a 62-year-old patient who suf- cacies of the visual cortex has blossomed over the last
fered from carbon monoxide poisoning who then pre- decade and technology has similarily progressed, intra-
sented with visual agnosia. His structural MRI indicated operative and extraoperative DCS for both research and
severe damage to parieto-occipital cortex bilaterally with clinical uses has seen a resurgence. By using extraopera-
no damage to V1. His VEPs to simple checkerboards tive DCS, Lee et al. showed that stimulation of early areas
also suggested intact V1. However, when presented with of the visual cortex mostly resulted in patient’s describ-
face stimuli, the patient had no early responses (which ing simple forms and stimulation of more distal areas re-
he should have had, as predicted by normal VEPs to sulted in more complex forms, laying credence to a hier-
checkerboards). The authors interpreted this finding as archical organization of visual processing.103 Our group
providing support for the presence of a feedback sys- has shown that the probability of eliciting precepts in the
tem from extrastriate cortices to V1, which suppressed visual cortex is indirectly related to the distance from the
the normally intact V1 response in the patient. occipital pole, with thresholds for these precepts being
In another interesting study, Kamada et al. (2005) directly related to this distance.104 Nguyen et al. recently
combined real-time VEPs and optic radiation DTI trac- reported a single case in which they used DCS and fre-
tography to monitor visual pathways intraoperatively. quent monitoring of a patient’s visual fields during an
They were able to spare functional visual areas in one awake craniotomy to resect an occipital glioma, while
of the patients reported and showed how loss of VEPs preserving the patient’s visual field.146 Subcortical stim-
occurred in a patient that had a subsequent visual field ulation can similarly be implemented to help define the
defect138 (see Fig. 16–9). maximum depth of resection.147

TRANSCRANIAL MAGNETIC
䉴 PEARLS AND PITFALLS
STIMULATION
In this chapter, we have provided an overview of the
Repetitive TMS (rTMS) has been used clinically to treat
clinical utility of mapping methods. The body of liter-
migraine, stroke, and Parkinson’s disease, as well as de-
ature that covers these topics is vast and complex and
pression (see Fitzgerald et al.139 for a review). There has
must be considered and applied carefully in clinical (and
been little use of TMS in planning surgical resections in
research) settings.
the visual cortex, or to avoid optic radiations, but TMS
Despite advances in neuroimaging and neurophys-
has shown promise in mapping motor areas140 and has
iology techniques over the past three decades that have
potential in unraveling visuospatial judgements and as-
helped in identifying structural lesions and functional
sociated defects.141,142
impairment, serious limitations exist with each method.
Some of these limitations are technological in nature,
LOCAL FIELD POTENTIAL such as the spatial resolution of fMRI, but many of them
RECORDINGS FROM INTRACRANIAL are inherent to the different techniques, such as the fact
ELECTRODES that one is only indirectly measuring neuronal activ-
ity with fMRI. While technologies such as fMRI contiue
There are few reports of using LFPs evoked by visual to improve, it is necessary for the clinician to under-
stimulation to tailor surgical resections to minimize visual stand inherent limitations in order to use and interpret
deficits.143,144 Curatolo et al.144 used a flashing strobe data effectively. Furthermore, it is in the combination
light to elicit LFPs in two patients with OLE being eval- of these technologies and their overlapping strengths
uated for surgery. They found that the use of photopic that the most comprehensive picture and understand-
stimulation to map visual cortex helped to preserve cen- ing of visual information processing in the brain will be
tral vision after occipital lobe surgery.144 While our group unraveled.
has primarily used retinotopic mapping of visual cortex Several clinical realities are particularly important
as a research tool,35 we have also found it useful for and are relevant even with the most judicious combina-
defining the location of early visual cortex and in tai- tion of various structural and functional mapping tech-
loring a resection with maximal preservation of visual niques. The first reality is the fact that many patients have
function. underlying pathological disease that may affect normal
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 235

C B

Figure 16–9. Combining visual evoked potentials (VEPs), tractography, and


navigation. (A) Consecutive changes in the VEPs during tumor resection. VEPs
became undetectable when the optic radiation was damaged at 13:12 (black arrow ).
(B) Two-dimensional magnetic resonance imaging (MRI) scans on neuronavigation
demonstrating that resection had reached the ventricular wall, where the optic
radiation was embedded. (C) Three-dimensionally reconstructed whole-head MRI
data on neuronavigation showing the optic radiation (purple), the tumor (yellow ), and
the navigation probe (blue bar ). (Reproduced with the permission of Wolters Kluwer
Inc., Kamada K, Todo T, Morita A, et al. Functional monitoring for visual pathway using
real-time visual evoked potentials and optic-radiation tractography. Neurosurgery
2005;57(1 Suppl):121–127; discussion 121–127.

structure/function relationships. Lesions, whether they gioma) may displace cortex and white matter tracts. The
are tumors, vascular or genetic in nature, may cause clinical management of these two situations is potentially
reorganization of visual areas, or affect the neuronal different and extent of resection, with known effects on
pathways leading to visual cortex.119,122 These poten- patient survival, must be weighed against neurological
tial changes must be taken into consideration by the deficit.
clinician. For example, there is evidence that low-grade Another clinical reality that is not addressed by com-
gliomas may have functional cortex and white matter binatorial use of preoperative structural and functional
tracts within them,130 while other lesions (e.g., menin- mapping techniques is intraoperative shifts in brain
236 SECTION II SYSTEMS

tissue. This should be helped with the advent 11. Lister WT, Holmes G. Disturbances of Vision from Cere-
of intraoperative structural and functional mapping bral Lesions, with Special Reference to the Cortical Rep-
techniques.122,131 resentation of the Macula. Proc R Soc Med 1916;9(Sect
Finally, it is important to recognize that unilateral Ophthalmol):57-96.
resection of later visual areas may be well tolerated due 12. Horton JC, Hoyt WF. The representation of the visual field
in human striate cortex. A revision of the classic Holmes
to the additional presence of these functional areas on
map. Arch Ophthalmol 1991;109(6):816-824.
the contralateral hemisphere. For example, unilateral re- 13. Daniel PM, Whitteridge D. The representation of the vi-
section of area IT, which is critical for object processing, sual field on the cerebral cortex in monkeys. J Physiol
does not result in overt deficits in object recognition, 1961;159:203-221.
presumably because the contralateral area IT in itself 14. Talbot SA, M WH. Physiological studies on neural mecha-
provides sufficient function. But resection of an early nisms of visual localization and discrimination. Am J Oph-
retinotopic visual area on one side, such as a portion of thal 1941;24:1255-1264.
V1, will typically result in a permanent visual field deficit 15. Hubel DH, Wiesel TN. Receptive fields, binocular interac-
because this particular spatial representation is not also tion and functional architecture in the cat’s visual cortex.
present in the contralateral hemisphere. J Physiol 1962;160:106-154.
As technology advances, different methods of vi- 16. Mountcastle VB. Modality and topographic properties of
single neurons of cat’s somatic sensory cortex. J Neuro-
sual mapping will provide a greater understanding of
physiol 1957;20(4):408-434.
this exciting and complicated field. Similarly, as basic re- 17. Hubel DH, Wiesel TN. Ferrier lecture. Functional architec-
search progresses, the clinician will have a broad span ture of macaque monkey visual cortex. Proc R Soc Lond
of literature on which to base clinical decisions and re- B Biol Sci 1977;198(1130):1-59.
search. But the road connecting technology and basic 18. Penfield W, Rasmussen T. The cerebral cortex of man;
research with clinical endeavors should by no means a clinical study of localization of function. New York:
be a one-way street. Rather, clinicians, with their ability Macmillan, 1950, p. xv, 248.
to probe different questions directly in human patients, 19. Brindley GS, Lewin WS. The sensations produced by elec-
whether it be subdural electrodes for epileptic seizure trical stimulation of the visual cortex. J Physiol 1968;
location or DCS, can play a major role in shaping our 196(2):479-493.
understanding of visual information processing in the 20. Dobelle WH, Mladejovsky MG. Phosphenes produced by
electrical stimulation of human occipital cortex, and their
brain.
application to the development of a prosthesis for the
blind. J Physiol 1974;243(2):553-576.
21. Knierim JJ, Van Essen DC. Visual cortex: cartography, con-
REFERENCES nectivity, and concurrent processing. Curr Opin Neurobiol
1992;2(2):150-155.
1. Wandell BA, Dumoulin SO, Brewer AA, Visual field maps 22. Merigan WH, Maunsell JH. How parallel are the primate
in human cortex. Neuron 2007;56(2):366-383. visual pathways? Annu Rev Neurosci 1993;16:369-402.
2. Maeda K, et al. Topography of the secondary somatosen- 23. Ungerleider LG, Haxby JV. ‘What’ and ‘where’ in the hu-
sory cortex in humans: a magnetoencephalo-graphic man brain. Curr Opin Neurobiol 1994;4(2):157-165.
study. Neuroreport 1999;10(2):301-306. 24. Fox PT, et al. Retinotopic organization of human vi-
3. Toni I, Thoenissen D, Zilles K. Movement preparation and sual cortex mapped with positron-emission tomography.
motor intention. Neuroimage 2001;14(1 Pt 2):S110-S117. J Neurosci 1987;7(3):913-922.
4. Glickstein M, Rizzolatti G. Francesco Gennari and the 25. Fox PT, et al. Mapping human visual cortex with positron
structure of the cerebral cortex. Trends Neurosci 1984; emission tomography. Nature 1986;323(6091):806-809.
7(12):464-467. 26. Schwartz EL, Christman DR, Wolf AP. Human primary vi-
5. Gross CG. Brain, vision, memory : tales in the history of sual cortex topography imaged via positron tomography.
neuroscience. Cambridge, Mass: MIT Press, 1998, p. xviii, Brain Res 1984;294(2):225-230.
255. 27. DeYoe EA, et al. Mapping striate and extrastriate visual
6. Finger S. Origins of neuroscience : a history of explo- areas in human cerebral cortex. Proc Natl Acad Sci U S A
rations into brain function. New York: Oxford University 1996;93(6):2382-2386.
Press, 1994, p. xviii, 462. 28. Engel SA, Glover GH, Wandell BA. Retinotopic organiza-
7. Henschen SE. On the visual path and centre. Brain tion in human visual cortex and the spatial precision of
1893;16(1-2):170-180. functional MRI. Cereb Cortex 1997;7(2):181-192.
8. Fishman RS. Gordon Holmes, the cortical retina, and the 29. Engel SA, et al. fMRI of human visual cortex. Nature
wounds of war. The seventh Charles B. Snyder Lecture. 1994;369(6481):525.
Doc Ophthalmol 1997;93(1-2):9-28. 30. Sereno MI, et al. Borders of multiple visual areas in hu-
9. Glickstein M, Whitteridge D. Tatsuji Inouye and the map- mans revealed by functional magnetic resonance imaging.
ping of the visual fields on the human cerebral cortex. Science 1995;268(5212):889-893.
Trends Neurosci 1987;10(9):350-353. 31. Brewer AA, et al. Visual areas in macaque cortex mea-
10. Holmes G. Disturbances of Vision by Cerebral Lesions. Br sured using functional magnetic resonance imaging.
J Ophthalmol 1918;2(7):353-384. J Neurosci 2002;22(23):10416-10426.
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 237

32. Tootell RB, et al. New images from human visual cortex. ing fMRI data: maps, modules and dimensions. Nat Rev
Trends Neurosci 1996;19(11):481-9. Neurosci 2008;9(2):123-135.
33. Tootell RB, Tsao D, Vanduffel W. Neuroimaging weighs 55. Pearlman AL, Birch J, Meadows JC. Cerebral color blind-
in: humans meet macaques in “primate” visual cortex. ness: an acquired defect in hue discrimination. Ann Neu-
J Neurosci 2003;23(10):3981-3989. rol 1979;5(3):253-261.
34. Allison T, et al. Human extrastriate visual cortex and the 56. Damasio A, et al. Central achromatopsia: behavioral,
perception of faces, words, numbers, and colors. Cereb anatomic, and physiologic aspects. Neurology 1980;
Cortex 1994;4(5):544-554. 30(10):1064-1071.
35. Yoshor D, et al. Receptive fields in human visual 57. Grill-Spector K, Sayres R, Ress D. High-resolution imaging
cortex mapped with surface electrodes. Cereb Cortex reveals highly selective nonface clusters in the fusiform
2007;17(10):2293-2302. face area. Nat Neurosci 2006;9(9):1177-1185.
36. Morin LP, Allen CN. The circadian visual system, 2005. 58. Tharin S, Golby A. Functional brain mapping and
Brain Res Rev 2006;51(1):1-60. its applications to neurosurgery. Neurosurgery 2007;60
37. Amini A, Digre K, Couldwell WT. Photophobia in a blind (4 Suppl 2):185–201; discussion 201-202.
patient: An alternate visual pathway. Case report. J Neu- 59. Wichmann W, Muller-Forell W. Anatomy of the visual sys-
rosurg 2006;105(5):765-768. tem. Eur J Radiol 2004;49(1):8-30.
38. Orban GA, Van Essen D. Vanduffel W. Comparative map- 60. Talairach J, Tournoux P. Co-planar stereotaxic atlas of
ping of higher visual areas in monkeys and humans. the human brain : 3-dimensional proportional system;
Trends Cogn Sci 2004;8(7):315-324. an approach to medical cerebral imaging. Stuttgart; New
39. Gazzaniga MS, Ivry RB, Mangun GR. Cognitive neuro- YorkNew York: G. Thieme; Thieme Medical Publishers,
science: the biology of the mind. 2nd ed. New York: Nor- 1988, p. viii, 122.
ton. 1 v, 2002 (various pagings). 61. Hinds OP, et al. Accurate prediction of V1 location from
40. Sparks D, Rohrer WH, Zhang Y. The role of the superior cortical folds in a surface coordinate system. Neuroimage
colliculus in saccade initiation: a study of express sac- 2008;39(4):1585-1599.
cades and the gap effect. Vision Res 2000;40(20):2763- 62. Van Essen DC. A Population-Average, Landmark- and
2777. Surface-based (PALS) atlas of human cerebral cortex.
41. Celesia GG, Brigell MG. Cortical visual processing. Elec- Neuroimage 2005;28(3):635-662.
troencephalogr Clin Neurophysiol Suppl 1999;50:202-209. 63. Hofer S, Karaus A, Frahm J. Reconstruction and dissection
42. Casagrande VA. A third parallel visual pathway to primate of the entire human visual pathway using diffusion tensor
area V1. Trends Neurosci 1994;17(7):305-310. MRI. Front Neuroanat 2010;4:15.
43. Hendry SH, Reid RC. The koniocellular pathway in pri- 64. Nimsky C, et al. Intraoperative visualization for resec-
mate vision. Annu Rev Neurosci 2000;23:127-153. tion of gliomas: the role of functional neuronavigation
44. Van Essen DC, Drury HA, Structural and functional analy- and intraoperative 1.5 T MRI. Neurol Res 2006;28(5):
ses of human cerebral cortex using a surface-based atlas. 482-487.
J Neurosci 1997;17(18):7079-7102. 65. Nimsky C, et al. Intraoperative high-field MRI: anatomical
45. Grill-Spector K, Malach R. The human visual cortex. Annu and functional imaging. Acta Neurochir Suppl 2006;98:
Rev Neurosci 2004;27:649-677. 87-95.
46. Felleman DJ, Van Essen DC. Distributed hierarchical pro- 66. Nimsky C, et al. Updating navigation with intraoperative
cessing in the primate cerebral cortex. Cereb Cortex image data. Top Magn Reson Imaging 2009;19(4):197-204.
1991;1(1):1-47. 67. Logothetis NK, et al. Neurophysiological investigation of
47. Ozdemir A, Black PM. Mapping of Human Visual Cortex. the basis of the fMRI signal. Nature 2001;412(6843):150-
Neurosurgery Quarterly 2005;15(2):65-71. 157.
48. Livingstone MS, Hubel DH. Specificity of intrinsic con- 68. Kolster H, et al. Visual field map clusters in macaque ex-
nections in primate primary visual cortex. J Neurosci trastriate visual cortex. J Neurosci 2009;29(21):7031-9.
1984;4(11):2830-2835. 69. Schira MM, et al. The foveal confluence in human visual
49. Konen CS, Kastner S. Two hierarchically organized neural cortex. J Neurosci 2009;29(28):9050-8.
systems for object information in human visual cortex. Nat 70. Wandell BA, Brewer AA, Dougherty RF. Visual field map
Neurosci 2008;11(2):224-231. clusters in human cortex. Philos Trans R Soc Lond B Biol
50. Sawatari A, Callaway EM. Convergence of magno- and Sci 2005;360(1456):693-707.
parvocellular pathways in layer 4B of macaque primary 71. Yacoub E, et al. Robust detection of ocular dominance
visual cortex. Nature 1996;380(6573):442-446. columns in humans using Hahn Spin Echo BOLD func-
51. Yabuta NH, Sawatari A, Callaway EM. Two functional tional MRI at 7 Tesla. Neuroimage 2007;37(4):1161-1177.
channels from primary visual cortex to dorsal visual cor- 72. Menon RS, et al. Ocular dominance in human V1 demon-
tical areas. Science 2001;292(5515):297-300. strated by functional magnetic resonance imaging. J Neu-
52. Sincich LC, Horton JC. The circuitry of V1 and V2: inte- rophysiol 1997;77(5):2780-2787.
gration of color, form, and motion. Annu Rev Neurosci 73. Bartels A, Zeki S. The architecture of the colour centre in
2005;28:303-26. the human visual brain: new results and a review. Eur J
53. Van Essen DC, et al. Functional and structural mapping of Neurosci 2000;12(1):172-193.
human cerebral cortex: solutions are in the surfaces. Adv 74. Hadjikhani N, et al. Retinotopy and color sensitivity in
Neurol 2000;84:23-34. human visual cortical area V8. Nat Neurosci 1998;1(3):235-
54. Op de Beeck HP, Haushofer J, Kanwisher NG. Interpret- 241.
238 SECTION II SYSTEMS

75. Wandell BA. Computational neuroimaging of human vi- 96. Kammer T, et al. Transcranial magnetic stimulation in the
sual cortex. Annu Rev Neurosci 1999;22:145-173. visual system. I. The psychophysics of visual suppression.
76. Ashida H, et al. FMRI adaptation reveals separate mech- Exp Brain Res 2005;160(1):118-128.
anisms for first-order and second-order motion. J Neuro- 97. Romei V, et al. Occipital transcranial magnetic stimula-
physiol 2007;97(2):1319-1325. tion has opposing effects on visual and auditory stimu-
77. Tootell RB, et al. Visual motion aftereffect in human cor- lus detection: implications for multisensory interactions.
tical area MT revealed by functional magnetic resonance J Neurosci 2007;27(43):11465-11472.
imaging. Nature 1995;375(6527):139-141. 98. Epstein CM, Verson R, Zangaladze A, Magnetic coil sup-
78. Morrone MC, et al. A cortical area that responds specif- pression of visual perception at an extracalcarine site.
ically to optic flow, revealed by fMRI. Nat Neurosci J Clin Neurophysiol 1996;13(3):247-252.
2000;3(12):1322-1328. 99. Hotson J, et al. Transcranial magnetic stimulation of ex-
79. Huk AC, Dougherty RF, Heeger DJ. Retinotopy and func- trastriate cortex degrades human motion direction dis-
tional subdivision of human areas MT and MST. J Neurosci crimination. Vision Res 1994;34(16):2115-2123.
2002;22(16):7195-7205. 100. Beckers G, Homberg V. Cerebral visual motion blind-
80. Grossman ED, Blake R. Brain Areas Active during Visual ness: transitory akinetopsia induced by transcranial mag-
Perception of Biological Motion. Neuron 2002;35(6):1167- netic stimulation of human area V5. Proc Biol Sci
1175. 1992;249(1325):173-178.
81. Kanwisher N, McDermott J, Chun MM. The fusiform face 101. Mundel T, et al. Transient inability to distinguish be-
area: a module in human extrastriate cortex specialized tween faces: electrophysiologic studies. J Clin Neurophys-
for face perception. J Neurosci 1997;17(11):4302-4311. iol 2003;20(2):102-110.
82. Haxby JV, Hoffman EA, Gobbini MI. The distributed hu- 102. Blanke O, et al. Direction-specific motion blindness in-
man neural system for face perception. Trends Cogn Sci duced by focal stimulation of human extrastriate cortex.
2000;4(6):223-233. Eur J Neurosci 2002;15(12):2043-2048.
83. Grill-Spector K, Kourtzi Z, Kanwisher N. The lateral oc- 103. Lee HW, et al. Mapping of functional organization in hu-
cipital complex and its role in object recognition. Vision man visual cortex: electrical cortical stimulation. Neurol-
Res 2001;41(10-11):1409-1422. ogy 2000;54(4):849-854.
84. Kanwisher N, et al. Functional imaging of human visual 104. Murphey DK, et al. Perceiving electrical stimulation of
recognition. Brain Res Cogn Brain Res 1996;5(1-2):55-67. identified human visual areas. Proc Natl Acad Sci U S A
85. Epstein R, Kanwisher N. A cortical representation of the 2009;106(13):5389-5393.
local visual environment. Nature 1998;392(6676):598-601. 105. Allison T, et al. Electrophysiological studies of color pro-
86. Watson JD. The human visual system, in Brain mapping cessing in human visual cortex. Electroencephalogr Clin
the systems, Toga AW, Mazziotta JC. editors. Orlando: Aca- Neurophysiol 1993;88(5):343-355.
demic Press, 2000, p. 263-289. 106. Puce A, Allison T, McCarthy G. Electrophysiological
87. Hamalainen MS. Magnetoencephalography: a tool for studies of human face perception. III: Effects of top-
functional brain imaging. Brain Topogr 1992;5(2): down processing on face-specific potentials. Cereb Cortex
95-102. 1999;9(5):445-458.
88. Ahlfors SP, Ilmoniemi RJ, Hamalainen MS. Estimates of 107. McCarthy G, et al. Electrophysiological studies of human
visually evoked cortical currents. Electroencephalogr Clin face perception. II: Response properties of face-specific
Neurophysiol 1992;82(3):225-236. potentials generated in occipitotemporal cortex. Cereb
89. Gramfort A, et al. Challenging the estimation of corti- Cortex 1999;9(5):431-444.
cal activity from MEG with simulated fMRI-constrained 108. Allison T, et al. Electrophysiological studies of human
retinotopic maps. Conf Proc IEEE Eng Med Biol Soc face perception. I: Potentials generated in occipitotem-
2007;2007:4945-4948. poral cortex by face and non-face stimuli. Cereb Cortex
90. Huang MX, et al. Vector-based spatial-temporal minimum 1999;9(5):415-430.
L1-norm solution for MEG. Neuroimage 2006;31(3):1025- 109. Yoshor D, et al. Spatial attention does not strongly mod-
1037. ulate neuronal responses in early human visual cortex.
91. Kaneoke Y, Magnetoencephalography: in search of neural J Neurosci 2007;27(48):13205-13209.
processes for visual motion information. Prog Neurobiol 110. Rokers B, Cormack LK, Huk AC. Disparity- and velocity-
2006;80(5):219-240. based signals for three-dimensional motion perception in
92. Handy T. Event-Related Potentials: A Methods Handbook. human MT+. Nat Neurosci 2009;12(8):1050-1055.
Cambridge, Mass: The MIT Press, 2004. 111. Weigelt S, et al. Orientation-selective functional magnetic
93. Skottun BC, Skoyles JR. A few remarks on relating reaction resonance imaging adaptation in primary visual cortex re-
time to magnocellular activity. J Clin Exp Neuropsychol visited. Hum Brain Mapp 2011.[CE: Please check this ref]
2007;29(8):860-866. 112. Logothetis NK. What we can do and what we cannot do
94. Marg E, Rudiak D. Phosphenes induced by magnetic stim- with fMRI. Nature 2008;453(7197):869-878.
ulation over the occipital brain: description and prob- 113. Schummers J, Yu H, Sur M. Tuned responses of astrocytes
able site of stimulation. Optom Vis Sci 1994;71(5):301- and their influence on hemodynamic signals in the visual
311. cortex. Science 2008;320(5883):1638-1643.
95. Ray PG, et al. Magnetic stimulation of visual cortex: factors 114. Nayar VV, et al. Preservation of visual fields with the infe-
influencing the perception of phosphenes. J Clin Neuro- rior temporal gyrus approach to the atrium. J Neurosurg
physiol 1998;15(4):351-357. 2009;110(4):740-743.
CHAPTER 16 MAPPING OF THE HUMAN VISUAL SYSTEM 239

115. Yasargil MG, et al. The selective amygdalohippocampec- 132. Shibasaki H, Ikeda A, Nagamine T. Use of mag-
tomy for intractable temporal limbic seizures. J Neurosurg netoencephalography in the presurgical evaluation of
2010;112(1):168-185. epilepsy patients. Clin Neurophysiol 2007;118(7):1438-
116. Taoka T, et al. Diffusion tensor tractography of the Meyer 1448.
loop in cases of temporal lobe resection for temporal lobe 133. Grover KM, et al. Retrospective review of MEG visual
epilepsy: correlation between postsurgical visual field de- evoked hemifield responses prior to resection of temporo-
fect and anterior limit of Meyer loop on tractography. parieto-occipital lesions. J Neurooncol 2006;77(2):161-
AJNR Am J Neuroradiol 2008;29(7):1329-1334. 166.
117. Roux FE, et al. Perimetric visual field and functional 134. Inoue T, et al. Combined three-dimensional anisotropy
MRI correlation: implications for image-guided surgery in contrast imaging and magnetoencephalography guidance
occipital brain tumours. J Neurol Neurosurg Psychiatry to preserve visual function in a patient with an occipi-
2001;71(4):505-514. tal lobe tumor. Minim Invasive Neurosurg 2004;47(4):249-
118. Hirsch J, et al. An integrated functional magnetic reso- 252.
nance imaging procedure for preoperative mapping of 135. Burneo JG, et al. Cortical reorganization in malformations
cortical areas associated with tactile, motor, language, and of cortical development: a magnetoencephalographic
visual functions. Neurosurgery 2000;47(3):711-21; discus- study. Neurology 2004;63(10):1818-1824.
sion 721-722. 136. Freye E. Cerebral Monitoring in the Operating Room and
119. Kollias SS, et al. Functional evaluation using magnetic the Intensive Care Unit: An introductory for the clinician
resonance imaging of the visual cortex in patients with and a guide for the novice wanting to open a window to
retrochiasmatic lesions. J Neurosurg 1998;89(5):780-790. the brain. Part III: Spinal cord evoked potentials. J Clin
120. Fried I, et al. Functional MR and PET imaging of rolandic Monit Comput 2005;19(1-2):169-178.
and visual cortices for neurosurgical planning. J Neuro- 137. Tobimatsu S, Celesia GG, Studies of human visual patho-
surg 1995;83(5):854-861. physiology with visual evoked potentials. Clin Neuro-
121. Peltier J, et al. Optic radiations: a microsurgical anatomical physiol 2006;117(7):1414-1433.
study. J Neurosurg 2006;105(2):294-300. 138. Kamada K, et al. Functional monitoring for visual
122. Sincoff EH, Tan Y, Abdulrauf SI. White matter fiber dis- pathway using real-time visual evoked potentials and
section of the optic radiations of the temporal lobe and optic-radiation tractography. Neurosurgery 2005;57(1
implications for surgical approaches to the temporal horn. Suppl):121–127; discussion 121-127.
J Neurosurg 2004;101(5):739-746. 139. Fitzgerald PB, Fountain S, Daskalakis ZJ. A comprehen-
123. Powell HW, et al. MR tractography predicts visual field sive review of the effects of rTMS on motor cortical ex-
defects following temporal lobe resection. Neurology citability and inhibition. Clin Neurophysiol 2006;117(12):
2005;65(4):596-599. 2584-2596.
124. Govindan RM, et al. Diffusion tensor imaging of 140. Neggers SF, et al. A stereotactic method for image-guided
brain plasticity after occipital lobectomy. Pediatr Neurol transcranial magnetic stimulation validated with fMRI and
2008;38(1):27-33. motor-evoked potentials. Neuroimage 2004;21(4):1805-
125. Silverman DH, et al. Evaluating early dementia with and 1817.
without assessment of regional cerebral metabolism by 141. Sack AT, et al. Imaging the brain activity changes underly-
PET: a comparison of predicted costs and benefits. J Nucl ing impaired visuospatial judgments: simultaneous FMRI,
Med 2002;43(2):253-266. TMS, and behavioral studies. Cereb Cortex 2007;17(12):
126. Chen W. Clinical applications of PET in brain tumors. 2841-2852.
J Nucl Med 2007;48(9):1468-1481. 142. Sack AT, et al. The temporal characteristics of motion pro-
127. Wong FC, et al. Occipital hypometabolism demon- cessing in hMT/V5+: combining fMRI and neuronavigated
strated by positron emission tomography after tempo- TMS. Neuroimage 2006;29(4):1326-1335.
ral lobectomy for refractory epilepsy. J Neuroophthalmol 143. Blume WT, Whiting SE, Girvin JP. Epilepsy surgery in the
2004;24(1):19-23. posterior cortex. Ann Neurol 1991;29(6):638-645.
128. Sturm JW, et al. Ictal SPECT and interictal PET in the lo- 144. Curatolo JM, et al. Intraoperative monitoring to pre-
calization of occipital lobe epilepsy. Epilepsia 2000;41(4): serve central visual fields during occipital corticectomy
463-466. for epilepsy. J Clin Neurosci 2000;7(3):234-237.
129. Eickhoff SB, et al. Organizational principles of human vi- 145. Penfield W. Some Mechanisms of Consciousness Discov-
sual cortex revealed by receptor mapping. Cereb Cortex ered during Electrical Stimulation of the Brain. Proc Natl
2008;18(11):2637-2645. Acad Sci U S A 1958;44(2):51-66.
130. Gerstl F, et al. Multimodal imaging of human early visual 146. Nguyen HS, et al. A method to map the visual cortex dur-
cortex by combining functional and molecular measure- ing an awake craniotomy. J Neurosurg 2011;114(4):922-
ments with fMRI and PET. Neuroimage 2008;41(2):204- 926.
211. 147. Duffau H, et al. Intra-operative mapping of the subcor-
131. Stefan H, et al. Magnetoencephalography (MEG) predicts tical visual pathways using direct electrical stimulations.
focal epileptogenicity in cavernomas. J Neurol Neurosurg Acta Neurochir (Wien) 2004;146(3):265-269; discussion
Psychiatry 2004;75(9):1309-1313. 269-2670.
This page intentionally left blank
Chapter 17
Mapping of Hearing
Albert J. Fenoy1 and Matthew A. Howard 2
1
Department of Neurosurgery, University of Texas Health Science Center, Houston, Texas
2
Department of Neurosurgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa

䉴 INTRODUCTION AND R share a common low-frequency border, and R and RT


HISTORICAL PERSPECTIVE may share a high-frequency border7,10 ; refer to Fig. 17–1.
As distance increases from the core region, frequency
More than 125 years ago, the superior temporal gyrus tuning to pure tones becomes less sharp, although tono-
(STG) was identified as the site of cortical areas involved topic gradients are shown.4,9,11 These belt neurons have
in auditory processing in both monkeys,1 and then later more complex receptive field properties than the core
in the human.2 It was thought that functional specializa- and prefer complex tones,12 showing optimal response
tion of cortex did exist, but it was not later until archi- profiles as a function of stimulus bandwidth for broad-
tectonic mapping techniques (e.g., Brodmann (1909)3 ) band sounds as well as sensitivity to direction and extent
could definitively differentiate cortical areas in the of frequency modulation.9,11,13−16 The parabelt region
human brain. has even more complex and less definable “preferred
Several investigators have mapped the subdivisions stimuli,” projecting more diffusely, with cortico-cortico
and connections of the auditory cortex in nonhuman connectivity patterns that are similarly complex.
primates using microelectrode recording techniques Human auditory cortex similarly exhibits tonotopic
and histological and tracing methods.4−8 In agreement organization. Several studies have found evidence for
among these studies is the existence of “core” auditory cortical tonotopy using noninvasive recordings to calcu-
fields, located on the superior temporal plane, which are late the equivalent dipole source location as a function
surrounded in a belt-like arrangement by other cortical of frequency.17−20 Howard21 has demonstrated the ex-
fields within STG with a variety of distinct cytoarchitec- istence of a tonotopic organization in human primary
tural and physiological features. auditory cortex (PAC) on medial Heschl’s gyrus (HG)
Tonotopic organization is a hallmark feature of the directly using single-unit recordings from a stereotacti-
auditory sensory system as it ascends from cochlea cally implanted depth electrode on STG. Over the past
to cortex. The degree of both the frequency tuning 10 years, our lab has been able to directly record si-
resolution—neuronal predilection to respond best (or multaneous activity within medial HG, lateral HG, and
fire) to a certain “best frequency”—and tonotopic organi- STG and provide evidence for the existence of multiple
zation changes as the encoded information moves from auditory fields in the human, each associated with dif-
subcortical regions to the cortex, where neurons with a ferent response latencies and preferred stimuli (Brugge
similar “best frequency” are arranged in rows that are and Howard, unpublished data).
organized along a frequency gradient (e.g., Merzenich
and Brugge (1973)4 ). Electrophysiological testing in each
defined field, in both the monkey and human, has pro-
vided well-defined characteristics of neuronal behavior 䉴 OVERVIEW OF NEUROANATOMY
that further help to distinguish and classify these regions. AND NEUROPHYSIOLOGY
Although homologies are not exact, it is useful to look
at these well-researched nonhuman primate studies as Nonhuman primate studies on architecture, connectivity,
an introduction to modeling human auditory cortical or- and tone-frequency serve as comparative models for the
ganization and function. sparse data obtained in humans, and continue to guide
In the macaque monkey, three distinguishable fre- ongoing research in human auditory cortical organiza-
quency gradients have been reported in the core re- tion and function.
gion, each responding well with short latencies to pure Kaas and Hackett10,22−24 present a model of au-
tones, with narrow frequency tuning at their character- ditory cortical organization in nonhuman primates in
istic frequency8,9 ; they are labeled A1 (primary area), which a primary “core” field, located upon the lower
R (rostral area) and RT (rostrotemporal area). A1 and bank of the transverse (sylvian) sulcus, is encircled by

241
242 SECTION II SYSTEMS

duced cell density and columnar spacing, and less dense


myelination relative to the core; inputs are received both
from the adjacent core field and from dorsal and medal
divisions of the MGN. The parabelt receives afferents
largely from neurons in the belt,10,22,24 but also from the
MGN.
Human auditory cortex is likewise comprised of
an array of fields distributed both on the exposed sur-
face of the STG and on the supratemporal plane, deep
within the sylvian fissure (SF) beneath the overlying
parietal cortex. Although the number and boundaries
A
of the fields are not well known, and homologies with
the well-researched cortical auditory fields of nonhuman
primates not well verified, cytoarchitectonic studies have
been able to consistently identify an area of koniocortex
confined to the posteromedial two-thirds of the first
transverse temporal gyrus, or HG, located on the
supratemporal plane.25,26 This area, considered by most
to be the site of PAC or A1, has a high-cell density (espe-
cially in layers II–IV), is heavily myelinated, and exhibits
a distinct but inhomogeneous chemoarchitecture27−31
suggesting that perhaps it may be more representative
of a primary cortical complex, or auditory core, homol-
ogous to the monkey and composed of more than one
field.32
B Anatomical studies have also consistently shown
a belt of cortical fields on the supratemporal plane
Figure 17–1. Lateral view of the macaque cerebral surrounding the core koniocortex, extending into the
cortex. (Adapted from Kaas JH, Hackett TA. insula31 and the frontal and parietal operculum.27 By
Subdivisions of auditory cortex and processing using morphological criteria, much of this nonprimary
streams in primates. Proc Natl Acad Sci U S A
region has been segregated into planum polare (PP, ante-
2000;97:11793–11799; copyright 2000, National
Academy of Sciences, USA). (A) The approximate rior to HG) and planum temporale (PT, posterior to HG),
location of the parabelt region of the superior but borders are not clearly defined (see Fig. 17–2).33
temporal gyrus (STG) (dashed orange line). (B) Architectonic3,25,27,32,34,35 and histochemical30,31 parcel-
Dorsolateral view after removal of the overlying lations of this nonprimary cortex, although not in full
parietal cortex, as demarcated by the dashed black agreement on number and location, have identified as
line. The approximate locations of the core region many as seven belt fields. One or two auditory fields
(solid red line), caudal and lateral portions of the belt
have been identified lateral to belt fields, on the postero-
region (dashed yellow line), and the parabelt region
(dashed orange line) are shown. The core region lateral exposed surface of the STG,30,36 possibly analo-
contains three subdivisions: primary area (A1), rostral gous to the parabelt fields seen in the monkey. Simi-
area (R), rostrotemporal area (RT). The medial larly, our laboratory has identified a circumscribed area
portion of the belt region and rostral pole of the core that overlaps a region on the posterolateral aspect of
in the ventral circular sulcus are not visible. AS, the STG and is activated by acoustic stimulation, a re-
arcuate sulcus; CS, central sulcus; LS, lateral sulcus; gion we refer to as posterior lateral superior temporal
INS, insula, STS, superior temporal sulcus.
area (PLST); see later and Fig. 17–2.33
A hierarchical model derived from anatomical and
nonprimary belt fields and on the lateral aspect by para- physiological studies suggests that information about
belt fields (see Fig. 17–1), which, in turn, make connec- spectrotemporal features of a natural sound are pre-
tions with more distant cortex of the temporal, parietal served in core cortex and from there transmitted to belt
and frontal lobes. and parabelt fields in serial as well as parallel fash-
The core has a highly granular and densely myeli- ion, where ultimately through further circuitous interac-
nated appearance, and receives ascending inputs from tions they are transformed into more complex cerebral
the ventral medial geniculate nucleus (MGN). The core representations.10,37
projects to ipsilateral and contralateral core areas, as well The exact homology between human and nonhu-
as to the adjacent belt region, which includes seven man primate for belt and parabelt fields is less known
or eight nonprimary fields. These belt areas have re- relative to that observed in core koniocortex.26,29,32,36
CHAPTER 17 MAPPING OF HEARING 243

A B

Figure 17–2. Photographs of the lateral surface (A) and superior temporal plane
(B, C) of a postmortem human brain showing the relative size and location of
posterior lateral superior temporal area (PLST) (blue) estimated from the distributions
of averaged click-evoked evoked potentials (EPs) recorded in our experimental
subjects (see Howard MA, Volkov IO, Mirsky R, et al. Auditory cortex on the posterior
superior temporal gyrus of human cerebral cortex. J Comp Neurol 2000;416:76–92,
for more details). The mesial auditory field on Heschl’s gyrus (HG) from which auditory
EPs were recorded is designated A1 (green). The dashed arrow indicates a projection
from the mesial Heschl’s gyrus (HG) auditory field (A1) to PLST. (Reprinted with
permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.)

Cortico-cortico connectivity studies10 as well as single In humans, a recent meta-analysis of functional


cell recordings15,38 have been used to define parabelt imaging studies by Arnott et al.43 has supported the dual
borders in the monkey, with rostral (anterior) parabelt stream hypothesis by demonstrating that posterior pari-
receiving afferents from anterior belt areas and project- etal and superior frontal regions were more frequently
ing anteriorly to multiple sites within the temporal lobe activated during auditory spatial processing.30,44−46 Fur-
and into the ventrolateral frontal cortex, and caudal (pos- thermore, fMRI studies have provided evidence for
terior) parabelt receiving input from posterior belt, and the implication of posterior temporal cortex in spatial
projecting posteriorly and dorsally into the temporopari- processing.47,48 In contrast, anterior temporal and infe-
etal junction and dorsolateral prefrontal cortex.10,24,38 rior frontal areas were more often activated during non-
For illustrative purposes, Fig. 17–314 depicts the so-called spatial auditory processing.45,49,50 Fig. 17–4 depicts the
“what” and “where” streams as hypothesized in their results of the meta-analysis of the 11 sound localization
studies on the macaque. and 27 nonspatial sound studies in which activation was
Nonetheless, evidence from functional magnetic seen by either positron emission tomography (PET) or
resonance imaging (fMRI) studies suggests that a func- fMRI, plotted in Talairach space.43
tional hierarchy may also exist for human auditory Such noninvasive methods favor homology be-
cortex,39 where cortical processing of complex sound tween human and nonhuman primate data, but it is still
occurs in two separate streams.14,40−42 essential that studies using complimentary experimental
244 SECTION II SYSTEMS

Figure 17–3. Schematic flow diagram of “what” and “where” streams in the auditory
cortical system of primates, as depicted by Rauschecker and Tian. (Rauschecker JP,
Tian B. Mechanisms and streams for processing of “what” and “where” in auditory
cortex. Proc Natl Acad Sci U S A 2000;97(22):11800–11806; copyright 2000, National
Academy of Sciences, USA). The ventral “what” stream is shown in green, the dorsal
“where” stream in red. PFC, prefrontal cortex; PP, posterior parietal cortex; PB,
parabelt cortex; AL, ML, CM and CL, anterolateral, middle lateral, caudomedial, and
caudolateral auditory belt; T2/T3, anterior temporal lobe; MGd and MGv, dorsal and
ventral parts of the medial geniculate nucleus.

approaches, including invasive mapping, be undertaken and location of HG and more lateral STG. More specific
if we are to fully understand the functional organization anatomy of the upper surface of the temporal lobe is
of human auditory cortex and the substrates involved in shown in a dissection by Rhoton52 in Fig. 17–5. Here, the
the processing and perception of complex sound, such three gross partitions of the superior temporal plane—
as speech. PP, HG, and PT, can be clearly visualized.
von Economo and Horn,53 as well as Pfeifer54,55 out-
lined in detail the normal anatomy of the supratemporal
䉴 MAPPING METHODS plane, characterized by one strongly developed convo-
lution, or transverse gyrus, known as HG on the left side
STRUCTURAL IMAGING AND and two on the right, as confirmed by others.56−58 This
OPERATIVE ANATOMY pattern, so-called “Pfeifer’s norm,”53 is however quite
variable, with the frequent occurrence of several trans-
Gross Anatomy
verse gyri on either side of the brain.
Understanding the neurophysiology and neuroanatomy Broca59 and Wernicke60 first inferred a functional
of the auditory cortex at the superstructural or gross asymmetry of the brain for language following from ob-
anatomical level provides insights into the spatial or- servations that language could be disrupted in patients
ganization of distributed neural networks that mediate affected by lesions on the left side of the brain. Anatom-
different aspects of acoustic sound perception. ically, it has been shown that the SF is asymmetric at its
Although “auditory cortex” may not signify a specific, posterior end,61 where the right side curves upward pos-
easily definable brain structure, historic cross-modality teriorly and ends at a higher position that the left, and in
experimentation on both human and nonhuman pri- the majority of cases of shorter length than the left. The
mates has revealed that most neurons chiefly responsive PT, which lies on the superior surface of the temporal
to acoustic stimuli reside in the superior portion of the lobe posterior to HS, (Heschl’s sulcus, the sulcus pos-
temporal lobe, both on the exposed STG and on the terior to the first transverse temporal gyrus (or HG)) is
hidden supratemporal plane, deep within the SF.51 influenced by SF asymmetry as it is mostly larger on the
In Fig. 17–233 are photographs of a postmortem hu- left side.54−55 Such asymmetry might be contributive in
man brain specimen depicting the lateral surface (A) and defining language dominance, although no defining evi-
superior temporal plane (B, C), showing the relative size dence has been provided. (see Shapleske et al. (1999)62 ).
CHAPTER 17 MAPPING OF HEARING 245

Planum polare

Superior temporal gyrus

Temporal stem
Temporal horm

Heschl’s gyrus

Transverse temp. gyri


Planum temporale

Atrium

Figure 17–5. Superior view of the upper surface of


the left temporal lobe that forms the floor of the
sylvian fissure (SF). It has been disconnected medially
from the thalamus through the choroidal fissure,
anteriorly and medially from the globus pallidus,
anteriorly and laterally from the insula, and superiorly
from the temporal stem. The superior surface is also
called the opercular surface, and it is the temporal
Figure 17–4. Sagittal and axial views of all spatial operculum of the SF. It presents three
(blue triangles) and nonspatial (red spheres) data morphologically distinct parts, from anterior to
points showing activation as compiled from the posterior: the planum polare (PP) (free of gyri and
meta-analysis by Arnott et al. (2004) (Arnott SR, has a shallow trough to accommodate the course of
Binns MA, Grady CL, Alain C. Assessing the auditory the middle cerebral artery), Heschl’s gyrus (or the
dual-pathway model in humans. Neuroimage most anterior transverse temporal gyrus), and the
2004;22(1):401–408.), plotted in Talairach space. planum temporale (usually formed by the transverse
Sagittal (x = +29 mm) and axial (z = −4 mm) Talairach temporal gyri posterior to Heschl’s gyrus). The lateral
images are displayed for reference. edge of the PP is formed by the superior temporal
gyrus. The stem of the temporal lobe, the relatively
thin layer of white and gray matter that connects the
Structural Imaging: MRI and Diffusion temporal lobe to the lower insula, is positioned above
Tensor MRI the lateral and anterior edge of the temporal horn.
The temporal horn and the atrium are inferior and
Current MRI protocols are able to provide relatively fine posterior segments of the lateral ventricle,
definition of cortical and subcortical structures in indi- respectively. (Adapted from Rhoton AL Jr. The
vidual subjects, with quite exquisite detail that improves Supratentorial cranial space: microsurgical anatomy
with increased magnet strength. As elsewhere, our lab- and surgical approaches. Neurosurgery
oratory creates three-dimensional (3D) MRIs to help lo- 2003;53(Suppl, Part 2):39).
calize cortical areas of interest with respect to electrode
placement, and the derived models simulate anatomy
well (e.g., see Fig. 17–14B; Brugge and Howard, unpub-
lished data). (DW) MRI have shown that, by applying diffusion
Although conventional T2-weighted MRI of the weighting in at least six different directions, one can ob-
brain has been unable to provide information on sub- tain the diffusion tensor (DT) and quantitative measure-
cortical fiber direction, advances in diffusion weighted ments of the anisotropy of the white matter tracts. Studies
246 SECTION II SYSTEMS

of auditory cortex using DT imaging (DTI), though, are As there are many different parameters that con-
few. tribute to our understanding of auditory cortex physi-
By using DTI, Hiwatashi et al.63 were able to show ology, it is prudent to concentrate on the response of
on normal subjects that the fractional anisotropy of the auditory cortex to single parameter variance. Later in
subcortical white matter of first HG to be higher than that the chapter we have explored and summarized the cur-
in the ipsilateral STG. This finding supports the presence rent findings from functional imaging studies investigat-
of a more prominent “core” projection of the acoustic ing cortical response properties to individual acoustic
radiation to HG relative to STG, commensurate with the parameter testing.
evidence of “core” PAC on HG and “parabelt” on more
lateral STG.
Frequency
Recently, Upadhyay et al.64 through stroboscopic
event-related fMRI, were able to demonstrate mirror- Within the auditory system, by far the parameter of
symmetric tonotopic organization within the human frequency provides the most useful organizational infor-
PAC, and, by using anatomical functional image analy- mation. Several groups have utilized neuroimaging tech-
sis in conjunction with DT fiber tractography and prob- niques as a means to noninvasively detail tonotopy in
abilistic mapping, characterize the axonal projection humans.17,21,67−81 These studies have depicted spatially
patterns present within tonotopically defined areas. DTI distinct activity based on responses to tones varying in
reconstructed fiber projections revealed the presence of frequency, some depicting mirror-symmetric tonotopy,
both cross-field isofrequency and within-field nonisofre- arousing the hypothesis that the connectivity patterns
quency-specific axonal projections in human PAC, where observed within the PAC in nonhuman primate tracer
the number of short-length nonisofrequency-specific studies also exist in the human.
fibers located within the same auditory fields sig- Two PET studies have reported more laterally lo-
nificantly outnumbered the long-range isofrequency- cated auditory activation by a 500-Hz tone than by a
specific fibers located in distinct auditory fields (see 4000-Hz tone,67,76 although the precise locations are
Fig. 17–6)64 . This suggests that the majority of projec- indeterminate due to the low resolution of the tech-
tions within the human PAC remain intrinsic to that au- nique. fMRI, however, has corroborated these findings
ditory region, similar to the observations in primate audi- of separate response areas to low- and high-frequency
tory cortices,65 where structural connectivity within and tones.72,73 The fMRI investigation by Talvage et al.82 re-
among distinct auditory fields exists and is necessary to vealed multiple tonotopic fields encompassing both the
integrate and process various sound properties. HG and surrounding fields on the STG using a variety of
increasingly complex stimuli, from pure tones to music.
At least four tonotopic gradients were identified, with
locations ascertained by the overlay of individual func-
FUNCTIONAL IMAGING tional maps onto anatomical maps for each of six sub-
jects; one such high–low frequency gradient seems to ex-
PET and fMRI provide good spatial maps of brain acti- ist from posteromedial to anterolateral HG, with adjacent
vation, with resolution of a few centimeters or less, but tonotopic fields sharing either a low- or high-frequency
temporal resolution is poor because they reflect changes border, possibly homologous to the macaque.
in blood flow; they are both indirect measures of brain Recent fMRI work performed by Formisano et al.79
activity, and are fundamentally limited by the character- and Talvage et al.80 has shown activation maps in PAC
istics of the hemodynamic response on which the acti- similar to the mirror-symmetric tonotopic organization
vation signal is based.66 In contrast, electroencephalog- revealed by electrophysiological recordings in nonhu-
raphy (EEG) is sensitive to scalp surface measurements man primates24 and intracranially in humans, to be dis-
of electrical activity produced by synchronous neural fir- cussed next.21,33,83,84
ings in the brain, and magnetoencephalography (MEG) Nonprimary auditory areas corresponding to the
is sensitive to scalp magnetic fields directly generated by belt regions in the macaque are believed to be more pref-
this electrical activity. Thus both measures provide infor- erentially activated by more complex, broadband sounds
mation about the millisecond time course of brain activ- than pure tones, as in the core. This is supported by the
ity, but they suffer from estimations in the source of the fMRI study by Wessinger et al.39 in which responses to
electrical generators, leading to poor spatial resolution. single-frequency tones at 0.5 kHz, 2 kHz or 8 kHz were
Let us fist look at the information obtained by func- far less widespread than those obtained using 1-octave
tional mapping. fMRI has become more prevalent in re- wide, band-passed noise at the same center frequencies
cent years than PET due to the ease by which it can (see Fig. 17–7). Hall et al.85 also reported greater activa-
be administered, it’s more widespread availability, and tion beyond HG when activation by a single-frequency
the fact that multiple observations can be made on the tone was subtracted from that by a harmonic complex
accuracy of the derived maps. tone of the same pitch.
CHAPTER 17 MAPPING OF HEARING 247

A B

C D

Figure 17–6. Mirror symmetric tonotopic organization for one subject. (Adapted from
Upadhyay J, Ducros M, Knaus TA, et al. Function and connectivity in human primary
auditory cortex: a combined fMRI and DTI study at 3 Tesla. Cereb Cortex
2007;17(10):2420–2432). (A) Two high-frequency regions, one caudomedial and one
rostrolateral, were mapped within primary auditory cortex (PAC); between these
areas was a low-frequency region. Activation maps are present within the PAC,
specifically along the transverse temporal gyrus of Heschl (TTG) and transverse
temporal sulcus (TTS) and spanned a distance of 25.2 mm, as depicted on an inflated
brain representation in the Talairach coordinate system. (B) Three-dimensional
high-frequency (blue) and low-frequency (light blue–green) activation clusters were
projected and created in coregistered T1-weighted structural magnetic resonance
imaging and DTI datasets in the non-Talairach coordinate system. These
three-dimensional clusters were used as seeding points for fiber tractography and
probabilistic mapping. LS, lateral sulcus; PSTG, posterior superior temporal gyrus;
STG, superior temporal gyrus; SIG, short insular gyrus. (C) Zoomed view of a sagittal
slice depicting the isofrequency-specific fibers connecting the caudomedial and
rostrolateral high-frequency region of interests (ROIs) (blue). (D) Zoomed view
depicting the nonisofrequency-specific fibers (low-high frequency fibers) projecting
from the low-frequency ROI (light blue–green) to both caudomedial and rostrolateral
high-frequency regions, which are located within the dashed red lines

extent,76,86 or only an increase in either extent87 or


Sound Level
magnitude.88 A few fMRI studies have investigated the
PET and fMRI studies have reported systematic changes differential sensitivity to sound level across subregions
in activation of the auditory cortex with changing sound of the auditory cortex,86,89 agreeing that the majority of
level, although evidence is not uniform. Activation has cortex most sensitive to increasing sound level corre-
been seen with an increase in both magnitude and sponded to PAC on HG.
248 SECTION II SYSTEMS

Figure 17–7. Axial slices for two subjects showing their individual auditory activation
as a function of frequency spectrum. (Adapted from Wessinger CM, VanMeter J, Tian
B, Van Lare J, Pekar J, Rauschecker JP. Hierarchical organization of the human
auditory cortex revealed by functional magnetic resonance imaging. J Cogn Neurosci
2001;13:1–7). Activation is plotted for all voxels exceeding Z = 14.2. Areas shown in
blue represent activation by pure tones, yellow represents activation by band-passed
noise and green represents the overlap between the two activation patterns.

Temporal Aspects of Sound


pitch, or increased temporal regularity, perhaps giving
Animal vocalizations and human speech alike rely on a evidence for the engagement of HG based on temporal
processor that is able to continuously extract changing cues. To better differentiate responsiveness to different
frequency and amplitude information over time. Tem- parameters of temporal acoustic structure, Hall et al.91
poral acoustic patterns are important determinants of used fMRI to find the greatest activation to both pitch and
the sound stimulus and can be broadly classified into increasing temporal regularity (and thus pitch strength)
three divisions: envelope (<50 Hz), periodicity (50–500 in lateral HG, whereas reliable activations related to both
Hz), and fine structure (>500 Hz). 90,91 For human listen- pitch and the spatial width of sound to be localized to
ers, temporal variations ≤50 Hz are important for accu- PT (see Fig. 17–8). These activation patterns were in-
rate speech recognition (e.g., Drullman et al. (1994,)92,93 ; terpreted by the authors to be consistent with a larger
Shannon et al. (1995)94 ), whereas waveforms exceeding role of lateral HG in perceptual analysis than temporal
50 Hz create the percept of pitch. Acoustic temporal acoustic structure, and an overall concept that more lat-
structure can be present separately in each channel, eral auditory cortex (including PT) integrates different
but can also be created through binaural interaction classes of sound information to form auditory objects.
across channels. The fine temporal relationships be- Spectrotemporal patterns of sound have also been
tween the signals at the two ears generally determine studied using frequency- and amplitude-modulated (FM
spatial qualities such as the width and location of the and AM) tones. fMRI studies on AM (e.g., Giraud et al.
sound source.91,95 (2000)104 ) and FM tones85 reported significant responses
Pitch is the auditory percept associated with the fre- in bilateral primary and nonprimary areas, strongest
quency ( f ), and hence the period (T = 1/ f ), of sound in the posterolateral regions of the STG. Zatorre and
wave vibrations in the audible frequency range (∼20 Hz– Belin105 independently manipulated the frequency and
20 kHz).51,66 By using manipulations in temporal regu- timing information in sequences of single frequency
larity (and thus altering pitch), recent PET and fMR imaging tones, comparing responses to each modulated sound to
studies have identified regions of the auditory cortex each other, rather to an unmodulated control as in other
that are preferentially activated by temporal acoustic pat- studies;85,104 here, greater responses to spectral changes
terns at rate >50 Hz, defining that lateral HG and PT (over temporal changes) were found instead in bilateral
were the most highly sensitive to periodicity95−101 and anterior STG. Griffiths et al.96,106 found via PET imaging
fine structure102,103 above all areas studied. Such activa- that both anterior and posterior STG were preferentially
tion was also seen to increase with the strength of the activated by a melodic pitch sequence.
CHAPTER 17 MAPPING OF HEARING 249

Figure 17–8. Results from the group analyses showing the patterns of sensitivity to
pitch- and location-related acoustic attributes. The group activation maps were
derived from conventional t-test and the four stimulus effects are represented by solid
colors. All maps are overlaid onto the same five horizontal brain images (z = 16-16
mm). This base image is an average of the 64-slice whole brain set that was acquired
for all participants and is displayed in neurological convention (Left = Left). (Adapted
from Hall DA, Barrett DJ, Akeroyd MA, Summerfield AQ. Cortical representations of
temporal structure in sound. J Neurophysiol 2005;94(5):3181–3191, for individual
analyses).

Together, the results of these studies give evidence ies support lateralization in the processing of acous-
that the processing and analysis of combined spec- tic space,96,106,107,112,113 while others found no evidence
trotemporal information in sound relies mostly on non- of such.114,115 However, right hemisphere dominance is
PAC, with activity moving anterolaterally away from HG more often suggested than none. A behavioral study by
as the processing of melodic sounds proceeds. Zatorre and Penhune116 based on patients with unilateral
temporal lobe lesions concluded that damage to the right
anterior STG, not encroaching upon the PAC, produced
Sound Localization a sound localization deficit in both hemifields. Palomaki
Rauschecker and Tian,14 and similarly Kaas and et al.117 had similar observations in their MEG study find-
Hackett,24 suggest, based on nonhuman primate stud- ing strong N1m responses bilaterally but increased acti-
ies, that the auditory space is processed in a posterior vation in the right hemisphere for either right- or left-
(caudal) pathway leading dorsally from the PAC in HG lateralized noise bursts.
up to the inferior parietal cortex through the caudal belt Brunetti et al.118 employed both fMRI and MEG to
and parabelt in the STG, a so-called “where” pathway, localize activation as well as to disclose temporal dy-
whereas nonspatial auditory information is processed in namics during the processing of sounds coming from
an anterior (rostral) direction along the anterior portions different auditory locations (right, left, or both). The fMRI
of the lateral belt, or “what” stream (see Fig. 17–3).14 results indicated activation of a complex circuit involv-
Several studies in humans have corroborated this ing HG, posterior STG, supramarginal gyrus, and frontal
concept. Investigations using a sound localization lobe; concomitant MEG data analysis observed sequen-
task45,107 have found that the inferior parietal lob- tial increased latencies in activation times, starting with
ule is more preferentially activated by spatial process- HG, suggesting a pathway analogous to the “where”
ing than by frequency discrimination, or even sound pathway in monkeys. Both hemispheres were activated,
identification.46 This area’s ability to be activated in but the right hemisphere was activated more strongly
the disambiguation of overlapping auditory sources108 (see Fig. 17–9).118 These results are in accordance with
is suggestive that the spatial properties of sound acti- other studies confirming the presence of such a “dorsal
vate a posterior nonprimary auditory circuit. A recent stream” activation path in spatial processing involving
meta-analysis of PET and fMRI studies of auditory posterior STG, parietal and frontal regions.45,46,119
processing,43 also, underlined the importance of the in-
ferior parietal lobe in sound localization (as discussed
previously, see Fig. 17–4). As seen in noninvasive
recordings, moving sounds often produce bilateral ac- NONINVASIVE RECORDINGS
tivation in primary and nonprimary regions of the
Surface Electrical Recordings
auditory cortex101,109,110 but may incur greater activa-
tion in a lateral area of the right PT than stationary Unlike functional imaging, noninvasive electrophysio-
sounds89,47 as well as selectively recruit activity in pari- logical recordings provide information about the mil-
etal cortex.96,101,106,111 lisecond time course of brain activity, but have poor
On the other hand, the role of hemispheric domi- spatial resolution. Thus, EEG and MEG data comple-
nance in sound localization is still debated. Some stud- ment the spatial maps derived from PET and fMRI data
250 SECTION II SYSTEMS

x = 52 z = 11

A B

Figure 17–9. (A) Results from the group analysis of 11 patients ( p < 0.05, corr.):
activated areas observed by functional magnetic resonance imaging during auditory
stimulation in the three conditions: Mixed (upper ), right (middle), and Left (lower ).
(Adapted from Brunetti M, Belardinelli P, Caulo M, et al. Human brain activation during
passive listening to sounds from different locations: an fMRI and MEG study. Hum
Brain Mapp 2005;26(4):251–261). In the mixed condition the bilateral Heschl’s gyrus
(HG) and right posterior superior temporal gyrus (STG) were activated and activation
was greater than in the left and right conditions. The STG activation extends caudally
(y = −49). Activation in the supramarginal gyrus is present in this condition only. During
both the right and the left conditions, bilateral HG and the right posterior STG were
activated. (B) MEG analysis of one subject. The four equivalent current dipoles (ECDs)
of the source model are superimposed on the individual structural magnetic
resonance imaging (MRI): (a) bilateral sources in HG; (b) one source in the caudal
region of the STG; (c) one source in the supramarginal gyrus; and (d) the three ECDs
in the right hemisphere are shown in a sagittal view. The positions of the two ECDs in
(b) and (c) were constrained in a cube with 6 mm side centered on the “center of
mass” of the corresponding functional MRI (fMRI) activation. Across nine subjects
undergoing magnetoencephalography analysis for mixed presentations, the mean
peak latency from stimulus onset was 139 milliseconds in bilateral HG, 156 milliseconds
in caudal STG, and 162 milliseconds in the supramarginal gyrus. (Both (A) and (B) are
reprinted with permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.)

reviewed previously and for the most part corroborate Electrophysiology, however, has provided the most
such findings. The poor spatial resolution of EEG or MEG direct demonstration of a frequency gradient along HG.
techniques can be greatly offset when complemented by Using MEG and EEG, a systematic relationship has been
the adjunctive use of functional imaging studies (e.g., shown between dipole sources and stimulus frequency
Brunetti (2005)118 ), combing temporal resolution in the in the auditory cortex (e.g., Romani et al. (1982)17,18 ,
first modality and spatial localization in the latter. Huotilainen et al. (1995)19 , Pantev et al. (1995)68 , Pantev
CHAPTER 17 MAPPING OF HEARING 251

et al. (1988)69 , Yamamoto et al. (1992)120 , Lütkenhöner contralateral posterior temporoparietal areas were seen
and Steinsträter (1998)121 , Lütkenhöner et al. (2001)122 , to have enhanced spectral amplitude increases between
Rosburg et al. (2000)123 ). Lütkenhöner and Steinsträter121 approximately 60 Hz and 80 Hz to both sounds. On
demonstrated a lateral shift in the location of the re- the other hand, only biological sounds gave rise to 30
sponse within HG as tone frequency was presented at Hz desynchronization over contralateral sensorimotor ar-
steps of 2000 Hz, 1000 Hz, 500 Hz, and 250 Hz, although eas, parietofrontal gamma coherence increases as well
the exact spatial localization of these dipole locations as beta coherence reductions between sensorimotor and
could not be entirely validated due to the limitations of prefrontal sensors, possibly reflecting activation of mo-
the technique. tor networks involved in the automatic preparation for
On amplitude, EEG recordings show that the ampli- orientating.
tude of the main response progressively increases with Such localization of induced gamma activity at the
sound level,124,125 which is consistent with findings from level of cortical gyri has often been difficult to ascertain,
neuroimaging studies. By using MEG and a model of two though, as the skull acts as a low-pass filter,135 with ar-
dipole sources, Gutschalk et al.126 have shown that both tifacts present in the gamma band from cranial muscle
a source posterior to HG, as well as one on the border activity.136,137 Thus, the signal-to-noise ratio for gamma-
between HG and PT, exhibited an increase in response band activity in invasive electocorticography (ECoG) is
amplitude as the stimulus sound level was increased. much better than it is in scalp EEG, and combined
Most studies investigating a single acoustic parame- with shorter interelectrode distances (∼1 cm), spatial
ter have employed stationary sounds in their protocols, resolution is much improved. Even in studies not ana-
and it has now become apparent, only stationary sound lyzing gamma-band oscillatory activity, intracranial sub-
can be modeled using dipoles located in auditory dural electrode recordings provide detailed information
cortex. In their MEG study, Xiang et al.127 found that of cortical and/or deep white matter neuronal activity
responses to both moving and stationary tones can otherwise not sufficiently resolved. The results of this
be fitted using dipole sources in bilateral auditory technique provide detailed mapping of functional au-
cortex, but only moving sounds are best fitted with an ditory cortex, and examples of such will be elaborated
additional source located in the right parietal cortex. upon in the following.
Ducommum et al.128 used EEG to directly compare
auditory location and motion perception, finding that
moving sounds specifically involve the right parietal DIRECT CORTICAL STIMULATION
cortex. These evoked responses are suggestive of partly
segregated networks for stationary and moving sound, As discussed previously, the results of recent imaging
and corroborate the observations of functional imaging studies reveal that a hierarchical auditory processing
studies of nonauditory cortex activations for motion model for human cortex seems to exist as it does for non-
processing.66,96,106 human primates,39,138 but the anatomical connections
between cortical auditory fields are largely unknown.
Anatomical track-tracing methods using dyes cannot be
conducted in living humans, unlike in the primate. Post-
Gamma Range Oscillations
mortem examinations on human auditory cortex have
In the past decade, there has been much interest in the been performed using DiI injections139 as well as car-
oscillatory activity in the so-called “gamma range,” op- bocyanine dyes and have revealed the presence of both
erationally defined as more than 30 Hz, as it seems to intrinsic140 and extrinsic141 cortico-cortical connections
be indicative of regional cortical processing.129 Synchro- within the STG; only examination of short pathways,
nization at gamma frequencies was hypothesized to form however, can be performed due to technical limitations.
a temporal code that may dynamically “bind” spatially Diffusion tensor imaging is a promising new technique
segregated neurons into networks representing higher to assist in in vivo track tracing, but this method has
order stimulus properties,130,131 which after observation only begun to elucidate functional connectivity in audi-
in primate cortex,132,133 has since been studied exten- tory cortex.64
sively in humans with scalp EEG and MEG. An alternative method of tracing neural pathways in
In auditory cortex, such nonphase locked or “in- the cortical auditory system involves focal electrical stim-
ducible” activity at approximately 30 Hz has been ulation of one cortical site, while systematically mapping
demonstrated by EEG, correlating with word stimuli.68 the resultant evoked activity from distant sites.33,83,142−149
To further identify the networks involved with process- Although no direct information on the exact cellular ori-
ing of spatial auditory information, Kaiser et al.134 used gins, anatomical pathways, or terminal arborations can
MEG to visualize if oscillatory responses differed using a be detailed, this approach does provide us with func-
stationary lateralized artificial distorted noise compared tional connectivity between the site(s) of stimulation and
to a biological sound (barking dog). In this approach, recording in the living human.148
252 SECTION II SYSTEMS

Liegeois-Chauvel et al.83 reported the first evidence Our approach routinely employs a variety of con-
for a functional connection between auditory cortical trolled sounds, ranging from brief click trains to human
fields in the human. They found that electrical stimula- vocalizations, to generate auditory evoked potentials
tion of mesial HG in human evoked activity on laterally (AEPs) that, by simultaneous recordings through these
adjacent HG cortex as well as on the PT. These are ar- electrode arrays, have been used to characterize the
eas that subsequently have been identified both anatom- existence of at least three distinguishable auditory
ically and functionally as part of an auditory belt30,31,39,82 cortical fields—two on HG and a third on the pos-
that surrounds an auditory core.10,24 terolateral surface of the STG. Our ongoing work has
In our series of over 150 epilepsy-surgery patients, thoroughly detailed the functional connectivity of these
we have adopted this method of stimulation, via both regions via the analysis of generated waveforms to elec-
electrical and acoustic stimuli, and simultaneous record- trical stimulation33,148,150 (Brugge and Howard, unpub-
ing at multiple sites to characterize the response patterns lished data).
and functional connections of different auditory cortical Using bipolar electrical stimulation of HG, we have
fields. As part of the treatment plan, in most patients shown that polyphasic evoked activity occurs within a
depth electrodes were inserted into HG on the supratem- circumscribed area that overlaps a region on the pos-
poral plane, while grid electrodes were implanted over terolateral aspect of the STG activated by acoustic stim-
perisylvian cortex of either the left or right hemisphere. ulation, a region we refer to as area PLST33 (see Fig.
The modified hybrid depth electrodes (HDEs) were tar- 17–10).148 The most robust and complex waveform,
geted stereotactically for the left or right HG.21 characterized by having an initial positive component

B
A

C D
Figure 17–10. (A) Magnetic resonance imaging (MRI) showing location of recording
grid on lateral surface, overlaying posterior aspect of superior temporal gyrus (STG)
and extending above sylvian fissure (SF) and below superior temporal sulcus (STS), in
a representative subject. (See Brugge JF, Volkov IO, Garell PC, Reale RA, Howard MA
3rd. Functional connections between auditory cortex on Heschl’s gyrus and on the
lateral superior temporal gyrus in humans. J Neurophysiol 2003;90:3750–3763). (B)
Axial MRI section at level of Heschl’s gyrus (HG) showing trajectory of depth electrode
and location of bipolar stimulation sites within HG. (C) and (D) Response fields on
lateral STG resulting from click-train stimulation and electrical stimulation of mesial
HG. Averaged evoked potentials derived from 100 repetitions of respective stimuli.
Relative positions of recorded waveforms on maps of C and D represent relative
positions of electrodes on recording grid shown in A. Gray lines represent locations of
SF and STS. Asterisks mark sites of maximal response within two response fields.
CHAPTER 17 MAPPING OF HEARING 253

form altered its shape and diminished substantially in


amplitude, further supporting existence of a functional
HG boundary between PAC on mesial HG and an auditory
A belt situated more laterally on HG (see Fig. 17–12).148
These results provide direct evidence for a func-
B tional projection from auditory cortex on HG to an as-
sociational auditory cortex (PLST) on the lateral surface
[L4]
of the STG in human.148 This method, however, useful
intraoperatively for resection surgery (discussion to fol-
STMA low), does not provide detailed information about neural
pathway transmission. Regardless, as we shall see, these
STMB connections give further insight into interpreting the re-
sults of local field potential recordings and single-cell
recordings as our patients are presented with acoustic
STMA stimuli.
Recently, the application of the algorithm of di-
STMB rected coherence (DCOH) has shed new light on au-
50 μv ditory cortical connectivity patterns. Introduced by Saito
50 ms and Harashima151 and based on multivariate autoregres-
sive (AR) models and Granger causality, the algorithm
Figure 17–11. Effects of changing stimulus site on outputs the directional coherences between two EEG
Heschl’s gyrus (HG). (Adapted from Brugge JF, signals recorded from two given leads in distinct cor-
Volkov IO, Garell PC, Reale RA, Howard MA 3rd.
tical regions. This can be viewed in terms of delay in
Functional connections between auditory cortex on
Heschl’s gyrus and on the lateral superior temporal propagating a signal. In epilepsy patients undergoing
gyrus in humans. J Neurophysiol 2003;90:3750– invasive seizure monitoring, Gueguin and colleagues152
3763). Averaged waveforms recorded at eight recorded simultaneous responses to AM white noise in
recording sites at and around site of maximal depth electrodes implanted in the insula, HG, PT, and
amplitude in response to stimulating mesial (A) and Brodmann area (BA) 22. Among other findings, estima-
lateral (B) sites on HG. Asterisks mark site of maximal tions of causality using DCOH found a predominant au-
amplitude of response. Dashed lines mark latency of
ditory stream from medial to lateral within PAC on HG,
negative peaks obtained when stimulus was applied
to mesial HG. unidirectional connections from PAC on HG to lateral
HG and PT, and an overall auditory propagation from
posterior to anterior areas at low modulation frequen-
followed by twin negative peaks, occurred within 50 cies. (see Fig. 17–13).152 These findings suggest both
milliseconds of stimulus onset, and resulted from stim- serial and parallel cortical processing of auditory infor-
ulation of sites within about the mesial half of HG. Re- mation, as inferred in animal studies, which can be in-
ducing the interstimulus interval (ISI) from 1000 to 200 terpreted as reflective of a constant interaction between
milliseconds diminished the amplitude and lengthened adaptive and plastic cortical areas.
the latency of the first peak only. When more lateral
HG was stimulated, the recorded waveforms on PLST
had, when present, an earlier appearing and more at- LOCAL FIELD POTENTIAL
tenuated first negative peak; the second peak was not RECORDINGS FROM INTRACRANIAL
evident. Summarizing these results, it appears that both ELECTRODES
negative peaks in the evoked waveform arise from ac-
tivating one or more corticocortical circuits originating Evoked potentials (EPs) obtained in response to a wide
in mesial HG, as evidenced by the systematic decrease range of both simple and complex sound have been
in latency of the first peak with decreasing distance be- recorded directly from the superior temporal plane
tween stimulation and recording sites; the differential ef- of neurosurgical patients both acutely in the operat-
fect of changing stimulus rate on the response waveform ing room153−159 or chronically through implanted mul-
suggests a complex combination of circuitry and inputs tichannel depth electrodes and surface cortical grid
(see Fig. 17–11).148 Conversely, in the cases in which electrodes.83,84,160−165
PLST was stimulated, a positive–negative complex wave- With concurrent adequate anatomical localization of
form was recorded on HG, corroborating the existence recording sites, these EPs can be distinguished into their
of cortico-cortical circuitry. Moreover, the waveform pro- representation of distinct auditory fields. Posterome-
duced increased in amplitude mediolaterally on HG until dial HG, with robustly responsive, frequency tuned and
a transition zone was reached, whereupon the wave- tonotopically organized neuronal clusters, as recorded
254 SECTION II SYSTEMS

HG

P
L M 1 cm
A
100 μv
100 ms

HG

50 μv
100 ms

Figure 17–12. Waveforms evoked along Heschl’s gyrus (HG) by bipolar electrical
stimulation of most active site on PLST in two representative subjects. (Adapted from
Brugge JF, Volkov IO, Garell PC, Reale RA, Howard MA 3rd. Functional connections
between auditory cortex on Heschl’s gyrus and on the lateral superior temporal gyrus
in humans. J Neurophysiol 2003;90:3750–3763). Location and polarity of bipolar
stimulation shown. Arrows point to site along HG where evoked waveform changed
abruptly. Small circles, high-impedance contacts; Large circles, low-impedance sites.

via HDE by Howard et al.166 and presently in our lab, et al.167 also carried out anatomical reconstruction of
provide evidence for this area being field A1. More an- recording sites by visualizing the electrode tracks with
terolaterally recorded waveforms on HG are of relatively stereotaxic MRI after the electrodes had been removed.
longer latency and lower amplitude,83,84,154 perhaps in- All of these studies inserted depth electrodes perpendic-
dicative of a second auditory “belt” field on HG adja- ular to the exposed lateral surface of the STG, which
cent to the auditory core. EPs recorded directly from the resulted in traversing HG obliquely (see later).
exposed posterolateral STG exhibit waveforms and re- The progress of our laboratory in defining these
sponse sensitivity grossly different from that recorded three distinct auditory fields on the STG are partially
on HG, defined as the posterolateral superior temporal based, among many other stimuli, on the responses to
auditory field (area PLST).33,148,149 a brief click train (5 clicks at 100 Hz) recorded simul-
Over the past decades of recording auditory evoked taneously from 14 microcontacts and 4 macrocontacts
activity directly from the cortex deep within the SF, on a HDE that traversed within gray matter, the long
such clear anatomic reconstruction of electrode sites has axis of HG, and from 96 contacts of a grid overlaying
varied, and ultimately improved with imaging technol- the posterolateral STG. See Fig. 17–14 for a representa-
ogy. By using summary data maps estimating locations tion of a usual scenario (Brugge and Howard, unpub-
anatomically with reference to the temporal pole, de- lished data). At our institution, these recordings occur in
rived from pooled data, Celesia et al.154 unfortunately epilepsy surgery patients over 7–14 days, who as part of
did not take into account intersubject variability. their treatment plan, require such electrode placement
Liegeois-Chauvel and colleagues83,84 utilized cere- for diagnosis and ultimately surgical resection of their
bral angiography to obtain 3D coordinates of each HG epileptogenic focus. Details of the placement and MRI
electrode lead in relationship to the temporal branch of localization of the HDE and grid electrode can be found
the middle cerebral artery, the insula, and the SF. Godey elsewhere,148,166 but can be summarized as follows.
CHAPTER 17 MAPPING OF HEARING 255

By using this click train stimulus we, at our Hu-


man Brain Physiology Laboratory, have been able to
distinguish one field from another based not only on
the waveform of the AEP evoked by the abrupt onset of
the click train but also by the synchronized, frequency-
following response (FFR) to individual clicks in the train.
Figure 17–14 depicts filtered EPs, with details shown in
Fig. 17–15 (Brugge and Howard, unpublished data).
Supporting previous findings,148,149 the robust, polypha-
sic potentials of early latency (9.6 milliseconds) recorded
within posteromedial HG are in contrast to the smaller
amplitude, later negative deflections obtained further an-
terolaterally. Moreover, the ability of posteromedial HG
to have an FFR that was not present more anterolater-
ally signifies the simultaneous recording of two auditory
fields, with the characteristics of posteromedial HG con-
sistent with it being part of auditory core. Also simultane-
Figure 17–13. Corticocortical connections within the ous with the HG recordings were a cluster of polyphasic
auditory areas of one subject. (Adapted from AEPs recorded on the posterolateral aspect of STG that
Guéguin M, Le Bouquin-Jeannés R, Faucon G, were demonstrably different, characterizing field PLST,
Chauvel P, Liégeois-Chauvel C. Evidence of functional
as mentioned previously.33,148,149
connectivity between auditory cortical areas revealed
by amplitude modulation sound processing. Cereb
Differences in temporal response patterns between
Cortex 2007;17(2):304–313). Arrows indicate the auditory cortical fields can also be assessed by their ca-
direction of auditory stream. Localization of pacity to synchronize to AM noise, as has been a key
intracerebral electrodes in auditory cortex is component in our routine laboratory studies as well as
superimposed on the patient’s magnetic resonance elsewhere. Recently, Liegeois-Chauvel and colleagues168
imaging slices. Each dashed line (labeled T, H, and P) used their intracortical stereoelectroencephalography
indicates the anatomical location of a single electrode
array to determine the temporal response properties of
(each white segment corresponding to a given
electrode contact). Each blue, yellow, orange-red,
different auditory cortical areas in humans by record-
white, or green-circled line indicates which auditory ing the phase-locked neural activity to AM white noises.
region is recorded. Note that a single electrode can Traversing PAC in medial HG, secondary auditory cor-
record cortical activity from different areas. Primary tex on lateral HG and PT, the insula, and BA 22, the pre-
auditory cortex, medial and intermediate Heschl’s dominant response of the electrodes in the cortical areas
gyrus (HG); SAC, lateral HG and planum temporale was to the lowest AM frequencies (4–16 Hz) presented
(PT, posterior part of superior temporal gyrus (STG));
matching the range of AM frequencies crucial for speech
BA22 (Brodmann area 22), anterior STG, in front of
HG; HS, Heschl’s sulcus
intelligibility (e.g., Drullman et al. (1994)92,93 , Shannon
et al. (1995)94 , Smith et al. (2002)95 (see Fig. 17–16).168
Also, AEPs showing best synchronization to 16 Hz and
32 Hz appeared only in PAC, suggesting increased tem-
The HDE is stereotactically placed through the long poral resolution relative to more lateral areas and pos-
axis of either left or right HG, and consists of both macro- terior STG. These results corroborate those of an fMRI
contacts (impedance ∼5 k , 1.6 mm circumferential study on human subjects,104 which shows that regions
platinum discs spaced 10 mm apart) and microcontacts lateral and posterior to HG are more sensitive to the low
(impedance ∼0.08–0.2 M , consisting of 40-micrometer frequencies of 4 Hz and 8 Hz, and give support to the
exposed end wires cut flush with the electrode shaft probability of functional specialization of auditory areas.
and distributed at 1–2 mm intervals) (Howard, unpub-
lished data). Usually, the cortical surface array consists of
12 × 8 platinum–iridium disc electrodes embedded in a CONCORDANCE ACROSS METHODS
silicon membrane, with 5 mm center–center electrode
spacing (diameter 5 mm); references for all electrodes As is evident from the preceding sections, the use of var-
are at the vertex of the skull near the midline in con- ious methods to map the function of the auditory cor-
tact with the galea. Detailed intraoperative photographs, tex has largely been corroborative. As new techniques
postimplantation X-rays, and pre- and postimplantation emerge, they verify findings from older key studies and
3D MRIs are used together in localizing the grid and hone details, investigating parameters on a detail hereto-
depth electrode recording sites; coordinates of the depth fore unseen. The use of intracranial electrical stimulation
electrodes are determined by computer algorithm. and recording has been by far the most sensitive method
256 SECTION II SYSTEMS

Figure 17–14. (A) Auditory evoked potentials (AEPs) recorded from 14 microcontact
and 4 macrocontact sites along the length of Heschl’s gyrus (HG) of the left
hemisphere of one subject, in response to a click train (5 clicks at 100 Hz) (Brugge
and Howard, unpublished data). AEPs in left column were filtered from 1.6 to 1000 Hz,
those in right column from 70 to 1000 Hz. Asterisk marks two sites of maximal
amplitude of response, in posteromedial and anteromedial HG. Dashed line denotes
where a functional transition takes place in the sequence of AEPs. Drawings of
magnetic resonance imaging (MRI) cross-sections show the position of the recording
electrode (closed red circle) within the grey matter at the two representative
recording locations marked with the asterisk. Light red shading denotes the estimated
medio-lateral extent of HG. The electrode trajectory and location of each recording
site are shown on the surface rendering of the superior temporal plane. Open circles,
high-impedance contacts; closed circles, low-impedance contacts; ats, anterior
transverse sulcus. (B). All-pass AEPs recorded from the 96-contact grid on the
perisylvian cortex. The locations of the recording contacts are shown on the magnetic
resonance imaging rendering of the lateral surface of the cerebral hemisphere.
Asterisk marks the site of maximal amplitude of response. Expanded view shows the
AEPs recorded at each site. STG, superior temporal gyrus; SF, sylvian fissure.
CHAPTER 17 MAPPING OF HEARING 257

A E I

B F J

C G K

D H L

Figure 17–15. AEP waveforms obtained at the sites of maximal amplitude on


posteromedial (A–D) and anterolateral (E–G) Heschl’s gyrus and from posterolateral
superior temporal gyrus (I–L) plotted at different time scales, in reference to Figure
17–14 (Brugge and Howard, unpublished data). Filters: 1.6 to 1000 Hz for all but D, H,
L, for which a high-pass filter (70–1000 Hz) was employed. Latency (milliseconds) of
major deflections shown on figures. Note: different time scale than in Figure 17–14.

for defining eloquent auditory cortex and for differenti- method is gaining acceptance in its comparable results
ating various auditory fields. This invasive method has to intracranial stimulation and recording.
in general confirmed and refined the findings on EEG,
PET, and fMRI, but its invasive nature causes it to see
limited usage aside from presurgical candidates. 䉴 CLINICAL APPLICATIONS
As already mentioned, though, spectral analysis of
synchronized “gamma-range” oscillatory activity, more Direct cortical stimulation (electrical interference) is
than 30–40 Hz, is becoming more prominent as a means a well-established technique for mapping speech-
of studying and understanding auditory cortical physi- language and motor functions in patients who are
ology. Its presence in primate cortex,132,133 as well as surgical candidates for treatment of intractable seizures,
in human sensorimotor and visual cortex,169−174 seems tumors, or arteriovenous malformations.175−179 Origi-
to be indicative of “binding” spatially segregated neu- nally developed for intraoperative use to test for local-
rons into networks representing higher order stimulus ization of eloquent cortex,175,176 this technique induces
properties.130,131 As we shall see in the next section, this a low-level (10–15 mA) bipolar electrical current for
258 SECTION II SYSTEMS

A B

Figure 17–16. Spatial distribution of responses to amplitude modulation investigated in


the right (A) and left (B) auditory cortices in two patients. (Adapted from
Liégeois-Chauvel C, Lorenzi C, Trébuchon A, Régis J, Chauvel P. Temporal envelope
processing in the human left and right auditory cortices. Cereb Cortex 2004;14(7):
731–740). Thirty contacts in Case A, electrodes labeled T, H, and P, and 11 contacts in
Case B, electrodes labeled T and H (each white segment corresponding to a given
electrode contact). Each blue, yellow, red, or green line indicates which auditory
region is recorded and its anatomical location on the electrode superimposed on the
magnetic resonance imaging. Spectral magnitude is represented by a color code.
Note that a single electrode can record cortical activity from different areas, with
contacts numbered from medial to lateral. (A) Note the spatial segregation of corner
or best modulation frequencies (CMFs or BMFs) within primary auditory cortex (PAC):
BMFs of 8 Hz are observed in H3, BMFs of 32 Hz are observed in H4 and CMFs or
BMFs of 4, 8, and 16 Hz are observed in H5 (these leads are located in the anterior
PAC). The green arrow indicates that H6 passes through a sulcus between Heschl’s
gyrus and planum temporale. (B) BMFs of 16 Hz are observed in H13 and H14, while
CMFs of 4 Hz are observed in T11, T12, and T13.

2–3 seconds, using a 100-milliseconds pulse width be- electrode strip placed on adjacent cortex to the site of
tween pairs of electrodes located on the lateral surface of stimulation.
the cortex.33,148,150 The so-called “functional” lesion that We also routinely employ electrical stimulation
is induced is temporary (<5 seconds), discretely local- functional mapping (ESFM) extraoperatively to test for
ized to less than 1 cm2 , and highly reproducible.33,178−182 location of eloquent cortex in the language-dominant
To map language function in the awake patient intraop- hemisphere prior to planned resection in patients with
eratively, the stimulating current is carefully adjusted to medically intractable epilepsy. Most of these patients
the proper strength, using motor and/or sensory cortical have depth electrodes stereotactically implanted into bi-
stimulation induced movements or paresthesias as cues, lateral amygdala as well as into HG on the supratemporal
careful not to exceed afterdischarge threshold. This can plane, with grid electrodes implanted over perisylvian
be evaluated concurrently with the use of a four contact cortex of either the left or right hemisphere.21,33
CHAPTER 17 MAPPING OF HEARING 259

Other investigators may have variants on this tech- adjunct it is insufficient to be used alone as a tool to form
nique, but we usually implant a surface array of 12 × critical preresection decisions. In addition, as only three
8 platinum–iridium disc electrodes embedded in a sil- of eight patients examined with postresection fMRI cor-
icon membrane, with 5-mm center-to-center electrode roborated the language foci identified by intraoperative
spacing; electrode contact diameter is 1.5 mm. Strip elec- ECS, the authors contend that perhaps such low sensi-
trodes with 1-cm intercontact spacing are placed on the tivity and specificity can be improved by modifying the
mesial surface of the inferior temporal gyrus; reference behavioral paradigms employed. In contrast, in a study
electrodes are attached to the skull near the midline on patients with mesial temporal lobe epilepsy, Carpen-
in contact with the galea. Detailed intraoperative pho- tier et al.187 found good concordance in intrahemispheric
tographs, postimplantation X-rays, and pre- and postim- language maps generated by fMRI and intraoperative
plantation 3D MRIs are used together in localizing the ECS. Here, additional eloquent foci were identified by
grid and depth electrode recording sites. fMRI, which the authors attribute to limited cortical cov-
These patients usually remain on the epilepsy ward erage by electrodes and the inherent differences in the
for 10–14 days under video-EEG surveillance, where lan- techniques between fMRI (imaging) and ECS (electrical
guage mapping is performed just prior to resection. This interference).
is performed by presenting the patient with pictures of
common objects to name, while stimulating pairs of ad-
jacent electrodes on the lateral surface, searching for ACOUSTIC PROCESSING
sites within the field where deficits in speech occur. As
performed intraoperatively, afterdischarge threshold in Penfield and Rasmussen188 first reported that electrical
stimulation current is not exceeded by observing the pulses applied to the STG of patients undergoing awake
concurrently recorded EEG. craniotomies produced alterations in auditory percep-
Aside from electrical cortical stimulation (ECS), the tion, referred to as “auditory illusions.” Most of these
gold standard technique, there have been applications patients suffered from seizures and were undergoing
of noninvasive functional mapping modalities to identify exploratory stimulation for eloquent cortex. “Experien-
eloquent language cortex in presurgical planning. tial” responses were elicited from less than 10% patients
The Wada test,183 or the intracarotid injection of tested, ranging from “crude” auditory percepts such as a
sodium amobarbital, has been used to establish the lat- “tone, a buzzing or knocking sound” from HG stimula-
erality of language function in preoperative planning tion to “voices” or “someone calling” when lateral STG
for decades. Functional brain mapping using PET to was stimulated.189−192
measure regional cerebral blood flow (rCBF) has also These early records of objectively quantified per-
been used as a preoperative assessment of language ceptions confirmed the presence of a hierarchical audi-
function. Pardo and Fox184 used CBF PET in 11 pa- tory cortex on STG, with increased complexity of elicited
tients with complex partial epilepsy to detect language responses from laterally stimulated sites.
dominance and they found their results to be compa- In the course of performing ESFM in patients with
rable to those of Wada testing. Kaplan et al.185 used medically intractable epilepsy, we were also able to
2-deoxy-2-[18F]fluoro-D-glucose (FDG) and L-[methyl- evoke sound perceptions within posterior medial HG,
(11)C]methionine (CMET) PET scans coregistered with such as a “tone” or “chopping sound,” localized to the
MR images in five pediatric patients to accurately de- contralateral ear. In contrast, we also were able to in-
fine structural and functional cortical areas as well as to duce hearing suppression in both ears, which persisted
grade tumors, which was instrumental in achieving effec- beyond the period of electrical stimulation, during stimu-
tive neurosurgical planning. In this patient population, lation of sites within anterior lateral HG and posterior lat-
PET scanning was deemed advantageous over MRI as it eral STG, as rarely reported before.191 This suppression
provided less claustrophobic anxiety for the patient, tu- was perceived as a “muffling” sound, and in a patient
mor grading capability, and mapping in a single session. with long-standing tinnitus, caused it to “quiet down”
Although fMRI regimens demonstrate compara- (see Fenoy et al.150 ). This finding of hearing suppres-
ble results to Wada testing for language lateralization, sion due to stimulation of sites within known auditory
correlation between pre- and postoperative fMRI and cortical fields offers the suggestion of the presence of
intraoperative ECS remains inconsistent. Roux et al.186 corticofugal efferent pathways that project to auditory
compared language areas showing fMR activations by thalamic, midbrain and brainstem structures, and pos-
naming and verb generation tasks with intraoperative sibly the cochlea, as seen in other animals.193−199 The
speech mapping using ECS in 14 right-handed patients corticofugal pathways, which originate in different au-
with left hemisphere tumors. ditory cortical fields, including PAC on HG, may exert
Only 5 of 22 language sites identified by ECS were different functional influences on auditory processing;
associated with preoperative fMR activation, leading the subjective suppression of hearing induced by electrical
authors to conclude that although fMRI may be a helpful stimulation is one such effect.
260 SECTION II SYSTEMS

SPEECH AND LANGUAGE tion of the STG and other cortical regions that sup-
PROCESSING port speech perception is hierarchical, with more basic
speech processing (i.e., syllable discrimination) posteri-
By far, the most pertinent application of clinical brain orly based and larger network activation with increased
mapping in the realm of hearing is to identify and pre- complexity of speech processing, using different tempo-
serve human speech and language function. In recent ral lobe pathways extending both anteriorly and poste-
years, there has been a rise in the number of brain riorly. Such a view is consistent with results of primate
mapping studies using functional lesion methods to and human neuroimaging studies that suggest different
investigate the neural bases of speech perception. These transmission streams depending on stimulus and task
studies have been performed on neurosurgery patients demands.8,14,23,38,208
prior to planned resections of language-dominant tem- If by chance afterdischarges are evoked, these can
poral lobe. Boatman et al.200 reviewed these stud- evolve into clinical seizures if left unchecked.209,210 Af-
ies and summarized the individual functional circuitry terdischarges and seizures elicited during stimulation
activated for specific speech processing. Within left mapping do not accurately reflect the patient’s seizure
STG, electrocortical mapping studies identified a cir- focus.209
cumscribed anterior region within the middle-posterior Alternatively, Crone et al.129 contend that functional
left STG associated with acoustic–phonetic processing maps based on passive recordings of event-related ECoG
(as measured by auditory discrimination of phonetic gamma activity avoids the possibility of afterdischarges
features),179,182,200−204 anterior and posterior sites as- that may prevent comprehensive language mapping and
sociated with phonological processing, as assessed by shortens the time of acquisition, as a map can be ob-
phoneme identification (repetition, picture word match- tained for all implanted electrode sites at once. This vari-
ing, orthographic matching),179 and a wide cortical distri- ation on mapping using the same subdural electrode ar-
bution of sites associated with access to lexical–semantic rays improves upon limitations of functional imaging.
information (as measured by sentence comprehension), Whereas fMRI has been used preoperatively recently for
including the temporoparietal junction as well as anterior some functional mapping, it may not accurately predict
regions in the frontal lobe.176,179,182,205−207 The location eloquent areas186,211−213 ; the poor temporal resolution
of electrode sites where discrimination of both syllables caused by the time lag between synaptic activity and
and spectrally complex FM tones was impaired during changes in blood flow makes it unsuitable for monitor-
cortical stimulation is depicted in Fig. 17–17.202 ing language function.214
Boatman et al. suggest that, in accordance with Crone and colleagues have been in the process of
recent animal studies,9,23,38 the functional organiza- comparing the maps derived from event-related ECoG
gamma-band changes with maps obtained by electrical
stimulation mapping (ESM) through the same subdural
electrodes172,215 to evaluate their potential application to
functional brain mapping.
In previous studies, Crone et al.171 found event-
related synchronization (ERS) more than 30–40 Hz to
occur in a relatively focused spatial distribution over the
dominant superior gyrus, coinciding with the onset of
the N100 of the auditory EP but lasting longer, and occur-
ring with greater magnitude in association with speech
stimuli than with tone stimuli. This study analyzed over-
lapping frequency bands, finding, as in other studies, the
presence of activity well above 100 Hz. Edwards et al.216
in their study of deviant auditory stimuli detection, re-
Figure 17–17. Location of electrode sites where ported high-gamma oscillations in the frequency range
discrimination of consonant-vowel syllables and of 60–250 Hz, centered at approximately 100 Hz.
spectrally complex frequency-modulated (FM) tones Sinai et al.215 recently compared the spatial pat-
was impaired during cortical stimulation in 11 patients, terns of high-gamma (80–100 Hz) spectral changes
normalized to a standard brain representation. (See obtained during picture naming with ESM maps of nam-
Boatman DF. Cortical auditory systems: speech and ing in the same clinical subjects undergoing epilepsy
other complex sounds. Epilepsy Behav 2006;8(3):
surgery (see Fig. 17–18).215 By using the average number
494–503; Epub 2006. Review, 2006.). Discrimination
of pure tones, vowels, and simple FM sweeps of 12 electrode sites with statistically significant high-
remained intact at these sites. Light gray lines gamma ERS, electrocorticographic high-gamma ERS
represent sites where no auditory discrimination during confrontation naming appeared to predict ESM
deficits were induced. interference with naming and mouth-related motor
CHAPTER 17 MAPPING OF HEARING 261

200 200
Gamma Power Gamma Power

Percent Change

Percent Change
150 80–100 Hz 150 80–100 Hz

100 100

50 50

0 0
–50 –50
0.0 1.0 2.0 3.0 0.0 1.0 2.0 3.0
Time (seconds) Time (seconds)

200
Gamma Power
Percent Change

150 80–100 Hz

100

50

0
–50
0.0 1.0 2.0 3.0
Time (seconds) Electrical Cortical Stimulation
Mouth Naming
No Impairment Other Language

Figure 17–18. Comparison of event-related electocorticography (ECoG) high-gamma


activity with electrical stimulation mapping (ESM) in an individual subject. (See Sinai A,
Bowers CW, Crainiceanu CM, et al. Electrocorticographic high-gamma activity versus
electrical cortical stimulation mapping of naming. Brain 2005;128:1556–1570). White
circles denote electrode sites where ECoG was recorded. Yellow plots show the
magnitude of high-gamma activity as a percentage change (y-axis) with respect to
baseline. The onset of the pictured object to be named occurred 400 milliseconds
after disappearance of a fixation point at 0 seconds (x-axis). Colored bars join
electrode pairs where electrical cortical stimulation was performed, color-coded for
the occurrence and types of functional effects. Note that ESM was not performed at
some sites where ECoG was recorded (pain was encountered at some
temporal-occipital sites).

function with good specificity (84%) but relatively low between perisylvian cortical regions at multiple high-
sensitivity (43%). Such favorable specificity of high- gamma frequencies, especially bidirectional interactions
gamma ERS suggests that it could predict at which sites between STG and inferior frontal gyrus.
ESM would disrupt naming, and thus could possibly be Such studies using multichannel electrocorticogra-
useful in the construction of a functional map. However, phy as well as evaluating the role of high-gamma oscil-
due to the low sensitivity in the study, for sites within lations are likely in the future to provide much more
the planned cortical resection at which high-gamma ERS detailed information regarding dynamic interactions
was not observed, the authors agree that ESM would still across distributed cortical networks. Moreover, current
be necessary to protect eloquent cortex. The authors research is continually finding new applications for spec-
attribute the low sensitivity to perhaps too restricted tral analysis to uniquely investigate of brain function.
a frequency band of analysis, too wide electrode spac- In a recent study, Lachaux and colleagues218 used
ing (1 cm), or perhaps difficulty in the interpretation of real-time spectral analysis of brain signals in the post-
magnitude that is indicative of cortical processing.215 electrode implantation, preresection evaluation of two
To investigate whether causal interactions at high- patients with intractable epilepsy to find a highly specific
gamma frequencies belies functional cooperation of functional and spatial organization of high-gamma (60–
distributed cortical networks, SdDTF (short-time direct 140 Hz) power in the anterior superior temporal lobe
directed transfer function) has been used recently by associated with speech and music perception. These
Korzeniewska et al.217 to estimate event-related causal results complemented concurrent standard ESM, again
interactions between multiple electrocorticography sig- making this a promising tool in presurgical mapping.
nals during auditory word repetition and other speech Furthermore, using direct visual feedback, it shows po-
production tasks. Such interactions were seen to occur tential as an online exploratory tool that can lead to
262 SECTION II SYSTEMS

novel experimental paradigms that can be tailor de- 10. Kaas JH, Hackett TA. Subdivisions of auditory cortex and
signed to systematically probe the spatial, temporal, and processing streams in primates. Proc Natl Acad Sci U S A
spectral aspects of cognition. 2000;97:11793-11799.
11. Kosaki H, Hashikawa T, He J, Jones EG. Tonotopic or-
ganisation of auditory cortical fields delineated by parval-
bumin immunoreactivity in macaque monkeys. J Comput
䉴 PEARLS AND PITFALLS Neurol 1997;386:304-316.
12. Manley JA, Müller-Preuss P. Response variability of audi-
tory cortex cells in the squirrel monkey to constant acous-
There has been considerable scientific progress over the
tic stimuli. Exp Brain Res 1978;32(2):171-180.
past decade in the development of new technological
13. Recanzone GH, Guard DC, Phan ML. Frequency and
applications to elucidate our current thinking and un- intensity response properties of single neurons in the
derstanding of the human brain. These new applications, auditory cortex of the behaving macaque monkey. J Neu-
such as improved fMRI and MEG, and the spectral analy- rophysiol 2000;83:2315-2331.
sis of high-gamma activity, however, is most useful when 14. Rauschecker JP, Tian B. Mechanisms and streams for pro-
used together or in conjunction with preexisting “gold cessing of “what” and “where” in auditory cortex. Proc
standard” techniques, such as intracranial recording and Natl Acad Sci U S A 2000;97(22):11800-11806.
stimulation. It is of little doubt that less invasive imaging 15. Tian B, Reser D, Durham A, Kustov A, Rauschecker JP.
will hopefully one day supplant invasive recordings or Functional specialization in rhesus monkey auditory cor-
stimulations, but as we have reiterated, the most valuable tex. Science 2001;292:290-293.
16. Whitfield IC, Evans EF. Responses of auditory cortical neu-
information has been yielded by a combination of tech-
rons to stimuli of changing frequency. J Neurophysiol
niques, so as to make up for the disadvantages of any
1965;28:655-672.
one particular modality applied individually. The com- 17. Romani GL, Williamson SJ, Kaufman L. Tonotopic orga-
plexity of function of the human brain is not fully known, nization of the human auditory cortex. Science 1982;216:
and as each human has a specific, unique cortical map 1339-1340.
designated for audition, speech, and language, we must 18. Romani GL, Williamson SJ, Kaufman L, et al. Characteriza-
still proceed cautiously in presurgical mapping to ensure tion of the human auditory cortex by the neuromagnetic
that eloquent cortex is fully detailed. It shall be inter- method. Exp Brain Res 1982;47:381-393.
esting to see what developments in research occur in 19. Huotilainen M, Tiitinen H, Lavikainen J, et al. Sustained
the next decade to augment our artillery of investigative fields of tones and glides reflect tonotopy of the auditory-
tools. cortex. Neuro Report 1995;6:841-844.
20. Pantev C, Eulitz C, Elbert T, Hoke M. The auditory evoked
sustained field: origin and frequency dependence. Elec-
troenc Clin Neurophysiol 1994;90(1):82-90.
REFERENCES 21. Howard MA, Volkov IO, Abbas PJ, Damasio H, Ollendieck
MC, Granner MA. A chronic microelectrode investigation
1. Ferrier D. The Functions of the Brain. London: Smith El- of the tonotopic organization of human auditory cortex.
der, 1876, pp. 171-175. Brain Res 1996;724:260-264.
2. Flechsig P. Bemerkungen über die Hörsphäre des men- 22. Kaas JH, Hackett TA. Subdivisions of auditory cortex
schlichen Gehirns. Neurol Zentralbl 1908;27(2):2-7. and levels of processing in primates. Audiol Neurootol
3. Brodmann K. Vergleichende Lokalisationslehre der 1998;3:73-85.
Grosshirnrinde. Barth: Leipzig, 1909. 23. Kaas JH, Hackett TA, Tramo MJ. Auditory processing in
4. Merzenich MM, Brugge JF. Representation of the cochlear primate cerebral cortex. Curr Opin Neurobiol 1999;9:164-
partition of the superior temporal plane of the macaque 170.
monkey. Brain Res 1973;50:275-296. 24. Hackett TA, Stepniewska I, Kaas JH. Subdivisions of au-
5. Fitzpatrick KA, Imig TJ. Auditory cortico-cortical connec- ditory cortex and ipsilateral cortical connections of the
tions in the owl monkey. J Comp Neurol 1980;192:589- parabelt auditory cortex in Macaque monkeys. Brain Res
610. 1999;817:45-58.
6. Morel A, Kaas JH. Subdivisions and connections of audi- 25. Rademacher J, Morosan P, Schormann T, et al. Probabilis-
tory cortex in owl monkeys. J Comp Neurol 1992;318:27- tic mapping and volume measurement of human primary
63. auditory cortex. Neuroimage 2001;13:669-683.
7. Morel A, Garraghty PE, Kaas JH. Tonotopic organization, 26. Hackett TA. The comparative anatomy of the primate au-
architectonic fields, and connections of auditory cortex in ditory cortex. In Primate Audition: Ethology and Neuro-
macaque monkeys. J Comp Neurol 1993;335:437-459. biology, edited by AA Ghazanfar. Boca Raton: CRC Press,
8. Rauschecker JP, Tian B, Pons T, Mishkin M. Serial and 2003, pp. 199-219.
parallel processing in rhesus monkey auditory cortex. 27. Galaburda AM, Sanides F. Cytoarchitectonic organiza-
J Comp Neurol 1997;382:89-103. tion of the human auditory cortex. J Comp Neurol
9. Rauschecker JP, Tian B, Hauser M. Processing of com- 1980;190:597-610.
plex sounds in the macaque nonprimary auditory cortex. 28. Morosan P, Rademacher J, Schleicher A, Amunts K,
Science 1995;268:111-114. Schormann T, Zilles K. Human primary auditory cortex:
CHAPTER 17 MAPPING OF HEARING 263

cytoarchitectonic subdivisions and mapping into a spatial 47. Baumgart F, Gaschler-Markefski B, Worldorff MG, Heinze
reference system. Neuroimage 2001;13:684-701. HJ, Scheich H. A movement sensitive area in auditory cor-
29. Fullerton BC, Pandya DN. Architectonic analysis of the tex. Nature 1999;400:724-726.
auditory-related areas of the superior temporal region in 48. Krumbholz K, Schönwiesner M, Rübsamen R, Zilles K,
human brain. J Comp Neurol 2007;504:470-498. Fink GR, von Cramon DY. Hierarchical processing of
30. Wallace MN, Johnston PW, Palmer AR. Histochem- sound location and motion in the human brainstem
ical identification of cortical areas in the auditory and planum temporale. Eur J Neurosci 2005;21(1):230-
region of the human brain. Exp Brain Res 2002;143:499- 238.
508. 49. Belin P, Zatorre RJ. ‘What’, ‘where’ and ‘how’ in auditory
31. Rivier F, Clarke S. Cytochrome oxidase, acetyl- cortex. Nat Neurosci 2000;3:965-966.
cholinesterase, and NADPH-diaphorase staining in 50. Scott SK, Blank CC, Rosen S, Wise RJ. Identification of a
human supratemporal and insular cortex: evidence for pathway for intelligible speech in the left temporal lobe.
multiple auditory areas. Neuroimage 1997;6:288-304. Brain 2000;123(12):2400-2406.
32. Hackett TA, Preuss TM, Kaas JH. Architectonic identifica- 51. Tramo MJ, Cariani PA, Koh CK, Makris N, Braida LD. Neu-
tion of the core region in auditory cortex of macaques, rophysiology and neuroanatomy of pitch perception: au-
chimpanzees, and humans. J Comp Neurol 2001;441:197- ditory cortex. Ann N Y Acad Sci 2005;1060:148-174.
222. 52. Rhoton AL Jr. The supratentorial cranial space: micro-
33. Howard MA, Volkov IO, Mirsky R, et al. Auditory cortex on surgical anatomy and surgical approaches. Neurosurgery
the posterior superior temporal gyrus of human cerebral 2003;53(Suppl, Part 2):39.
cortex. J Comp Neurol 2000;416:76-92. 53. von Economo C, Horn L. Uber windungsrelief mabe
34. Rademacher J, Caviness V, Steinmetz H, Galaburda AM. und Rindenarchitektonic der supratemparalflache, ihre
Topographical variation of the human primary cortices: individuellen und seitenunterschiede. Neuropsychiatre
implications for neuroimaging, brain mapping and neu- 1930;30(2):678-757.
robiology. Cereb Cortex 1993;3:313-329. 54. Pfeifer RA. Mylogenetisch—Anatomische Untersuchun-
35. von Economo C, Koskinas GN. Die Cytoarchitektonik der gen Uber das Korticale Ende Untersuchungen end der
Hirnrinde des erwachsenen Menschen. Berlin: Springer, Heorleitung. Berlin: Acad Wiss, 1920, p. 37.
1925. 55. Pfeifer RA. Pathologie der Hoerstrahlung und der corti-
36. Sweet RA, Dorph-Petersen KA, Lewis DA. Mapping audi- calen Hoesphare. In Handbuch der Neurologie, edited by
tory core, lateral belt, and parabelt cortices in the human O Bumke und O Foerster. Berlin: Springer Verlag, 1936,
superior temporal gyrus. J Comp Neurol 2005;491:270- pp. 523-626.
289. 56. Beck E. Die myeloarchitektoniscke felderung des in
37. Rauschecker JP. Parallel processing in the auditory cortex der sylvischen furche gelegenen teils des menschlichen
of primates. Audiol Neurootol 1998;3:86-103. schlafenlappens. J Psychol Neurol 1928;36:1-21.
38. Romanski LM, Bates JF, Goldman-Rakic PS. Auditory 57. Beck E. Typologie des gehirns am beispiel des dorsalen
belt and parabelt projections to the prefrontal cortex menschlichen schlafenappens nebst weiteren beitrage
in the rhesus monkey. J Comp Neurol 1999;403:141- zur frage der links-rechtshirnigkeit. Dtsch Z Nervenheilk
157. 1955;173:267-308.
39. Wessinger CM, VanMeter J, Tian B, Van Lare J, Pekar 58. Campain R, Minckler JA. Note on the gross configurations
J, Rauschecker JP. Hierarchical organization of the hu- of the human auditory cortex. Brain Lang 1976;3:318-
man auditory cortex revealed by functional magnetic res- 323.
onance imaging. J Cogn Neurosci 2001;13:1-7. 59. Broca PM. Perte de la porole, ramolissment chronique
40. Griffiths TD, Warren JD, Scott SK, Nelken I, King AJ. Cor- et destruction partiele de lobe anterieur gauches de
tical processing of complex sound: a way forward? Trends cerveaux. Bull Soc Anthropol Paris 1861;2:235-238.
Neurosci 2004;27:181-185. 60. Wernicke C. Der Ophasische Symptomenkomplex. Eine
41. Hickok G, Poeppel D. Dorsal and ventral streams: a Psychologische Studie Auf Anatomische Basis. Bresiam:
framework for understanding aspects of the functional Max Cohn und Weigert, 1874.
anatomy of language. Cognition 2004;92:67-99. 61. Cunnigham DJ. Contribution to the Surface Anatomy of
42. Hickok G, Poeppel D. The cortical organization of speech the Cerebral Hemispheres. Dublin: Royal Irish Academy,
processing. Nat Rev Neurosci 2007;8:393-402. 1892.
43. Arnott SR, Binns MA, Grady CL, Alain C. Assessing the 62. Shapleske J, Rossell SL, Woodruff PWR, David AS. The
auditory dual-pathway model in humans. Neuroimage planum temporale: a systematic, quantitative review of its
2004;22(1):401-408. structural, functional and clinical significance. Brain Re-
44. Griffiths TD, Rees A, Witton C, et al. Evidence for a sound search Reviews 1999;29:26-49.
movement center in the human cerebral cortex. Nature 63. Hiwatashi A, Zhong J, Ekholm SE, Westesson PL. Frac-
1996;383:425-427. tional anisotropy is higher in Heschl’s gyrus than in su-
45. Alain C, Arnott SR, Hevenor S, Graham S, Grady CL. ‘What’ perior temporal gyrus in normal subjects. Acad Radiol
and ‘where’ in the human auditory system. Proc Natl Acad 2006;13(1):73-76.
Sci U S A 2001;98:12301-12306. 64. Upadhyay J, Ducros M, Knaus TA, et al. Function and
46. Maeder PP, Meuli RA, Adriani M, et al. Distinct pathways connectivity in human primary auditory cortex: a com-
involved in sound recognition and localization: a human bined fMRI and DTI study at 3 Tesla. Cereb Cortex
fMRI study. NeuroImage 2001;14:802-816. 2007;17(10):2420-2432.
264 SECTION II SYSTEMS

65. Tian B, Rauschecker JP. Processing of frequency- hemispheric comparison of primary auditory cortex in the
modulated sounds in the lateral auditory belt cortex of common marmoset (Callithrix jacchus). J Comp Neurol
the rhesus monkey. J Neurophysiol 2004;92:2993-3013. 2005;487(4):391-406.
66. Hall DA, Hart HC, Johnsrude IS. Relationships between 82. Talavage TM, Ledden PJ, Benson RR, Rosen BR, Melcher
human auditory cortical structure and function. [Review]. JR. Frequency-dependent responses exhibited by multiple
Audiol Neurootol 2003;8(1):1-18. regions in human auditory cortex. Hear Res 2000;150:225-
67. Lauter JL, Herscovitch P, Formby C, Raichle ME. Tono- 244.
topic organisation in human auditory cortex revealed by 83. Liegeois-Chauvel C, Musolino A, Chauvel P. Localization
positron emission tomography. Hear Res 1985;20:199-205. of the primary auditory area in man. Brain 1991;114:139-
68. Pantev C, Bertrand O, Eulitz C, et al. Specific tonotopic or- 153.
ganisations of different areas of the human auditory cortex 84. Liégeois-Chauvel C, Musolino A, Badier JM. Evoked po-
revealed by simultaneous magnetic and electric record- tentials recorded from the auditory cortex in man: evalu-
ings. Electroenceph Clin Neurophysiol 1995;94:26-40. ation and topography of the middle latency components.
69. Pantev C, Hoke M, Lehnertz K, Lütkenhöner B, Electroencephalogr Clin Neurophysiol 2004;92(3):204-
Anogianakis G, Wittkowski W. Tonotopic organization of 214.
the human auditory cortex revealed by transient auditory 85. Hall DA, Johnsrude IS, Haggard MP, Palmer AR, Akeroyd
evoked magnetic fields. Electroenceph Clin Neurophysiol MA, Summerfield AQ. Spectral and temporal process-
1988;69:160-170. ing in human auditory cortex. Cereb Cortex 2002;12:
70. Tiitinen H, Alho K, Huotilainen M, Ilmoniemi RJ, Simola 140-149.
J, Näätänen R. Tonotopic auditory cortex and the mag- 86. Brechmann A, Baumgart F, Scheich H. Sound-level-
netoencephalographic (MEG) equivalent of the mismatch dependent representation of frequency modulations in
negativity. Psychophysiology 1993;30(5):537-540. human auditory cortex: a low-noise fMRI study. J Neu-
71. Verkindt C, Bertrand O, Perrin F, Echallier JF, Pernier rophysiol 2002;87:423-433.
J. Tonotopic organization of the human auditory cor- 87. Jäncke L, Shah NJ, Posse S, Grosse-Ryuken M, Müller-
tex: N100 topography and multiple dipole model analy- Gärtner HW. Intensity coding of auditory stimuli: an fMRI
sis. Electroencephalogr Clin Neurophysiol 1995;96(2):143- study. Neuropsychologia 1998;36:875-883.
156. 88. Mohr CM, King WM, Freeman AJ, Briggs RW, Leonard
72. Wessinger CM, Buonocore MH, Kussmaul CL, Mangun CM. Influence of speech stimuli intensity on the activation
GR. Tonotopy in human auditory cortex examined with of auditory cortex investigated with functional magnetic
functional magnetic resonance imaging. Hum Brain Mapp resonance imaging. J Acoust Soc Am 1999;105:2738-2745.
1997;5:18-25. 89. Hart HC, Palmer AR, Hall DA. Heschl’s gyrus is more sen-
73. Bilecen D, Scheffler K, Schmid N, Tschopp K, Seelig J. sitive to sound level than non-primary auditory cortex.
Tonotopic organization of the human auditory cortex as Hearing Res 2002;171:177-190.
detected by BOLD-FMRI. Hear Res 1998;126:19-27. 90. Rosen S. Temporal information is speech: acoustic, audi-
74. Hoke ES, Ross B, Hoke M. Auditory afterimage: tono- tory, and linguistic aspects. Phil Trans Roy Soc Lond B
topic representation in the auditory cortex. Neuroreport 1992;336:367-373.
1998;9(13):3065-3068. 91. Hall DA, Barrett DJ, Akeroyd MA, Summerfield AQ. Corti-
75. Rosburg T, Kreitschmann-Andermahr I, Emmerich E, cal representations of temporal structure in sound. J Neu-
Nowak H, Sauer H. Hemispheric differences in fre- rophysiol 2005;94(5):3181-3191.
quency dependent dipole orientation of the human au- 92. Drullman R, Festen JM, Plomp R. Effect of temporal en-
ditory evoked field component N100m. Neurosci Lett velope smearing on speech reception. J Acoust Soc Am
1998;258(2):105-108. 1994;95:1053-1064.
76. Lockwood AH, Salvi RJ, Coad ML, et al. The functional 93. Drullman R, Festen JM, Plomp R. Effect of reducing slow
anatomy of the normal human auditory system: responses temporal modulations on speech reception. J Acoust Soc
to 0.5 and 4.0 kHz tones at varied intensities. Cereb Cortex Am 1994;95:2670-2680.
1999;9:65-76. 94. Shannon RV, Zeng F, Kamath V, Wygonski J, Ekelid M.
77. Engelien A, Yang Y, Engelien W, Zonana J, Stern E, Sil- Speech recognition with primarily temporal cues. Science
bersweig DA. Physiological mapping of human auditory 1995;270:303-304.
cortices with a silent event-related fMRI technique. Neu- 95. Smith ZM, Delgutte B, Oxenham AJ. Chimaeric sounds
roimage 2002;16(4):944-953. reveal dichotomies in auditory perception. Nature 416:87-
78. Schönwiesner M, von Cramon YD, Rübsamen R. Is it tono- 90, 2002.
topy after all? NeuroImage 2002;17:1144-1161. 96. Griffiths TD, Buchel C, Frackowiak RSJ, Patterson RD.
79. Formisano E, Kim DS, Di Salle F, van de Moortele PF, Analysis of temporal structure in sound by the human
Ugurbil K, Goebel R. Mirror-symmetric tonotopic maps in brain. Nat Neurosci 1998;1:422-427.
human primary auditory cortex. Neuron 2003;40:59-69. 97. Griffiths TD, Uppenkamp S, Johnsrude I, Josephs O, Pat-
80. Talavage TM, Sereno MI, Melcher JR, Ledden PJ, Rosen terson RD. Encoding of the temporal regularity of sound
BR, Dale AM. Tonotopic organization in human auditory in the human brainstem. Nat Neurosci 2001;4:633-637.
cortex revealed by progressions of frequency sensitivity. 98. Krumbholz K, Patterson RD, Seither-Preisler A, Lammert-
J Neurophysiol 2004;91:1282-1296. mann C, Lütkenhöner B. Neuromagnetic evidence for a
81. Philibert B, Beitel RE, Nagarajan SS, Bonham BH, pitch processing center in Heschl’s gyrus. Cereb Cortex
Schreiner CE, Cheung SW. Functional organization and 2003;13:765-772.
CHAPTER 17 MAPPING OF HEARING 265

99. Patterson RD, Uppenkamp S, Johnsrude I, Griffiths TD. 117. Palomaki KJ, Tiitinen H, Mäkinen V, May P, Alku P.
The processing of temporal pitch and melody information Cortical processing of speech sounds and their ana-
in auditory cortex. Neuron 2002;36:767-776. logues in a spatial auditory environment. Cogn Brain Res
100. Penagos H, Melcher JR, Oxenham AJ. A neural representa- 2002;14:294-299.
tion of pitch salience in nonprimary human auditory cor- 118. Brunetti M, Belardinelli P, Caulo M, et al. Human brain
tex revealed with functional magnetic resonance imaging. activation during passive listening to sounds from differ-
J Neurosci 2004;24:6810-6815. ent locations: an fMRI and MEG study. Hum Brain Mapp
101. Warren JD, Griffiths TD. Distinct mechanisms for process- 2005;26(4):251-261.
ing spatial sequences and pitch sequences in the human 119. Zatorre RJ, Bouffard M, Ahad P, Belin P. Where is ‘where’
auditory brain. J Neurosci 2003;23:5799-5804. in the human auditory cortex? Nat Neurosci 2002;5:905-
102. Budd TW, Hall DA, Goncalves MS, et al. Binaural spe- 909.
cialisation in human auditory cortex: an fMRI investi- 120. Yamamoto T, Uemura T, Llinas R. Tonotopic organiza-
gation of interaural correlation sensitivity. Neuroimage tion of human auditory cortex revealed by multi-channel
2003;20:1783-1794. SQUID system. Acta Otolaryngol 1992;112:201-204.
103. Krumbholz K, Schonwiesner M, Yves von Cramon D, et al. 121. Lütkenhöner B, Steinsträter O. High-precision neuromag-
Representation of interaural temporal information from netic study of the functional organization of the human
left and right auditory space in the human planum tem- auditory cortex. Audiol Neurootol 1998;3:191-213.
poral and inferior parietal lobe. Cereb Cortex 2005;15:317- 122. Lütkenhöner B, Lammertmann C, Ross B, et al. Tonotopic
324. organization of the human auditory cortex revisited: high-
104. Giraud AL, Lorenzi C, Ashburner J, et al. Representation precision neuromagnetic studies. In Proceedings of the
of the temporal envelope of sounds in the human brain. 12th International Symposium on Hearing. Physiological
J Neurophysiol 2000;84:1588-1598. and Psychophysical Bases of Auditory Function, edited
105. Zatorre RJ, Belin P. Spectral and temporal processing in by DJ Breebaart, AJM Houtsma, A Kohlrausch, VF Prijs, R
human auditory cortex. Cereb Cortex 2001;11:946-953. Schoonhoven. Maastricht: Shaker, 2001, pp. 129-136.
106. Griffiths TD, Rees G, Rees A, et al. Right parietal cortex 123. Rosburg T, Kreitschmann-Andermahr I, Ugur T, et al.
is involved in the perception of sound movement in hu- Tonotopy of the auditory-evoked field component N100m
mans. Nat Neurosci 1998;1:74-79. in patients with schizophrenia. J Psychophysiol 2000;14:
107. Weeks R, Aziz-Sultan A, Bushara KO, et al. A PET 131-141.
study of human auditory spatial processing. Neurosci Lett 124. Beagley H, Knight J. Changes in auditory evoked response
1999;262:155-158. with intensity. J Laryngol Otol 1967;81:861-873.
108. Zatorre RJ, Belin P, Penhune VB. Structure and function 125. Rapin I, Schimmel H, Tourk L, Krasnegor NA, Pollak C.
of auditory cortex: music and speech. Trends Cogn Sci Evoked responses to clicks and tones of varying intensity
2002;6:37-46. in waking adults. Electroencephalogr Clin Neurophysiol
109. Bremmer F, Schlack A, Shah NJ, et al. Polymodal motion 1966;21:335-344.
processing in posterior parietal and premotor cortex: a 126. Gutschalk A, Patterson RD, Rupp A, Uppenkamp S, Scherg
human fMRI study strongly implies equivalencies between M. Sustained magnetic fields reveal separate sites for
humans and monkeys. Neuron 2001;29:287-296. sound level and temporal regularity in human auditory
110. Lewis JW, Beauchamp MS, Deyoe EA. A comparison of cortex. Neuroimage 2002;15:207-216.
visual and auditory motion processing in human cerebral 127. Xiang J, Chuang S, Wilson D, et al. Sound motion evoked
cortex. Cereb Cortex 2000;10:873-888. magnetic fields. Clin Neurophys 2002;113:1-9.
111. Griffiths TD, Green GGR. Cortical activation during per- 128. Ducommun CY, Murray MM, Thut G, et al. Segregated
ception of a rotating wide-field acoustic stimulus. Neu- processing of auditory motion and auditory location: an
roimage 1999;10:84-90. ERP mapping study. Neuroimage 2002;16:76-88.
112. Kaiser J, Lutzenberger W, Preissl H, Ackermann H, Bir- 129. Crone NE, Sinai A, Korzeniewska A. High-frequency
baumer N. Right-hemisphere dominance for the pro- gamma oscillations and human brain mapping with elec-
cessing of sound-source lateralization. J Neurosci 2000; trocorticography. Prog Brain Res 2006;159:275-295.
20:6631-6639. 130. Singer W, Gray CM. Visual feature integration and the
113. Kaiser J, Lutzenberger W. Location changes enhance temporal correlation hypothesis. Annu Rev Neurosci
hemispheric asymmetry of magnetic fields evoked by lat- 1995;18:555-586.
eralized sounds in humans. Neurosci Lett 2001;314(1-2): 131. Von der Malsburg C. Binding in models of perception and
17-20. brain function. Curr Opin Neurobiol 1995;5:520-526.
114. Bushara KO, Weeks RA, Ishii K, et al. Modality-specific 132. Murthy VN, Fetz EE. Coherent 25- to 35-Hz oscillations
frontal and parietal areas for auditory and visual spatial in the sensorimotor cortex of awake behaving monkeys.
localization in humans. Nat Neurosci 1999;2:759-766. Proc Natl Acad Sci U S A 1992;89:5670-5674.
115. Woldorff MG, Tempelmann C, Fell J, et al. Lateralized 133. Sanes JN, Donoghue JP. Oscillations in local field poten-
auditory spatial perception and the contralaterality of tials of the primate motor cortex during voluntary move-
cortical processing as studied with functional magnetic ment. Proc Nat Acad Sci U S A 1993;90:4470-4474.
resonance imaging and magnetoencephalography. Hum 134. Kaiser J, Lutzenberger W, Ackermann H, Birbaumer N.
Brain Mapp 1999;7:49-66. Dynamics of gamma-band activity induced by auditory
116. Zatorre RJ, Penhune VB. Spatial localization after excision pattern changes in humans. Cereb Cortex 2002;12:212-
of human auditory cortex. J Neurosci 2001;21:6321-6328. 221.
266 SECTION II SYSTEMS

135. Pfurtscheller G, Cooper R. Frequency dependence of the nectivity between auditory cortical areas revealed by
transmission of the EEG from cortex to scalp. Electroen- amplitude modulation sound processing. Cereb Cortex
cephalogr Clin Neurophysiol 1975;38:93-96. 2007;17(2):304-313.
136. Akay M, Daubenspeck JA. Investigating the contami- 153. Sem-Jacobsen CW, Petersen MC, Dodge HW Jr, Lazarte
nation of electroencephalograms by facial muscle elec- JA, Holman CB. Electroencephalographic rhythms from
tromyographic activity using matching pursuit. Brain Lang the depths of the parietal, occipital and temporal lobes in
1999;66:184-200. man. EEG Clin Neurophysiol 1956;8:263-278.
137. Loring DW, Sheer DE. Laterality of 40 Hz EEG and 154. Celesia GG. Organization of auditory cortical areas in
EMG during cognitive performance. Psychophysiology man. Brain 1976;99:403-414.
1984;21:34-38. 155. Celesia GG, Puletti F. Auditory cortical areas of man. Neu-
138. Binder JR, Frost JA, Hammeke TA, et al. Human temporal rology 1969;19:211-220.
lobe activation by speech and nonspeech sounds. Cereb 156. Celesia GG, Puletti F. Auditory input to the human cor-
Cortex 2000;10:512-528. tex during states of drowsiness and surgical anesthesia.
139. Tardif E, Clarke S. Intrinsic connectivity of human au- Electroencephalogr Clin Neurophysiol 1971;31:603-609.
ditory areas: a tracing study with DiI. Eur J Neurosci 157. Celesia GG, Broughton RJ, Rasmussen T, Branch C. Audi-
2001;13(5):1045-1050. tory evoked responses from the exposed human cortex.
140. Galuske RA, Schlote W, Bratzke H, Singer W. Interhemi- Electroencephalogr Clin Neurophysiol 1968;24:458-466.
spheric asymmetries of the modular structure in human 158. Chatrian GE, Petersen MC, Lazarte JA. Responses to
temporal cortex. Science 2000;289:1946-1949. clicks from the human brain: some depth electro-
141. Galuske RAW, Schuhmann A, Schlote W, et al. Interareal graphic observations. Electroencephalogr Clin Neuro-
connections in the human auditory cortex. Neuroimage physiol 1960;12:479-489.
1999;9:S994. 159. Puletti F, Celesia GG. Functional properties of the primary
142. Ades HW. Functional relationship between the middle cortical auditory area in man. J Neurosurg 1970;32:244-
and posterior ectosylvian areas in the cat. Am J Physiol 247.
1949;159:561. 160. Lee YS, Lueders H, Dinner DS, Lesser RP, Hahn J, Klem
143. Bignall KE. Bilateral temporofrontal projections in the G. Recording of auditory evoked potentials in man using
squirrel monkey: origin, distribution and pathways. Brain chronic subdural electrodes. Brain 1984;107:115-131.
Res 1969;13:319-327. 161. Steinschneider M, Volkov IO, Noh MD, Garell PC, Howard
144. Bignall KE, Imbert M. Polysensory and cortico-cortical MA 3rd. Temporal encoding of the voice onset time
projections to frontal lobe of squirrel and rhesus monkeys. (VOT) phonetic parameter by field potentials recorded
Electroencephalogr Clin Neurophysiol 1969;26:206-215. directly from human auditory cortex. J Neurophysiol
145. Bremer F, Bonnet V, Terzuolo C. Etude electrophysiolo- 1999;82(5):2346-2357.
giaque des aires auditives corticales chez le chat. Arch Int 162. Steinschneider M, Volkov IO, Fishman YI, Oya H, Arezzo
Physiol 1954;62:390-428. JC, Howard MA 3rd. Intracortical responses in human and
146. Downman CB, Woolsey CN, Lende RA. Auditory areas monkey primary auditory cortex support a temporal pro-
I, II and Ep: cochlear representation, afferent paths and cessing mechanism for encoding of the voice onset time
interconnections. Bull Johns Hopkins Hosp 1960;106:127- phonetic parameter. Cereb Cortex 2005;15:170-186.
142. 163. Fishman YI, Volkov IO, Noh MD, et al. Consonance and
147. Imbert M, Bignall KE, Buser P. Neocortical interconnec- dissonance of musical chords: neural correlates in au-
tions in the cat. J Neurophysiol 1966;29:382-395. ditory cortex of monkeys and humans. J Neurophysiol
148. Brugge JF, Volkov IO, Garell PC, Reale RA, Howard MA 2001;86:2761-2788.
3rd. Functional connections between auditory cortex on 164. Yvert B, Fischer C, Bertrand O, Pernier J. Localization of
Heschl’s gyrus and on the lateral superior temporal gyrus human supratemporal auditory areas from intracerebral
in humans. J Neurophysiol 2003;90:3750-3763. auditory evoked potentials using distributed source mod-
149. Brugge JF, Volkov IO, Reale RA, et al. The posterolateral els. Neuroimage 2005;28:140-153.
superior temporal auditory field in humans. Functional 165. Yvert B, Fischer C, Guenot M, Krolak-Salmon P, Isnard
organization and connectivity. In The Auditory Cortex— J, Pernier J. Simultaneous intracerebral EEG recordings of
Toward a Synthesis of Human and Animal Research, early auditory thalamic and cortical activity in human. Eur
edited by R Konig, P Heil, E Budinger, H Scheich. Toyko: J Neurosci 2002;16:1146-1150.
Erlbaum, 2005, pp. 145-162. 166. Howard MA, Volkov IO, Granner MA, Damasio HM, Ol-
150. Fenoy AJ, Severson MA, Volkov IO, Brugge JF, Howard lendieck MC, Bakken HE. A hybrid clinical-research depth
MA 3rd. Hearing suppression induced by electri- electrode for acute and chronic in vivo microelectrode
cal stimulation of human auditory cortex. Brain Res recording of human brain neurons [Technical note]. J Neu-
2006;1118(1):75-83. rosurg 1996;84:129-132.
151. Saito Y, Harashima H. Tracking of information within mul- 167. Godey B, Schwartz D, de Graaf JB, Chauvel P, Liégeois-
tichannel EEG record-causal analysis in EEG. In Recent Chauvel C. Neuromagnetic source localization of auditory
Advances in EEG and EMG Data Processing, edited by evoked fields and intracerebral evoked potentials: a com-
N Yamaguchi and K Fujisawa. Amsterdam: Elsevier, 1981, parison of data in the same patients. Clin Neurophysiol
pp. 133-146. 2001;112:1850-1859.
152. Guéguin M, Le Bouquin-Jeannés R, Faucon G, Chau- 168. Liégeois-Chauvel C, Lorenzi C, Trébuchon A, Régis J,
vel P, Liégeois-Chauvel C. Evidence of functional con- Chauvel P. Temporal envelope processing in the human
CHAPTER 17 MAPPING OF HEARING 267

left and right auditory cortices. Cereb Cortex 2004;14(7): 186. Roux FE, Boulanouar K, Lotterie JA, Mejdoubi M, LeSage
731-740. JP, Berry I. Language functional magnetic resonance imag-
169. Crone NE, Hao L. Functional dynamics of spoken and ing in preoperative assessment of language areas: cor-
signed word production: a case study using electrocor- relation with direct cortical stimulation. Neurosurgery
ticographic spectral analysis. Aphasiology 2002;16:903- 2003;52:1335-1347.
926. 187. Carpentier A, Pugh KR, Westerveld M, et al. Functional
170. Crone NE, Hao L, Hart J Jr, et al. Electrocorticographic MRI of language processing: dependence on input modal-
gamma activity during word production in spoken and ity and temporal lobe epilepsy. Epilepsia 2001;42:1241-
sign language. Neurology 2001;57(11):2045-2053. 1254.
171. Crone NE, Boatman D, Gordon B, Hao L. Induced electro- 188. Penfield W, Rasmussen T. The Cerebral Cortex of Man—
corticographic gamma activity during auditory perception. A Clinical Study of Localization of Function. New York:
Clin Neurophysiol 2001;112(4):565-582. MacMillan, 1950.
172. Crone NE, Miglioretti DL, Gordon B, et al. Functional map- 189. Penfield W. The Excitable Cortex in Conscious Man. Liv-
ping of human sensorimotor cortex with electrocortico- erpool: Liverpool Press, 1958.
graphic spectral analysis. I. Alpha and beta event-related 190. Penfield W, Jasper H. Epilepsy and the Functional
desynchronization. Brain 1998;121(Pt 12):2271-2299. Anatomy of the Human Brain. Boston: Little Brown, 1954.
173. Crone NE, Miglioretti DL, Gordon B, et al. Functional map- 191. Penfield W, Perot P. The brain’s record of auditory and
ping of human sensorimotor cortex with electrocortico- visual experience: a final summary and discussion. Brain
graphic spectral analysis. II. Event-related synchronization 1963;86:595-696.
in the gamma band. Brain 1998;121(Pt 12):2301-2315. 192. Mullen S, Penfield W. Illusions of comparative interpre-
174. Pfurtscheller G, Graimann B, Huggins JE, Levine SP, Schuh tation and emotion. Arch Neurol Psychiatry 1959;81:269-
LA. Spatiotemporal patterns of beta desynchronization 284.
and gamma synchronization in corticographic data during 193. Hazama M, Kimura A, Donishi T, Sakoda T, Tamai Y.
self-paced movement. Clin Neurophysiol 2003;114:1226- Topography of corticothalamic projections from auditory
1236. cortex of the rat. Neuroscience 2004;124:655-667.
175. Penfield W, Roberts L. Speech and brain mechanisms. 194. He J. Corticofugal modulation of the auditory thalamus.
Princeton, NJ: Princeton University Press, 1959. Exp Brain Res 2003;153:579-590.
176. Ojemann GA. Electrical Stimulation and the neurobiology 195. Jacomme AV, Nodal FR, Bajo VM, et al. The projection
of language. Behav Brain Sci 1983;6:221-226. from auditory cortex to cochlear nucleus in guinea pigs:
177. Ojemann GA, Engel J Jr. Acute and chronic intracra- an in vivo anatomical and in vitro electrophysiological
nial recording and stimulation. In Surgical Treatment of study. Exp Brain Res 2003;153:467-476.
Epilepsies, edited by J Engel Jr. New York: Raven Press, 196. Pandya DN, Rosene DL, Doolittle AM. Corticothalamic
1986, pp. 263-288. connections of auditory-related areas of the temporal lobe
178. Lesser RP, Luders H, Klm G, et al. Extraoperative cortical in the rhesus monkey. J Comp Neurol 1994;345:447-471.
functional localization in patients with epilepsy. [Review]. 197. Suga N, Ma X. Multiparametric corticofugal modulation
J Clin Neurophysiol 1987;4:27-53. and plasticity in the auditory system. Na Rev Neurosci
179. Boatman D, Gordon B, Hart J, Selnes O, Miglioretti D, 2003;4:783-794.
Lenz F. Transcortical sensory aphasia: revisited and re- 198. Weedman DL, Ryugo DK. Projections from auditory cortex
vised. Brain 2000;123(Pt 8):1634-1642. to the cochlear nucleus in rats: synapses on granule cell
180. Gordon B, Lesser RP, Rance NE, et al. Parameters for direct dendrites. J Comp Neurol 1996;371:311-324.
cortical electrical stimulation in the human: histopatho- 199. Weedman DL, Ryugo DK. Pyramidal cells in primary au-
logic confirmation. Electroencephalogr Clin Neurophysiol ditory cortex project to cochlear nucleus in rat. Brain Res
1990;75(5):371-377. 1996;706:97-102.
181. Gordon B, Hart J, Boatman D, et al. Cortical stimula- 200. Boatman D. Cortical bases of speech perception: evidence
tion (interference) during behavior. In Behavioral Neu- from functional lesion studies. Cognition 2004;92(1–2):47–
rology and Neuropsychology, edited by TE Feinberg and 65Review, 2004.
MJ Farah. New York: McGraw-Hill, 1997, pp. 667-672. 201. Boatman D, Hart J Jr, Lesser RP, et al. Right hemisphere
182. Boatman D, Lesser R, Gordon B. Auditory speech pro- speech perception revealed by amobarbital injection and
cessing in the left temporal lobe: an electrical interference electrical interference. Neurology 1998;51(2):458-464.
study. Brain Lang 1995;51:269-290. 202. Boatman DF. Cortical auditory systems: speech and other
183. Wada J, Rasmussen T. Intracarotid injection of sodium complex sounds. Epilepsy Behav 2006;8(3):494-503; Epub
amytal for the lateralization of cerebral speech domi- 2006. Review, 2006.
nance. Experimental and clinical observations. J Neuro- 203. Boatman D, Hart J, Lesser R, et al. Right hemisphere
surg 1960;17:266-282. speech perception as revealed by amobarbital injection
184. Pardo JV, Fox PT. Preoperative assessment of the cerebral and electrical interference. Neurology 1998;51:458-464.
hemispheric dominance for language with CBF PET. Hum 204. Boatman D, Lesser R, Hall C, et al. Auditory perception
Brain Map 1993;1:57-68. of segmental features: a functional neuroanatomic study.
185. Kaplan AM, Bandy DJ, Manwaring KH, et al. Functional J Neurolinguistics 1994;8:225-234.
brain mapping using positron emission tomography scan- 205. Bhatnagar SC, Mandybur G, Buckingham H, Andy OJ.
ning in preoperative neurosurgical planning for pediatric Language representation in the human brain: evidence
brain tumors. Neurosurg Focus 2000;8(2):1-7. from cortical mapping. Brain Lang 2000;74:238-259.
268 SECTION II SYSTEMS

206. Lurito J, Lowe M, Sartorius C, Mathews VP. Comparison onance imaging protocol for intraoperative localization
of fMRI and intraoperative direct cortical stimulation in of critical temporoparietal language areas. Ann Neurol
localization of receptive language areas. J Comput Assist 2002;51:350-360.
Tomogr 2000;24(1):99-105. 213. Pouratian N, Bookheimer SY, Rex DE, Martin NA, Toga
207. Schaffler L, Luders H, Dinner D, Lesser RP, Chelune GJ. AW. Utility of preoperative functional magnetic resonance
Comprehension deficits elicited by electrical stimulation imaging for identifying language cortices in patients with
of Broca’s area. Brain 1993;116:695-715. vascular malformations. J Neurosurg 2002;97:21-32.
208. Wise R, Scott S, Blank C, Mummery CJ, Murphy K, War- 214. Magistretti PJ, Pellerin L, Rothman DL, Shulman RG. En-
burton EA. Separate neural subsystems within ‘Wernicke’s ergy on demand. Science 1999;283:496-497.
area’. Brain 2001;124(1):83-95. 215. Sinai A, Bowers CW, Crainiceanu CM, et al. Electrocor-
209. BlumeWT, Jones DC, Pathak P. Properties of after- ticographic high-gamma activity versus electrical corti-
discharges from cortical electrical stimulation in focal cal stimulation mapping of naming. Brain 2005;128:1556-
epilepsies. Clin Neurophysiol 2004;115:982-989. 1570.
210. Lesser RP, Luders H, Klem G, Dinner DS, Morris HH, 216. Edwards E, Soltani M, Deouell LY, Berger MS, Knight RT.
Hahn J. Cortical after discharge and functional response High-gamma activity in response to deviant auditory stim-
thresholds: results of extraoperative testing. Epilepsia uli recorded directly from human cortex. J Neurophysiol
1984;25:615-621. 2005;94:4269-4280.
211. FitzGerald DB, Cosgrove GR, Ronner S, et al. Location of 217. Korzeniewska A, Crainiceanu CM, Kus R, et al. Dynamics
language in the cortex: a comparison between functional of event-related causality in brain electrical activity. Hum
MR imaging and electrocortical stimulation. Am J Neuro- Brain Mapp 2007; 29(10):1170-92..
radiol 1997;18:1529-1539. 218. Lachaux JP, Jerbi K, Bertrand O, et al. A blueprint for real-
212. Rutten GJ, Ramsey NF, van Rijen PC, Noordmans HJ, van time functional mapping via human intracranial record-
Veelen CW. Development of a functional magnetic res- ings. PLoS ONE 2007;2(10):e1094.
Chapter 18
Mapping of Memory
Jeffrey G. Ojemann and Richard G. Ellenbogen
Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington

䉴 INTRODUCTION Storage refers generically to all processes that


extend beyond “working memory.” Working memory
Though extensive neurosurgical experience has shaped refers to the “scratch pad” (see also Baddeley (1992)).9
our understanding of motor and language systems, other of information that we use during a task. This type of
functions have not been consistently explored. The un- memory is limited. For example the number of digits
derstanding of mapping memory suffers not only from that you can easily remember at one time may be a
this dearth of neurosurgical experience, but also from the telephone number, while a longer string of numbers
many different aspects of memory1 and the correspond- may be more difficult. Working memory is also gener-
ing taxonomy (e.g., encoding vs. retrieval, short-term vs. ally preserved in cases of amnesia. Once the working
long-term, semantic vs. episodic). Further difficulties re- memory phase is exceeded, some sort of storage oc-
late to the fundamentally mercurial nature of memory, curs. Over time, memories are consolidated. These con-
meaning that only when memories are formed or re- solidated memories are much more stable. For example,
trieved we are able to directly assess memory processes, in head injury patients a retrograde amnesia may occur
even though memory itself covers the “storage” interval for some period of time (days) but longerterm mem-
between these two tangible events. ories are preserved. Longer term memories are dam-
Memory has been studied first through neuropsy- aged in more diffuse pathologies, such as Alzheimer’s
chological measures from subjects with normal memory disease.
function and from patients with neurological disorders, Retrieval is the event most typically thought of
and more recently through neuroimaging studies, such in memory studies, as it is unambiguously a memory
as functional magnetic resonance imaging (fMRI).2−4 Im- phenomenon. However, overlap with the brain regions
pairment of several different cortical and subcortical involved in encoding is seen consistently in imaging
structures are associated with memory disorders.5 The studies.
role of the medial temporal lobe for memory formation Memory can also be broken down into different
and retention is well established, especially as the se- types. So-called episodic memory, refers to memory for
vere consequences of bilateral damage to medial tem- specific events. For example, if you remember what you
poral structures.6 Other areas such as the frontal lobe had for breakfast yesterday, that requires memory for a
have particular research and clinical relevance.7,8 specific moment and context. However, if you remem-
ber that eggs are a common breakfast food you are
accessing semantic memory, or memory for facts and
knowledge of the world.1 Semantic memory strongly
WHAT IS MEMORY? overlaps with language systems from a clinical perspec-
tive.
The memory process may be conceptualized as consist- Finally, procedural memory is another term used
ing of the steps of encoding (initial exposure to the thing to describe skills or changes associated with practice
being remembered), storage (the retention of the mem- and repetition.1 This type of memory appears to be de-
ory), and retrieval (the culmination of memory in the act pendent on a certain system, such as the motor sys-
of remembering). tem for the acquisition of motor learning, and largely
Encoding has significant overlap with perception happens on a subconscious level. It can be seen in
and attention. For example, a word that is studied or ma- simple motor tasks, repetitive cognitive tasks, or even
nipulated will be remembered better than a word that is repetitive exposure to the same list of words. Again,
merely viewed or read. Thus, if you see the word “build- the deficits seen with this type of memory would be
ing” and are asked to think of a synonym for the word, expected to overlap with the primary system affected,
you will have a better chance of recognizing the word such as visual processing, somatosensory, or language
on a list than if you merely read the word. processing.

269
270 SECTION II SYSTEMS

Figure 18–1. Schematic diagram showing some of the areas involved in memory.
Though the medial temporal regions (black, left panel) are well known to show
memory deficits with lesions, cortical regions including lateral temporal, parietal, and
frontal lobe (black, right panel, indicating approximate regions involved) play a role in
various memory processes.

MEMORY ANATOMY rather than medial temporal lobe resection was the best
predictor of postoperative memory deficit.11 .
Memory deficits can result from lesions in various brain
regions (Fig. 18–1). Limbic system lesions are classically
associated with significant memory problems, especially 䉴 TECHNIQUES FOR ASSESSMENT
when bilateral. In addition to medial temporal struc- OF MEMORY
tures, damage to the fornix, mamillary bodies, and an-
terior thalamic nuclei are all associated with memory FUNCTIONAL IMAGING
disturbances, each with specific designated syndromes.
Frontal lobe damage seems to affect memory as well, Investigators using fMRI have attempted to activate the
though the nature of the deficit may not be obvious on medial temporal lobe reliably.12−15 However, it has been
cursory clinical evaluation.8,10 However, on tests of in- noticed for some time that a simple memory task is
terference in the memory process, frontal lobe patients more likely to activate frontal lobe systems than the
perform quite poorly and their clinical complaints of hippocampus.1,7,8 More successful efforts to activate the
memory loss often reflect problems with semantic mem- hippocampus and medial temporal lobe have focused on
ory retrieval, such as being unable to remember names tasks that require the patient to assess stimuli for nov-
or places. For some patients following epilepsy surgery, elty or for specific relations between stimuli.12 In addi-
material specific memory (verbal vs. nonverbal material) tion to an incomplete understanding of the physiology
may be the most affected process. Attempts to identify of hippocampus and medial temporal lobes (e.g., What
these processes have been ongoing, if only partially suc- activates them?) technical limitations of fMRI include
cessful. the relatively small size of the hippocampus and local
From an anatomic perspective of memory, the neu- susceptibility artifacts from the nearby aerated petrous
rosurgically relevant observations are limited to two ma- bone.16 These limitations are not insurmountable and
jor findings. First, it is consistently seen that memory the next few years are likely to bring progress in the
deficits are lateralized and thus resections of the dom- understanding, if not the mapping, of these functions.
inant hemisphere temporal lobe will be more likely to
give verbal memory deficits compared to nondominant
resections. Second, the deficit may not be directly linked NONINVASIVE ASSESSMENTS
to hippocampal resection. When a memory deficit is
found following temporal lobe resection, a traditional For elective cases, such as surgical treatment of epilepsy,
view may suggest that this be due to medial temporal a thorough neuropsychological assessment can be in-
resection. However, one of the few studies of the effect valuable for understanding memory function. In partic-
of resection on deficit found that the extent of lateral, ular, well-defined cases of dominant hemisphere mesial
CHAPTER 18 MAPPING OF MEMORY 271

temporal sclerosis are typically associated with decreases


in measures of verbal learning. Preoperative verbal
memory performance is a very strong negative indica-
tor of memory loss with surgery.17 A variety of studies
have shown material specific deficits depending on the
side of surgery,18,19 which may have variable affects on
activities of daily living.

CEREBRAL AMOBARBITAL (WADA)

The intracarotid amobarbital (Wada) test (see chapter 6)


is frequently used to assess memory dominance for pre-
operative neurosurgical evaluation. It has been used to
identify which candidates for temporal lobectomy might
be at risk for global amnesia due to preexisting dysfunc-
tion of the side opposite to that considered for surgery. It
has also been reported to be a predictor of verbal mem- Figure 18–3. A typical map of memory may find
errors (asterisks) in several temporoparietal sites
ory decline from anterior temporal lobectomy20,21 ; how- including fairly anteriorly in lateral temporal cortex.
ever, the actual predictive value may be relatively low
and depend on the type of psychological test used.22
tions sparing these sites.23 This would be in line with
findings that the extent of lateral temporal resection is a
CORTICAL STIMULATION predictor of postoperative verbal memory declines11 in
dominant temporal lobectomy. Further support for the
There have been a very limited number of studies using role of the lateral temporal lobe in memory comes from
cortical stimulation to map memory. Stimulation through the observation that experiential phenomenon can occur
intraoperative or extraoperative mapping11,23 can be ap- from lateral temporal lobe structures alone.24
plied during a short-term memory task with the electri-
cal current applied during encoding, storage, or retrieval
(see Fig. 18–2). Errors in any aspects of memory can be INTRACRANIAL RECORDINGS
found focally throughout frontal, temporal, or parietal
cortex (see Fig. 18–3). In one reported series of patients, The study of memory with electrical recordings has been
resection of neocortical sites showed a greater decline of long interest, but intracranial studies are rare. Repeat
in postoperative verbal memory than those with resec- exposure to an item can alter the response of a sen-
sory evoked potential and novelty responses, such as the
P300, can be modified by repeated exposure.25 Frontal
lobe changes in high-gamma (chi band) frequency have
been reported preliminarily with working memory tasks,
consistent with fMRI findings.26 Extensive research on
intracranial recordings from the lateral temporal lobe27
or hippocampus28 has provided some insight on basic
mechanisms of the neuronal basis for memory in these
regions.
Figure 18–2. Type of stimuli used for memory
mapping. A typical trial would include a target
presentation (encoding), which is named, a distracter
task during storage, and a recall cue (retrieval). 䉴 CLINICAL APPLICATION
Stimulation during any of these stages can result in
the eventual inability to recall the target. With this Memory mapping techniques used to determine a “risk
technique, correct recall can occur even if stimulation versus benefit ratio” in a neurosurgical setting have
induces a naming error during target presentation. If largely focused on patients undergoing anterior temporal
the error occurs with stimulation during recall or if lobe resection (ATR) of seizure foci. Neuropsychological
the error is due to disruption of language circuits, an
error at target presentation naming would be
assessment, intracarotid injection of amobarbital (Wada
expected, as oppose to an error only with stimulation testing), and more recently fMRI, are the primary tools
during recall implying that the site stimulated is used to determine the functional status of the temporal
specific for memory retrieval. lobe to be resected. The “functional adequacy” model
272 SECTION II SYSTEMS

would predict greater risk of decline if memory is ade- is predictive of increased risk of postsurgical verbal
quately supported by the temporal lobe to be resected.29 memory decline (e.g., Stroup et al. (2003)17 , Kneebone
On the other hand, if memory is poor for the ipsilat- et al. (1995)33 Furthermore, adequate Wada testing per-
eral temporal lobe before surgery, then the risk is re- formance after injection of the hemisphere contralateral
duced because there may be a limited amount of func- to the to be resected temporal lobe is predictive of good
tion or nothing to lose. Memory mapping techniques verbal memory outcome (e.g., Bell et al. (2000)30 , Chiar-
are also used to determine the functional status of the avalloti et al. (2001)31 . However, not all studies are in
temporal lobe contralateral to the side of resection. The agreement. For example, Kubu et al.39 demonstrated
“cognitive reserve” model would predict better memory that patients who failed the Wada test bilaterally had
outcome for patients with a functional contralateral tem- relatively few losses in memory abilities after surgery
poral lobe.29 (and no global amnesia), and as previously discussed,
In this section, Wada testing and fMRI memory map- the predictive value of Wada testing is more limited
ping validation studies are briefly reviewed. Both of when taking into account other factors such as presur-
these techniques are discussed in further detail else- gical neuropsychology-defined verbal memory ability
where in this volume (Chapters 3 and 4 for fMRI and and side of surgery.17,20,40 Few studies have evaluated
Chapter 8 for Wada testing). There is also an extensive Wada testing in predicting nonverbal memory decline
discussion of the utility of presurgical neuropsychologi- after a nonlanguage dominant ATR with results being
cal assessment in predicting postsurgical memory decre- equivocal.
ments associated with ATR in Chapter 7.

FUNCTIONAL MRI
WADA TESTING
Table 18–2 summarizes all of the studies that have eval-
Table 18–1 summarizes the extant literature studying uated the efficacy of fMRI activation as a predictor of
the utility of Wada testing in predicting material spe- postoperative memory impairment after ATR. In all of the
cific memory impairments after unilateral temporal lobe studies, greater ipsilateral activation (or greater asymme-
resection. A variation in Wada testing methods and pro- try vs. the contralateral hemisphere; commonly known
cedures make comparisons across studies difficult. How- as a laterality index) is related to greater memory de-
ever, generally speaking, most studies find that ade- cline after surgery to varying degrees using different
quate patient performance after amobarbital injection testing materials. Two of the studies compared the pre-
of the to be resected language-dominant temporal lobe dictive validity of both fMRI and Wada testing.42,46 Us-
ing an auditory semantic memory encoding task, Binder
et al.42 showed that greater left-sided fMRI activation be-
䉴 TABLE 18–1. WADA PREDICTION OF fore surgery predicted postsurgical verbal memory de-
POSTOPERATIVE NEUROPSYCHOLOGICAL cline, whereas Wada testing was noncontributory. By
MEMORY OUTCOME AFTER UNILATERAL using a visual encoding of scenes task, Rabin et al.46
ANTERIOR TEMPORAL LOBE RESECTION showed that greater ipsilateral fMRI activation of the hip-
Predicts Verbal Memory Does not Predict pocampus, parahippocampal gyrus, and fusiform gyrus
Decline Verbal Memory Decline predicted visual memory decline after surgery, whereas
Wada testing was noncontributory.
Baxendale et al. (2007)20 Kirsch et al. (2005)38
Bell et al. (2000)30 Kubu et al. (2000)39
Chiaravalloti and Glosser Lineweaver et al. (2006)40
(2001)31
FUNCTIONAL MRI VERSUS
Joiket et al. (1997)32 Loringet al. (1990) 41 WADA TESTING
Kneebone et al. (1995)33
Lacruz et al. (2004)34 Table 18–3 summarizes all of the studies that validated
Lee et al. (2005)35 fMRI memory paradigms compared to Wada testing.
Loringet al. (1995)21 fMRI would be an attractive alternative to Wada testing
Sabsevitz et al. (2001)36 in determining cognitive risks associated with surgery
Stroupet al. (2003)17 given that it is less invasive, can be repeated multi-
Wyllie et al. (1990)37 ple times, is widely available and poses minimal risks.
Predicts Nonverbal Does not Predict Nonverbal However, it is unclear if fMRI methodology is valid and
Decline Memory Memory Decline reliable enough to replace Wada testing given the vari-
ability in results shown in Table 18–3. The first studies to
Lacruz et al. (2004)34 Chiaravalloti and Glosser
publish data were very promising14,15 as there was 100%
Lineweaver et al. (2006)40 (2001)31
concordance between fMRI and Wada in identifying
CHAPTER 18 MAPPING OF MEMORY 273

䉴 TABLE 18–2. FUNCTIONAL MAGNETIC RESONANCE IMAGING (fMRI) PREDICTION OF


POSTOPERATIVE NEUROPSYCHOLOGICAL MEMORY CHANGE AFTER UNILATERAL ANTERIOR
TEMPORAL LOBE RESECTION

Study N/Sample Task Results

Binder et al. (2008)42 60 left-ATR Auditory encoding animal Greater left-side fMRI activation before
surgery predicted
62 right-ATR Decision task 11% of variance in postsurgical verbal
memory decline beyond the 54% of
variance accounted for by preoperative
memory testing and age at onset
Frings et al. (2008)43 22 ATR Object location memory Greater ipsilateral hippocampal fMRI
activation correlated (r = 0.49) with
verbal learning decline after surgery
Janszky et al. (2005)44 16 right-ATR Route learning task Greater ipsilateral fMRI activation before
surgery correlated (r = 0.71) with visual
memory decline after right-ATL
Powell et al. (2007)45 15 ATR Visual encoding of words, Greater ipsilateral mesial temporal lobe
pictures of common fMRI activation (compared to
objects, and faces contralateral activation) correlated with
postsurgical verbal memory decline after
left-ATR and nonverbal memory decline
after right-ATR
Rabin et al. (2004)46 25 ATR Visual encoding of novel Greater ipsilateral fMRI activation of
scenes mesial temporal lobe before surgery was
correlated (r = − 0.56) with visual
memory decline after surgery
Richardson et al. (2004)47 10 left-ATR with HS Visual encoding of words Greater left hippocampal activation (vs.
right) correlated (r = 0.82) with verbal
memory decline after surgery
Richardson et al. (2006)48 12 left-ATR with HS Visual encoding of words Greater left hippocampal activation, but
not right hippocampus or amygdala
activation, predicted worsening in word
list learning ability after surgery

ATR, anterior temporal lobe resection; HS, hippocampal sclerosis.

䉴 TABLE 18–3. COMPARISON OF FMRI MEMORY LATERALIZATION PARADIGMS TO WADA TESTING


IN INTRACTABLE EPILEPSY SURGERY CANDIDATES

Study N/Sample Task Concordance Rates with Wada

Branco et al. (2006)49 5 epilepsy Visual encoding of novel 60% concordance


patterns, scenes, and words
Deblaere et al. (2005)50 18 TLE Visual encoding of novel vs. old Concordance rates not reported; fMRI
pictures of common objects activation correlated with Wada results
in right-TLE but not left-TLE
Detre et al. (1998)15 9 TLE Visual encoding of scenes 100% concordance
Golby et al. (2002)14 9 TLE Visual encoding of novel 100% concordance
patterns, faces, scenes, and
words
Rabin et al. (2004)46 35 TLE Visual encoding of novel scenes Concordance rates not reported;
correlation (r = 0.60) between fMRI and
WADA asymmetry ratios for right-TLE
but not significant for left-TLE
Szaflarski et al. (2004)51 5 TLE Visual encoding of novel scenes 50% concordance; fMRI nonlateralizing in
1 ET that varied in verbal two cases and discordant in one case
encodability and the presence vs. Wada
of people

TLE, temporal lobe epilepsy; ET, extratemporal.


274 SECTION II SYSTEMS

the functional status of the mesial temporal lobes. 13. D’Arcy RC, Bolster RB, Ryner L, Mazerolle EL, Grant J, Song
Problematically, later studies showed that concordance X. A site directed fMRI approach for evaluating functional
was not perfect, and suggested that fMRI and Wada re- status in the anterolateral temporal lobes. Neurosci Res
sults are similar in identifying the functional status of 2006;57:120-128.
the nonlanguage dominant hemisphere, but not the lan- 14. Golby AJ, Poldrack RA, Illes J, Chen D, Desmond JE,
Gabrieli JD. Memory lateralization in medial temporal lobe
guage dominant hemisphere.46,50
epilepsy assessed by functional MRI. Epilepsia 2002;43:855-
863.
15. Detre J, Maccotta L, King D, et al. Functional MRI lateral-
SUMMARY OF CLINICAL APPLICATION ization of memory in temporal lobe epilepsy. Neurology
1998;50:926-932.
Practically speaking, the best predictors of memory dis- 16. Ojemann J, Akbudak E, Snyder AZ, McKinstry RC, Raichle
turbance following epilepsy surgery remain the preop- ME, Conturo TE. Anatomic localization and quantitative
erative MRI and preresection neuropsychological status. analysis of gradient refocused echo-planar fMRI suscep-
Patients with normal MRI and good presurgical function tibility artifacts. Neuroimage 1997;6:156-167.
17. Stroup E, Langfitt J, Berg M, McDermott M, Pilcher W,
have the greatest risk of new deficit. The use of Wada
Como P. Predicting verbal memory decline following ante-
testing still has a role in patient assessment although
rior temporal lobectomy. Neurology 2003;43:1800-1805.
somewhat limited by the factors discussed previously. 18. Alpherts WC, Vermeulen J, van Rijen PC, da Silva FH, van
The role of fMRI is still evolving. The role of intraop- Veelen CW. Dutch collaborative epilepsy surgery program:
erative mapping remains largely unexplored as the very verbal memory decline after temporal epilepsy surgery?:
nature of memory is much less well understood than A 6-year multiple assessments follow-up study. Neurology
other, more commonly mapped functions. 2006;67(4):626-631.
19. Meador K. Memory loss after left anterior temporal lobec-
tomy in patients with mesial temporal lobe sclerosis.
REFERENCES Epilepsy Curr 2006;6(2):44-45.
20. Baxendale S, Thompson P, Harkness W, Duncan J. The
1. Squire LR. Memory and Brain. New York: Oxford University role of the intracarotid amobarbital procedure in predict-
Press, 1987. ing verbal memory decline after temporal lobe resection.
2. Dolan RJ, Paulesu E, Fletcher P. Human memory systems. Epilepsia 2007;48(3):546-552.
In Human Brain Function, edited by RSJ Frackowiak, KJ 21. Loring DW, Meador KJ, Lee GP, et al. Wada memory asym-
Friston, CD Frith, et al. San Diego: Academic Press, 1997, metries predict verbal memory decline after anterior tem-
pp. 367-404. poral lobectomy. Neurology 1995;45:1329-1333.
3. Ojemann JG, Buckner RL, Cortetta M, Raichle ME. Imaging 22. Dodrill CB, Ojemann GA. An exploratory comparison of
studies of memory and attention. Neurosurg Clin N Am three methods of memory assessment with the intracarotid
1997;8:307-319. amobarbital procedure. Brain Cogn 1997;33:210-223.
4. Tharin S, Golby A. Functional brain mapping and its ap- 23. Perrine K, Devinsky O, Uysal S, Luciano DJ, Dogali M. Left
plications to neurosurgery. Neurosurgery 2007;60(4 Suppl temporal neocortex mediation of verbal memory: evidence
2):185-201. from functional mapping with cortical stimulation. Neurol-
5. Ojemann RG. Correlations between specific human brain ogy 1994;44:1845-1850.
lesions and memory changes: a critical survey of the liter- 24. Moriarity JL, Boatman D, Krauss GL, Storm PB, Lenz FA.
ature. Neurosci Res Program Bull Suppl 1966;4:1-70. Human ‘memories’ can be evoked by stimulation of the lat-
6. Corkin S. Lasting consequences of bilateral medial tempo- eral temporal cortex after ipsilateral medial temporal lobe
ral lobectomy. Semin Neurol 1984;4:249-259. resection. J Neurol Neurosurg Psychiatry 2001;71:549-551.
7. Buckner RL, Kelley WM, Petersen SE. Frontal cortex 25. van Hooff JC. The influence of encoding intention on elec-
contributes to human memory formation. Nat Neurosci trophysiological indices of recognition memory. Int J Psy-
1999;2:311-314. chophysiol 2005;56:25-36.
8. Ojemann JG, Kelley WM. The frontal lobe role in memory. 26. Miller KJ, Ojemann JG. Electrocorticographic cortical
Epilepsy Behav 2003;3:309-315. changes during a working memory task. Soc Neurosci Abs
9. Baddeley A. Working memory. Science 1992;255:556-559. 2006;610-612.
10. Kho KH, Rutten GJM, Leijten FSS, et al. Working mem- 27. Ojemann GA, Schoenfield-McNeill J, Corina D. Different
ory deficits after resection of the dorsolateral prefrontal neurons in different regions of human temporal lobe dis-
cortex predicted by functional magnetic resonance imag- tinguish correct from incorrect identification or memory.
ing and electrocortical stimulation mapping. J Neurosurg Neuropsychologia 2004;42(10):1383-1393.
2007;106(6 Suppl):501-505. 28. Viskontas IV, Knowlton BJ, Steinmetz PN, Fried I. Differ-
11. Ojemann GA, Dodrill CB. Verbal memory deficits after left ences in mnemonic processing by neurons in the human
temporal lobectomy for epilepsy: mechanism and intraop- hippocampus and parahippocampal regions. J Cogn Neu-
erative prediction. J Neurosurg 1985;62(1):101-107. rosci 2006;18(10):1654-1662.
12. Binder JR, Bellgowan PS, Hammeke TA, Possing ET, Frost 29. Chelune GJ. Hippocampal adequacy versus functional re-
JA. A comparison of two fMRI protocols for eliciting hip- serve: predicting memory functions following temporal
pocampal activation. Epilepsia 2005;46:1061-1070. lobectomy. Arch Clin Neuropsychol 1995;10:413-432.
CHAPTER 18 MAPPING OF MEMORY 275

30. Bell BD, Davies KG, Haltiner AM, Walters GL. Intracarotid 41. Loring DW, Lee GP, Meador KJ, et al. The intracarotid
amobarbital procedure and prediction of postoperative amobarbital procedure as a predictor of memory failure
memory in patients with left temporal lobe epilepsy and following unilateral temporal lobectomy. Neurology 1990;
hippocampal sclerosis. Epilepsia 2000;41:992-997. 40:605-610.
31. Chiaravalloti ND, Glosser G. Material-specific memory 42. Binder JR, Sabsevitz DS, Swanson SJ, Hammeke TA, Ragha-
changes after anterior temporal lobectomy as predicted van M, Mueller WM. Use of preoperative functional MRI to
by the intracarotid amobarbital test. Epilepsia 2001;42: predict verbal memory decline after temporal lobe epilepsy
902-911. surgery. Epilepsia 2008;49(8):1377-1-394.
32. Jokeit H, Ebner A, Holthausen H, et al. Individual predica- 43. Frings L, Wagner K, Halsband U, Schwarzwald R, Zentner J,
tion of change in delayed recall of prose passages after left- Schulze-Bonhage A. Lateralization of hippocampal activa-
sided anterior temporal lobectomy. Neurol 1997;49:481- tion differs between left and right temporal lobe epilepsy
487. patients and correlates with postsurgical verbal learning
33. Kneebone AC, Chelune GJ, Dinner DS, et al. Intracarotid decrement. Epilepsy Res 2008;78:161-170.
amobarbital procedure as a predictor of material-specific 44. Janszky J, Jokeit H, Kontopoulou K, et al. Functional MRI
memory change after anterior temporal lobectomy. Epilep- predicts memory performance after right mesiotemporal
sia 1995;36:857-865. epilepsy surgery. Epilepsia 2005;46:244-250.
34. Lacruz ME, Alarcón G, Akanuma N, et al. Neuropsycholog- 45. Powell HW, Richardson MP, Symms MR, et al. Preop-
ical effects associated with temporal lobectomy and amyg- erative fMRI predicts memory decline following anterior
dalohippocampectomy depending on Wada test failure. J temporal lobe resection. J Neurol Neurosurg Psychiatry
Neurol Neurosurg Psychiatry 2004;75:600-607. 2007;79(6):686-693.
35. Lee GP, Westerveld M, Blackburn LB, Park YD, Loring DW. 46. Rabin ML, Narayan VM, Kimberg DY, et al. Functional MRI
Prediction of verbal memory decline after epilepsy surgery predicts post-surgical memory following temporal lobec-
in children: effectiveness of Wada memory asymmetries. tomy. Brain 2004;127:2286-2298.
Epilepsia 2005;46:97-103. 47. Richardson MP, Strange BA, Thompson PJ, et al. Pre-
36. Sabsevitz DS, Swanson SJ, Morris GL, Mueller WM, Sei- operative verbal memory fMRI predicts post-operative
denberg M. Memory outcome after left anterior temporal memory decline after left temporal lobe resection. Brain
lobectomy in patients with expected and reversed Wada 2004;127:2419-2426.
memory asymmetry scores. Epilepsia 2001;42:1408-1415. 48. Richardson MP, Strange BA, Duncan JS, Dolan RJ. Memory
37. Wyllie E, Naugle R, Awad I, et al. Intracarotid amo- fMRI in left hippocampal sclerosis: optimizing the approach
barbital procedure. I. Prediction of decreased modality- to predicting postsurgical memory. Neurology 2006;66:699-
specific memory scores after temporal lobectomy. Epilep- 705.
sia 1991;32:857-864. 49. Branco DM, Suarez RO, Whalen S, et al. Functional MRI
38. Kirsch HE, Walker JA, Winstanley FS, et al. Limitations of of memory in the hippocampus: laterality indices may be
Wada memory asymmetry as a predictor of outcomes after more meaningful if calculated from whole voxel distribu-
temporal lobectomy. Neurology 2005;65:676-680. tions. Neuroimage 2006;32:592-602.
39. Kubu CS, Girvin JP, McLachlan RS, Pavol M, Harnadek MC. 50. Deblaere K, Backes WH, Tieleman A, et al. Lateralized ante-
Does the intracarotid amobarbital procedure predict global rior mesiotemporal lobe activation: semirandom functional
amnesia after temporal lobectomy? Epilepsia 2000;41:1321- MR imaging encoding paradigm in patients with temporal
1329. lobe epilepsy-initial experience. Radiology 2005;236:996-
40. Lineweaver TT, Morris HH, Naugle RI, Najm IM, Diehl 1003.
B, Bingaman W. Evaluating the contributions of state- 51. Szaflarski JP, Holland SK, Schmithorst VJ, et al. High-
of-the-art assessment techniques to predicting memory resolution functional MRI at 3T in healthy and epilepsy sub-
outcome after unilateral anterior temporal lobectomy. jects: hippocampal activation with picture encoding task.
Epilepsia 2006;47:1895-1903. Epilepsy Behav 2004;5:244-252.
This page intentionally left blank
INDEX
Note: Page numbers followed by f and t indicate figures and tables, respectively.

A localization, 8–9, 9f starting parameters, 95t


Acoustic processing applications, 259 optical imaging in animals and humans, subject preparation, 94
AEFs. See Auditory evoked magnetic fields 135 video-EEG, 96, 98t
AEPs. See Auditory evoked potentials Auditory evoked magnetic fields (AEFs), 123 intraoperative cortical mapping
AF. See Arcuate fasciculus Auditory evoked potentials (AEPs), 256f–257f anesthesia, 104
Afterdischarge (AD) threshold, 97 Auditory illusions, 259 in clinical practice, 106
Alexia, 105 Auditory stimulation, 43f, 137, 250 craniotomy, 104
Amygdala, 27 AVMs. See Arteriovenous malformations electrocorticography, 105
Anatomic landmarks, 3 vs. extraoperative cortical mapping,
Anatomic modularity, 56 B 98–99
Anatomical localization Barbiturates, 110–11 language mapping, 105–6
auditory function, 8–9, 9f Basal temporal language area (BTLA), 213 positioning, 104
cortical structural anatomy, 4 Baseline, or Frankfurt plane, 4 postoperative outcomes, 106
language function, 5, 7–8, 7f Benzodiazepines, 110 principles, 103
motor and sensory functions, 4–5 Bipolar stimulation, 95 sensorimotor stimulation, 105
visual function, 8, 9f Blood oxygen level dependent (BOLD) fMRI, somatosensory evoked potential
Anesthesia, brain mapping surgery 31, 33, 34f, 195–96 recording, 104
anesthetic management, 114–15 Brain architecture, optical imaging sensorimotor cortex
anesthetics effects, 109 auditory cortex, 135 cerebral hemisphere, 19–20
barbiturates, 110–11 language cortex, 137, 139 clinical applications, 198–99
benzodiazepines, 110 somatosensory cortex, 135–37, 136f direct cortical stimulation, 192–94
craniotomy, 113 visual cortex, 134–35 electrocorticographic spectral
dexmedetomidine, 112 Brain diseases, optical imaging pathology analysis, 156–57
etomidate, 111 epilepsy, 139–40 functional MRI, 195–96
general, 115–16 intracranial hematomas, 143–45 historical perspectives, 189–90
intraoperative cortical mapping, 104 stroke, 141–43 localization, 191–92
intraoperative monitoring, 114 traumatic brain injury, 143–45 magnetoencephalography, 196–97
ketamine, 111 Brain fissural patterns, 13 positron emission tomography, 196
local anesthetics, 110 Brain mapping, 94 primary motor cortex, 190–91
management, 114–15 electrocorticographic spectral analysis somatosensory-evoked potentials,
nitrous oxide, 112–13 advantages and pitfalls, 161–62 194–95
opiates, 111 auditory function localization, 157 transcranial magnetic stimulation, 197
preoperative care, 113–14 in clinical practice, 160–61 Brain mapping in children
propofol, 112 historical perspectives, 151 clinical indications, 167
sedatives, 109 instrumentation methodology, 153 diffusion tensor imaging, 171
volatile anesthetic neuropharmacology, language cortex mapping, 157–58 diffusion-weighted imaging, 170–71
113, 113t mapping functional connectivity, 159 functional MRI
Anesthetic agents mapping sensorimotor function, brain tumors, 170
barbiturates, 110–11 155–56 cortical plasticity and reorganization,
benzodiazepines, 110 memory function mapping, 158–59 170
dexmedetomidine, 112 principles, 152–53 epilepsy, 170
etomidate, 111 recording techniques and data general considerations in pediatrics,
ketamine, 111 acquisition, 154–55 169–70
nitrous oxide, 112–13 signal analysis, 155 magnetoencephalography
opiates, 111 strengths and weaknesses, 159–60 epilepsy, 173–74
propofol, 112 subject inclusion and preparation, 153 functional mapping, 172–73
Anterior basal orbitofrontal sulci, 24f, 25 visual function localization, 156–57 general considerations in pediatrics,
Anterior temporal lobectomy (ATL), 62–63, extraoperative functional mapping 172
63f, 79 cortical stimulation, 96, 96f perfusion-weighted imaging, 171
Arcuate fasciculus (AF), 204f–205f electronic principles, 95 positron emission tomography
Arteriovenous malformations (AVMs), 48 historical perspectives, 93–94 brain tumors, 174
Association of deficits, 57 instrumentation, 95 epilepsy, 174–75
ATL. See Anterior temporal lobectomy vs. intraoperative functional mapping, functional mapping, 174
Attention, 60, 66 98–99 general considerations in pediatrics,
Auditory cortex language task, 96 174
electrocorticographic spectral analysis, methodology, 94 single photon emission tomography
157 saftey and adverse effects, 97–98 additional clinical applications, 176

277
278 INDEX

Brain mapping in children (Cont.) Central sulcus superior temporal sulcus, 16–17
brain tumors, 175 cerebral hemisphere, 15, 15f sylvian opercula, 13–15, 14f
epilepsy, 175–76 localization, 4–5, 6f temporal lobe, 15f, 18–19
general considerations in pediatrics, imaging approaches, 20 Cerebral localization
175 Cerebral amobarbital testing activation map creation, 39, 40f
specific techniques, 167–68, 168f advantages and pitfalls, 87–88 clinical inferences, 41–44, 43f
Brain mapping surgery applications, 86 data analysis, 33–39, 36f, 38f
anesthetic agents in clinical practice, 87 MRI and MR scanners, 33, 34f
barbiturates, 110–11 data acquisition and data analysis MRI data collection, 33, 35f
benzodiazepines, 110 language, 84–85 thumbnail sketch, 31–33, 32f
dexmedetomidine, 112 memory, 85–86 visualization on cortical surface, 39–41,
etomidate, 111 historical perspectives, 79–80 40f, 42f
ketamine, 111 instrumentation methods, 81–83 Cerebral oxygen volume (COV), 142
nitrous oxide, 112–13 invasive pediatric brain mapping, Cerebral sulci, 13, 14t
opiates, 111 177 Cingulate sulcus, 22, 22f
propofol, 112 principles, 79–80 Clinical applications
anesthetic management, 114–15 recording techniques, 83–84 functional MRI
anesthetics effects, 109 strengths and weakness, 87 arteriovenous malformations, 48
craniotomy, 113 subject preparation methods, 80–81 brain tumors, 47–48, 47f, 49f
with general anesthesia, 115–16 validation techniques, 86–87 epilepsy, 48, 50
intraoperative monitoring, 114 Cerebral blood oxygenation (CBO), stereotactic radiosurgery, 50, 50f
local anesthetics, 110 141–42 surgical navigation, 46–47, 48f
preoperative care, 113–14 Cerebral blood volume (CBV), 132 surgical planning, 46, 47f
sedatives, 109 Cerebral hemisphere hearing, 257–59
volatile anesthetic neuropharmacology, anterior speech area, 17f, 20–21 acoustic processing, 259
113, 113t calcarine sulcus, 23 speech and language processing,
Brain morphology, 13, 14t central sulcus, 15, 15f 260–62
Brain sulcation, 13 cingulate sulcus, 22, 22f language
Brain sulci frontal lobe, 15f, 17–18, 17f basal temporal language area, 213
primary, 13, 14t frontomarginal sulcus, 17 multilingual patients, 212–13
secondary, 13, 14t gyri notion of degeneracy, 213–14
tertiary, 13, 14t and anterior basal orbitofrontal sulci, young children, 212
Brain tumors 24f, 25 magnetoencephalography
functional MRI, 47–48, 47f, 49f and posterior basal temporal sulci, vs. electroencephalography, 119
intrinsic, 198–99 24f, 25 evoked magnetic activity, 122
pediatric brain mapping gyri of mesial surface language-related brain magnetic
functional MRI, 170 cingulate gyrus, 22f, 24 fields, 124–26
positron emission tomography, 174 cuneus, 22f, 25 movement-related magnetic fields,
single photon emission tomography, gyrus rectus, 23, 24f 123
175 lingual gyrus, 22f, 25 somatosensory evoked magnetic
Brain-behavior relationship medial frontal gyrus, 24 fields, 123
data analysis, 61 paracentral lobule, 24 spontaneous magnetic activity,
data interpretation, 61–62 precuneus, 22f, 24 120–22
evaluation methods, 58–59 supplementary motor area, 24 visual evoked magnetic fields, 123
historical perspective, 55–56 inferior and mesial surface, 24f, 25 memory
investigation tools inferior frontal sulcus, 15–16 functional MRI, 272, 273t
attention, 60 inferior precentral sulcus, 15 functional MRI vs. Wada testing, 272,
cognitive speed, 60–61 insula of Reil, 19 273t, 274
executive functions, 61 intraparietal sulcus, 16, 16f Wada testing, 272, 272t
intelligence, 59 lateral sulcus, 13–15, 14f optical spectroscopic imaging
language, 60 lateral surface, 13 diffuse optical tomography, 133
memory, 59–60, 60f gyri, 15f, 17 near infrared spectroscopy, 133
visual perception, 60 limbic lobe region, 25–28, 26f neurovascular coupling, 131–32
working memory, 60 mesial surface, 22, 22f optical recording of intrinsic signal,
principles mesial temporal region, 25–28, 26f 132–33, 132f
clinical mapping, 56–57 occipital lobe, 18f, 19 physiological characteristics, 131
lesion method, 57–58 parietal lobe, 18, 18f optical spectroscopic imaging
Broca’s area, 7, 8f parieto-occipital sulcus, 22–23 architecture
BTLA. See Basal temporal language posterior speech area, 18f, 21–22 auditory cortex, 135
area precentral sulcus, 14f, 16 language cortex, 137, 139
premotor cortex, 20 somatosensory cortex, 135–37, 136f
C rostral sulci, 22, 22f visual cortex, 134–35
Calcarine sulcus, 8, 23 sensorimotor cortex optical spectroscopic imaging pathology
Callosomarginal sulcus, 22 postcentral gyrus, 20 epilepsy, 139–40
CBO. See Cerebral blood oxygenation precentral gyrus, 19–20 intracranial hematomas, 143–45
CBV. See Cerebral blood volume striate visual cortex, 23 stroke, 141–43
Central pain, 199 superior frontal sulcus, 16, 16f traumatic brain injury, 143–45
INDEX 279

pediatric brain mapping mapping sensorimotor function, starting parameters, 95t


single photon emission tomography, 155–56 subject preparation, 94
176 memory function mapping, 158–59 video-EEG, 96, 98t
sensorimotor cortex visual function localization, 156–57
motor cortical stimulation, 199 in clinical practice, 160–61 F
Rolandic cortex, 198–99 historical perspectives, 151 Face sensory function, 5, 7f
visual cortex mapping functional connectivity, 159 Fixed battery approach, 58
direct cortical stimulation, 235 methodology Flexible battery approach, 58
event-related scalp visual evoked instrumentation, 153 Fluorine-18 2-fluoro-2-deoxy-D-glucose
potential, 233–34 recording techniques and data (FDG), 228
functional MRI, 232–33 acquisition, 154–55 PET, 233
local field potential recordings, 235 signal analysis, 155 uptake, 174
magnetoencephalography, 233 subject inclusion and preparation, 153 fMRI. See Functional magnetic resonance
positron emission tomography, 233 principles, 152–53 imaging
transcranial magnetic stimulation, strengths and weaknesses, 159–60 Frontal lobe
234–35 Electrocorticography cerebral hemisphere, 15f, 17–18, 17f
Clinical indications brain mapping, 105 neuropsychological testing
language-related brain magnetic fields, invasive pediatric brain mapping, 180 attention, 66
124 Electroencephalography (EEG) design and verbal fluency, 66–67
pediatric brain mapping, 167 dose responses, 110f executive functioning, 67, 68f
Cognitive speed, 60–61 vs. magnetoencephalography, 119 working memory, 66
Collateral sulcus, 25 Wada testing, 82f–83f Frontomarginal sulcus, 17
Concordance across methods Encoding, 269 Full scale IQ, 59
hearing, 255, 257 Epidural stimulation, 98 Functional brain regions
visual cortex, 231 Epilepsy anterior speech area, 17f, 20–21
Cortical plasticity, 170 executive functions, 68f calcarine sulcus, 23
Cortical stimulation, 271 functional MRI, 48, 50 central sulcus, 15, 15f
Cortical stimulation mapping (CSM), 209 imaging pathology in animals and cingulate sulcus, 22, 22f
Cortical substrate, 204–7 humans, 139–40 frontal lobe, 15f, 17–18, 17f
Craniotomy, 104, 113, 180 pediatric brain mapping gyri
Cuneus, 25 functional MRI, 170 and anterior basal orbitofrontal sulci,
Current intensity, 95 magnetoencephalography, 173–74 24f, 25
positron emission tomography, and posterior basal temporal sulci,
D 174–75 24f, 25
DCS. See Direct cortical stimulation single photon emission tomography, gyri of mesial surface
Design fluency, 66–67 175–76 cingulate gyrus, 22f, 24
Dexmedetomidine, 112 Episodic memory cuneus, 22f, 25
Diffuse optical tomography (DOT), 133, 137f definition, 269 gyrus rectus, 23, 24f
Diffusion tensor imaging (DTI), 171, 251 nonverbal, 63–64, 64t lingual gyrus, 22f, 25
tractography, 211 verbal, 62–63, 63f medial frontal gyrus, 24
Diffusion-weighted imaging (DWI), 170–71 ERS. See Event-related synchronization paracentral lobule, 24
Direct cortical stimulation (DCS), 257 ESM. See Electrocortical stimulation mapping precuneus, 22f, 24
hearing, 251–53 Etomidate, 88, 111 supplementary motor area, 24
sensorimotor cortex, 192–94 Event-related potentials, 152 inferior and mesial surface, 24f, 25
visual cortex, 230, 235 Event-related scalp visual evoked potential, inferior frontal sulcus, 15–16
Domain specificity hypothesis, 57 233–34 inferior precentral sulcus, 15
Dorsal stream, 223 Event-related synchronization (ERS), 152 insula of Reil, 19
DOT. See Diffuse optical tomography Evoked-related potentials, 152 intraparietal sulcus, 16, 16f
Double dissociation, 58 Executive functioning lateral sulcus, 13–15, 14f
DTI. See Diffusion tensor imaging epilepsy, 68f lateral surface, 13
DWI. See Diffusion-weighted imaging frontal lobe localization, 67 gyri, 15f, 17
neuropsychological testing tool, 61 limbic lobe region, 25–28, 26f
E Expressive language-specific cortex, mesial surface, 22, 22f
Echo-planar imaging (EPI), 33 125–26 mesial temporal region, 25–28, 26f
eCSM. See Extraoperative CSM Extraoperative cortical stimulation, 178–80, occipital lobe, 18f, 19
EEG. See Electroencephalography 207 parietal lobe, 18, 18f
Electrical interference. See Direct cortical Extraoperative CSM (eCSM), 207 parieto-occipital sulcus, 22–23
stimulation (DCS) Extraoperative functional cortical mapping posterior speech area, 18f, 21–22
Electrical stimulation, 69 cortical stimulation, 96, 96f precentral sulcus, 14f, 16
Electrocortical stimulation mapping (ESM), electronic principles, 95 premotor cortex, 20
151, 154f, 158 historical perspectives, 93–94 rostral sulci, 22, 22f
Electrocorticographic (EcoG) spectral instrumentation, 95 sensorimotor cortex
analysis vs. intraoperative functional mapping, postcentral gyrus, 20
advantages and pitfalls, 161–62 98–99 precentral gyrus, 19–20
applications language task, 96 striate visual cortex, 23
auditory function localization, 157 methodology, 94 superior frontal sulcus, 16, 16f
language cortex mapping, 157–58 saftey and adverse effects, 97–98 superior temporal sulcus, 16–17
280 INDEX

Functional brain regions (Cont.) positioning, 104 gamma range oscillations, 251
sylvian opercula, 13–15, 14f postoperative outcomes, 106 surface electrical recordings, 249–51
temporal lobe, 15f, 18–19 principles, 103 structural imaging methods, 245–46
Functional imaging methods sensorimotor stimulation, 105 Hemifield stimulation, 123
hearing somatosensory evoked potential Hemorrhage, 98
frequency, 246 recording, 104 Heschl’s gyrus, 9, 9f
sound level, 247 visual cortex Human language mapping
sound localization, 249 advantages and pitfalls, 235–36 clinical applications
temporal aspects of sound, 248–49 clinical applications, 232–35 basal temporal language area, 213
language concordance across methods, 231 multilingual patients, 212–13
vs. cortical stimulation mapping, direct cortical stimulation, 230 notion of degeneracy, 213–14
210–11 electrocorticographic spectral young children, 212
modalities, 210 analysis, 156–57 cortical substrate, 204–7
memory assessment technique, 270 historical perspectives, 219–20 direct cortical stimulation, 207–10
functional magnetic resonance imaging local field potential recordings, functional imaging mapping, 210–11
(fMRI) 230–31 historical background, 203
cerebral localization neuroanatomy, 220–23 intracranial electrodes, 211–12
activation map creation, 39, 40f neurophysiology, 220–23 intraoperative cortical mapping, 105–6
clinical inferences, 41–44, 43f noninvasive neuroimaging, 227–30 lateralization versus localization, 203–4
data analysis, 33–39, 36f, 38f operative anatomy, 225–27 local field potential recordings, 211–12
MRI and MR scanners, 33, 34f optical imaging in animals and localization, 5, 7–8, 7f
MRI data collection, 33, 35f humans, 134–35 neuropsychological testing, 60
thumbnail sketch, 31–33, 32f organizational principles, 223–24 structural imaging mapping
visualization on cortical surface, structural imaging, 225–27 Broca’s morphology, 211
39–41, 40f, 42f DTI tractography, 211
future experiments, 50–52 G subcortical substrate, 204–7
memory, 272, 273t Gamma range oscillations, 251 temporal lobe localization, 65
neurosurgical applications General linear model, 35 Wada test, 84–85
arteriovenous malformations, 48 General anesthesia, 115–16. See also Human visual system
brain tumors, 47–48, 47f, 49f Anesthesia advantages and pitfalls, 235–36
epilepsy, 48, 50 Granger causality, 159 clinical applications
stereotactic radiosurgery, 50, 50f Gyri direct cortical stimulation, 235
surgical navigation, 46–47, 48f and anterior basal orbitofrontal sulci, 24f, event-related scalp visual evoked
surgical planning, 46, 47f 25 potential, 233–34
paradigms, 45–46, 46f lateral surface, 15f, 17 functional MRI, 232–33
pediatric brain mapping of mesial surface local field potential recordings, 235
brain tumors, 170 cingulate gyrus, 22f, 24 magnetoencephalography, 233
cortical plasticity and reorganization, cuneus, 22f, 25 positron emission tomography, 233
170 gyrus rectus, 23, 24f transcranial magnetic stimulation,
epilepsy, 170 lingual gyrus, 22f, 25 234–35
general considerations in pediatrics, medial frontal gyrus, 24 concordance across methods, 231
169–70 paracentral lobule, 24 electrocorticographic spectral analysis,
pitfalls, 50, 51f precuneus, 22f, 24 156–57
sensorimotor cortex, 195–96 supplementary motor area, 24 historical perspectives, 219–20
techniques, 45 and posterior basal temporal sulci, local field potential recordings, 230–31
visual cortex, 227–28, 232–33 24f, 25 neuroanatomy, 220–23
vs. Wada testing, 272, 273t, 274 neurophysiology, 220–23
Functional mapping techniques H noninvasive neuroimaging techniques
extraoperative Hearing functional MRI, 227–28
cortical stimulation, 96, 96f advantages and pitfalls, 262 magnetoencephalography, 229
electronic principles, 95 clinical applications, 257–59 positron emission tomography,
historical perspectives, 93–94 acoustic processing, 259 228–29
instrumentation, 95 speech and language processing, transcranial magnetic stimulation,
vs. intraoperative, 98–99 260–62 229–30
language task, 96 concordance across methods, 255, 257 visually evoked potentials, 229
methodology, 94 direct cortical stimulation, 251–53 operative anatomy, 225–27
saftey and adverse effects, 97–98 functional imaging methods optical imaging in animals and humans,
starting parameters, 95t frequency, 246 134–35
subject preparation, 94 sound level, 247 organizational principles, 223–24
video-EEG, 96, 98t sound localization, 249 structural imaging, 225–27
intraoperative temporal aspects of sound, 248–49
anesthesia, 104 gross anatomy, 244 I
in clinical practice, 106 local field potential recordings, 253–55 ICP. See Intracranial pressure
craniotomy, 104 localization of, 8–9, 9f iCSM. See Intraoperative CSM
electrocorticography, 105 neuroanatomy and neurophysiology, Imaging pathology, animals and humans
vs. extraoperative, 98–99 241–44 epilepsy, 139–40
language mapping, 105–6 noninvasive recordings intracranial hematomas, 143–45
INDEX 281

stroke, 141–43 visual perception, 60 cortical structural anatomy, 4


traumatic brain injury, 143–45 working memory, 60 language function, 5, 7–8, 7f
Implicit memory, 59, 64–65 IQ. See Intelligence quotient vs. lateralization in language, 203–4
Inferior frontal sulcus, 15–16 motor and sensory functions, 4–5, 7f
Inferior precentral sulcus, 15 K visual function, 8, 9f
Insula of Reil, 19 Ketamine, 111 LRFs. See Language-related brain magnetic
Intelligence, 59 fields
Intelligence quotient (IQ) L
full scale, 59 Language M
performace, 59 clinical applications Magnetic source imaging, 229
verbal, 59 basal temporal language area, 213 Magnetoencephalography (MEG), 119
verbal vs. performance, 62 multilingual patients, 212–13 auditory evoked magnetic fields, 123
Intracarotid amobarbital testing notion of degeneracy, 213–14 vs. electroencephalography, 119
advantages and pitfalls, 87–88 young children, 212 evoked magnetic activity, 122
applications, 86 cortical substrate, 204–7 language-related brain magnetic fields
in clinical practice, 87 direct cortical stimulation, 207–10 clinical indications, 124
data acquisition and data analysis functional imaging mapping expressive language-specific cortex,
language, 84–85 vs. cortical stimulation mapping, 125–26
memory, 85–86 210–11 hemispheric dominance, 124–25
historical perspectives, 79–80 modalities, 210 movement-related magnetic fields, 123
instrumentation methods, 81–83 historical background, 203 pediatric brain mapping
invasive pediatric brain mapping, 177 intracranial electrodes, 211–12 epilepsy, 173–74
principles, 79–80 intraoperative cortical mapping, 105–6 functional mapping, 172–73
recording techniques, 83–84 lateralization versus localization, 203–4 general considerations in pediatrics,
strengths and weakness, 87 local field potential recordings, 211–12 171–72
subject preparation methods, 80–81 localization, 5, 7–8, 7f sensorimotor cortex, 196–97
validation techniques, 86–87 neuropsychological testing, 60 somatosensory evoked magnetic fields,
Intracarotid Amytal test. See Wada testing structural imaging mapping 123
Intracranial hematomas, 143–45 Broca’s morphology, 211 spontaneous magnetic activity, 120–22
Intracranial pressure (ICP), 143 DTI tractography, 211 visual cortex, 223, 229
Intracranial recordings, 271 subcortical substrate, 204–7 visual evoked magnetic fields, 123
Intraoperative cortical mapping, 103 temporal lobe localization, 65 Mapping functional connectivity, 159
in clinical practice, 106 Wada test, 84–85 Matching pursuit (MP) algorithm, 155
vs. extraoperative cortical mapping, 98–99 Language cortex Material specificity hypothesis, 57
operating room methods electrocorticographic spectral analysis, Medial frontal gyrus, 24
anesthesia, 104 157–58 Medial occipitotemporal sulcus, 25
craniotomy, 104 optical imaging in animals and humans, MEG. See Magnetoencephalography
electrocorticography, 105 137, 139 Memory
language mapping, 105–6 Language-related brain magnetic fields anatomy, 270
positioning, 104 (LRFs) assessment techniques
sensorimotor stimulation, 105 clinical indications, 124 cerebral amobarbital, 271
somatosensory evoked potential expressive language-specific cortex, cortical stimulation, 271
recording, 104 125–26 functional imaging, 270
postoperative outcomes, 106 hemispheric dominance, 124–25 intracranial recordings, 271
principles, 103 Lateral occipitotemporal sulcus, 25 noninvasive assessments, 270–71
Intraoperative cortical stimulation, 177–78 Lateral sulcus, 13–15, 14f clinical applications
Intraoperative CSM (iCSM), 207 Lateralization versus localization, 203–4 functional MRI, 272, 273t
Intraoperative monitoring, 114 Lesion neurological testing method, functional MRI vs. Wada testing, 272,
Intraoperative seizures, 104 57–58 273t, 274
Intraparietal sulcus, 16, 16f LFPs. See Local field potentials Wada testing, 272, 272t
Intrinsic brain tumors, 198–99 Lingual gyrus, 22f, 25 definition, 269
Invasive pediatric brain mapping techniques Local anesthetics, 110 electrocorticographic spectral analysis,
awake craniotomy, 180 Local field potentials (LFPs) 156–57
electrocorticography, 180 hearing, 253–55 episodic, 269
extraoperative cortical stimulation, language, 211–12 definition, 269
178–79 visual cortex, 230–31, 235 nonverbal, 63–64, 64t
intraoperative cortical stimulation, 177–78 Localization verbal, 62–63, 63f
vs. noninvasive techniques, 176 auditory function, 8–9, 9f implicit, 64–65
Wada testing, 177 cerebral, using fMRI neuropsychological testing, 59–60, 60f
Investigation tools, neuropsychological activation map creation, 39, 40f procedural, 269
testing clinical inferences, 41–44, 43f Wada test, 85–86
attention, 60 data analysis, 33–39, 36f, 38f working, 60, 66, 269
cognitive speed, 60–61 MRI and MR scanners, 33, 34f Mesial temporal sclerosis (MTS), 62
executive functions, 61 MRI data collection, 33, 35f Methohexital, 88
intelligence, 59 thumbnail sketch, 31–33, 32f MNI. See Montreal Neurological Institute
language, 60 visualization on cortical surface, Monopolar stimulation, 95
memory, 59–60, 60f 39–41, 40f, 42f Montreal Neurological Institute (MNI), 79
282 INDEX

Motor and sensory functions surgical navigation, 46–47, 48f Optical spectroscopic imaging method
anatomical localization, 4–5, 7f surgical planning, 46, 47f diffuse optical tomography, 133
central cortex, 20 magnetoencephalography imaging architecture in animals and
Motor cortical stimulation, 199 auditory evoked magnetic fields, 123 humans, 133–34
Motor hand function, 5, 7f vs. electroencephalography, 119 auditory cortex, 135
Movement-related magnetic fields (MRFs), evoked magnetic activity, 122 language cortex, 137, 139
123 language-related brain magnetic somatosensory cortex, 135–37, 136f
Moyamoya disease, 176 fields, 124–26 visual cortex, 134–35
MRFs. See Movement-related magnetic fields movement-related magnetic fields, near infrared spectroscopy, 133
MRI scanners, 33 123 neurovascular coupling, 131–32
MTS. See Mesial temporal sclerosis pediatric brain mapping, 172–74 optical recording of intrinsic signal,
Multilingual patients, 212–13 sensorimotor cortex, 196–97 132–33, 132f
Multivariate generalized linear hypothesis, 35 somatosensory evoked magnetic pathology in animals and humans
fields, 123 epilepsy, 139–40
N spontaneous magnetic activity, 120–22 intracranial hematomas, 143–45
Near infrared spectroscopy (NIRS), 133, visual cortex, 223 stroke, 141–43
144f visual evoked magnetic fields, 123 traumatic brain injury, 143–45
Neurologic specificity, 56 Neurovascular coupling, 131–32 physiological characteristics, 131
Neuropathic pain, 199 NIRS. See Near infrared spectroscopy Orientation preference, 224
Neuropharmacology, volatile anesthetic, 113 Nitrous oxide, 112–13 ORIS. See Optical recording of intrinsic
Neuropsychological testing Noninvasive imaging techniques signal
data analysis, 61 pediatric brain mapping Oxygen-15 (O-15), 228
data interpretation, 61–62 diffusion tensor imaging, 171
evaluation methods, 58–59 diffusion-weighted imaging, 170–71 P
historical perspective, 55–56 functional MRI, 169–70 Paracentral lobule, 24
investigation tools vs. invasive mapping, 168–69 Parallel processing technique, 223f, 224
attention, 60 magnetoencephalography, 172–74 Parallel sulcus, 16
cognitive speed, 60–61 perfusion-weighted imaging, 171 Parietal lobe
executive functions, 61 positron emission tomography, cerebral hemisphere, 18, 18f
intelligence, 59 174–75 neuropsychological testing, 67, 69
language, 60 single photon emission tomography, Parieto-occipital sulcus, 22–23
memory, 59–60, 60f 175–76 Pediatric brain mapping
visual perception, 60 visual cortex clinical indications, 167
working memory, 60 functional MRI, 227–28 diffusion tensor imaging, 171
principles magnetoencephalography, 229 diffusion-weighted imaging, 170–71
clinical mapping, 56–57 positron emission tomography, functional MRI
lesion method, 57–58 228–29 brain tumors, 170
subject preparation methods, 59 transcranial magnetic stimulation, cortical plasticity and reorganization,
Neuropsychological testing applications 229–30 170
case study, 70–71 visually evoked potentials, 229 epilepsy, 170
frontal lobe localization Noninvasive recordings general considerations in pediatrics,
attention, 66 gamma range oscillations, 251 169–70
design and verbal fluency, 66–67 surface electrical recordings, 249–51 invasive techniques
executive functioning, 67, 68f Nonverbal memory, 63–64, 64t awake craniotomy, 180
working memory, 66 Notion of degeneracy, 213–14 electrocorticography, 180
left versus right hemisphere Nyquist–Shannon sampling theorem, extraoperative cortical stimulation,
global versus local processing of 154 178–79
information, 62 intraoperative cortical stimulation,
verbal versus performance IQ, 62 O 177–78
parietal and occipital lobes localization, Occipital lobe vs. noninvasive techniques, 176
67, 69 cerebral hemisphere, 18f, 19 Wada testing, 177
strengths and weaknesses, 69–70 neuropsychological testing, 67, 69 magnetoencephalography
temporal lobe localization Ocular dominance, 224 epilepsy, 173–74
episodic nonverbal memory, 63–64, Ohm’s law, 95 functional mapping, 172–73
64t Olfaction, 65, 66t general considerations in pediatrics,
episodic verbal memory, 62–63, 63f Operating room methods, intraoperative 172
face processing, 65 mapping perfusion-weighted imaging, 171
implicit memory, 64–65 craniotomy, 104 positron emission tomography
language, 65 electrocorticography, 105 brain tumors, 174
olfaction, 65, 66t language mapping, 105–6 epilepsy, 174–75
Neuropsychology, 55 positioning, 104 functional mapping, 174
Neurosurgical applications sensorimotor stimulation, 105 general considerations in pediatrics,
functional MRI somatosensory evoked potential 174
arteriovenous malformations, 48 recording, 104 single photon emission tomography
brain tumors, 47–48, 47f, 49f Opiates, 111 additional clinical applications,
epilepsy, 48, 50 Optical recording of intrinsic signal (ORIS), 176
stereotactic radiosurgery, 50, 50f 132–33, 132f, 138f, 140f brain tumors, 175
INDEX 283

epilepsy, 175–76 Sensorimotor functional unit, 19 paracentral lobule, 24


general considerations in pediatrics, Sensorimotor stimulation, 105 precuneus, 22f, 24
175 Short-time directed transfer function (SDTF), supplementary motor area, 24
specific techniques, 167–68, 168f 159 inferior and mesial surface, 24f, 25
Performance IQ (PIQ), 62 Single dissociation, 57–58 inferior frontal sulcus, 15–16
Perfusion-weighted imaging (PWI), 171 Single photon emission tomography (SPECT) inferior precentral sulcus, 15
Peripheral vision, 8 noninvasive pediatric brain mapping insula of Reil, 19
PET. See Positron emission tomography additional clinical applications, 176 intraparietal sulcus, 16, 16f
Pfeifer’s norm, 244 brain tumors, 175 lateral sulcus, 13–15, 14f
Phase encoding procedure, 227 epilepsy, 175–76 lateral surface, 13
Phonological loop, 158 general considerations in pediatrics, gyri, 15f, 17
PIQ. See Performance IQ 175 limbic lobe region, 25–28, 26f
Positron emission tomography (PET) Single quadrant stimulation, 123 mesial surface, 22, 22f
noninvasive pediatric brain mapping Somatosensory cortex mesial temporal region, 25–28, 26f
brain tumors, 174 optical imaging in animals and humans, occipital lobe, 18f, 19
epilepsy, 174–75 134–35 parietal lobe, 18, 18f
functional mapping, 174 Somatosensory evoked magnetic fields parieto-occipital sulcus, 22–23
general considerations in pediatrics, (SEFs), 123 posterior speech area, 18f, 21–22
174 Somatosensory evoked potential recording, precentral sulcus, 14f, 16
sensorimotor cortex, 196 104 premotor cortex, 20
visual cortex, 228–29, 233 Somatosensory-evoked potentials (SSEPs), rostral sulci, 22, 22f
Posterior basal temporal sulci, 24f, 25 194–95 sensorimotor cortex
Precentral sulcus, 14f, 16 Sound postcentral gyrus, 20
Precuneus, 24 level, 247 precentral gyrus, 19–20
Premotor cortex, 20 localization, 249 striate visual cortex, 23
Primary brain sulci, 13, 14t temporal aspects of, 248–49 superior frontal sulcus, 16, 16f
Primary visual cortex, 8, 9f, 221f, 224 SPECT. See Single photon emission superior temporal sulcus, 16–17
Procedural memory, 269 tomography sylvian opercula, 13–15, 14f
Propofol, 112 Speech temporal lobe, 15f, 18–19
PWI. See Perfusion-weighted imaging localization of, 7–8, 8f Subcortical substrate, 204–7
processing applications, 260–62 Subdural electrodes (SDEs), 94, 212
R SSEPs. See Somatosensory-evoked potentials Subdural grids, 94
Radiosurgery, stereotactic, 50, 50f Stereotactic radiosurgery, 50, 50f Subdural strips, 94
Retinotopic mapping techniques, 223 Stereotactically implanted depth electrodes, Subject preparation methods
Retrieval, 269 153 extraoperative functional cortical
RFFT. See Ruff figural fluency test Stimulation mapping, 94
Rolandic cortex, 198–99 auditory, 43f, 137, 250 neuropsychological testing, 59
Rostral sulci, 22, 22f bipolar, 95 Wada testing, 80–81
Ruff figural fluency test (RFFT), 61 cortical, 271 Subparietal sulcus, 24
electrical, 69 Superior frontal sulcus, 16, 16f
S epidural, 98 Superior temporal sulcus, 16–17
SDEs. See Subdural electrodes Hemifield, 123 Supplementary motor area, 24
SDTF. See Short-time directed transfer monopolar, 95 Surface electrical recordings, 249–51
function motor cortical, 199 Surgical navigation, 46–47, 48f
Secondary brain sulci, 13, 14t sensorimotor, 105 Surgical planning, 46, 47f
Sedatives, 109 single quadrant, 123 Sylvian fissure, 4
SEFs. See Somatosensory evoked magnetic tactile, 123 Sylvian opercula, 13–15, 14f
fields visual, 37, 43f
Semantic memory, 269 Storage, 269 T
Sensorimotor cortex Striate visual cortex, 23 Tactile stimulation, 123
cerebral hemisphere Stroke, 141–43 Taylor–Haughton lines, 4, 4f
postcentral gyrus, 20 Structural brain imaging regions TBI. See Traumatic brain injury
precentral gyrus, 19–20 anterior speech area, 17f, 20–21 Temporal lobe
clinical applications calcarine sulcus, 23 cerebral hemisphere, 15f, 18–19
motor cortical stimulation, 199 central sulcus, 15, 15f neuropsychological testing
Rolandic cortex, 198–99 cingulate sulcus, 22, 22f episodic nonverbal memory, 63–64,
direct cortical stimulation, 192–94 gyri 64t
electrocorticographic spectral analysis, and anterior basal orbitofrontal sulci, episodic verbal memory, 62–63, 63f
156–57 24f, 25 face processing, 65
functional MRI, 195–96 and posterior basal temporal sulci, implicit memory, 64–65
historical perspectives, 189–90 24f, 25 language, 65
localization, 191–92 gyri of mesial surface olfaction, 65, 66t
magnetoencephalography, 196–97 cingulate gyrus, 22f, 24 Temporal lobe epilepsy (TLE), 79
positron emission tomography, 196 cuneus, 22f, 25 Tertiary brain sulci, 13, 14t
primary motor cortex, 190–91 gyrus rectus, 23, 24f TLE. See Temporal lobe epilepsy
somatosensory-evoked potentials, 194–95 lingual gyrus, 22f, 25 TMS. See Transcranial magnetic stimulation
transcranial magnetic stimulation, 197 medial frontal gyrus, 24 Tongue sensory function, 5, 7f
284 INDEX

Tonotopic organization, 241 functional MRI, 232–33 Visually evoked potentials (VEPs), 229,
Tractography, 227f, 234f local field potential recordings, 235 233–34
Traditional connectionists, 56 magnetoencephalography, 233 Volatile anesthetic neuropharmacology, 113,
Traditional generalists, 56 positron emission tomography, 233 113t
Traditional localizationists, 56 transcranial magnetic stimulation,
Transcranial magnetic stimulation 234–35 W
(TMS) concordance across methods, 231 Wada testing
sensorimotor cortex, 197 direct cortical stimulation, 230 advantages and pitfalls, 87–88
visual cortex, 229–30, 234–35 electrocorticographic spectral analysis, applications, 86
Traumatic brain injury (TBI), 143–45 156–57 in clinical practice, 87
historical perspectives, 219–20 data acquisition and data analysis
V local field potential recordings, 230–31 language, 84–85
VEFs. See Visual evoked magnetic fields neuroanatomy, 220–23 memory, 85–86
VEPs. See Visually evoked potentials neurophysiology, 220–23 historical perspectives, 79–80
Verbal fluency, 66–67 noninvasive neuroimaging techniques instrumentation methods, 81–83
Verbal IQ (VIQ), 59, 62 functional MRI, 227–28 invasive pediatric brain mapping,
Verbal memory, 62–63, 63f magnetoencephalography, 229 177
Vertical ramus, 15 positron emission tomography, principles, 79–80
Video-EEG, 96, 98t 228–29 recording techniques, 83–84
VIQ. See Verbal IQ transcranial magnetic stimulation, strengths and weakness, 87
Vision 229–30 subject preparation methods, 80–81
definition, 219 visually evoked potentials, 229 validation techniques, 86–87
localization, 8, 9f operative anatomy, 225–27 Warrington’s recognition memory test, 59
Visual adaptation, 231 optical imaging in animals and humans, Wechsler adult intelligence scale, 59
Visual cortex 134–35 Wernicke’s area, 8, 8f
advantages and pitfalls, 235–36 organizational principles, 223–24 Wisconsin card sorting test, 61
clinical applications structural imaging, 225–27 Working memory, 60, 66, 269
direct cortical stimulation, 235 Visual evoked magnetic fields (VEFs), 123
event-related scalp visual evoked Visual perception, 60 Y
potential, 233–34 Visual stimulation, 37, 43f Young children, language mapping, 212

You might also like