You are on page 1of 284

Intracranial Stereotactic Radiosurgery

2nd Edition

L. Dade Lunsford, MD, FACS


Lars Leksell Professor and Distinguished Professor
Department of Neurological Surgery
Director
Center for Image Guided Neurosurgery
Director
Residency Training Program
University of Pittsburgh
Pittsburgh, Pennsylvania

Jason P. Sheehan, MD, PhD, FACS


Harrison Distinguished Professor
Vice Chair of Neurological Surgery
Professor of Radiation Oncology and Professor of Neuroscience
Department of Neurological Surgery
Director
UVA Gamma Knife Center
University of Virginia School of Medicine
Charlottesville, Virginia

Thieme
New York • Stuttgart • Delhi • Rio de Janeiro
Executive Editor: Timothy Y. Hiscock Important note: Medicine is an ever-changing science undergoing
Managing Editor: Sarah Landis continual development. Research and clinical experience are contin-
Director, Editorial Services: Mary Jo Casey ually expanding our knowledge, in particular our knowledge of proper
Editorial Assistant: Nikole Connors treatment and drug therapy. Insofar as this book mentions any dosage
Production Editor: Sean Woznicki or application, readers may rest assured that the authors, editors, and
International Production Director: Andreas Schabert publishers have made every effort to ensure that such references are
Vice President, Editorial and E-Product Development: Vera Spillner in accordance with the state of knowledge at the time of production
International Marketing Director: Fiona Henderson of the book.
International Sales Director: Louisa Turrell Nevertheless, this does not involve, imply, or express any guarantee
Director of Sales, North America: Mike Roseman or responsibility on the part of the publishers in respect to any dosage
Senior Vice President and Chief Operating Officer: Sarah Vanderbilt instructions and forms of applications stated in the book. Every user is
President: Brian D. Scanlan requested to examine carefully the manufacturers’ leaflets accompa-
nying each drug and to check, if necessary in consultation with a
Library of Congress Cataloging-in-Publication Data physician or specialist, whether the dosage schedules mentioned there-
in or the contraindications stated by the manufacturers differ from the
Intracranial stereotactic radiosurgery / [edited by] L. Dade
statements made in the present book. Such examination is particularly
Lunsford, Jason P. Sheehan. – 2nd edition.
important with drugs that are either rarely used or have been newly
p. ; cm.
released on the market. Every dosage schedule or every form of appli-
Includes bibliographical references and index.
cation used is entirely at the user’s own risk and responsibility. The
Summary: "Written by international experts in the fields of neuro-
authors and publishers request every user to report to the publishers
surgery, neurology, physics, and radiation oncology, many of them
any discrepancies or inaccuracies noticed. If errors in this work are
pioneers in radiosurgery, the book is a concise yet comprehensive
found after publication, errata will be posted at www.thieme.com on
reference on current techniques for radiosurgery of the brain and all
the product description page.
the contextual information needed to understand why and how these
Some of the product names, patents, and registered designs
current techniques are used, such as the evolution of the radiosurgery
referred to in this book are in fact registered trademarks or proprietary
modality, the sciences of radiosurgery such as physics and radiobiology,
names even though specific reference to this fact is not always made in
and the devices/technology systems used. The core of the book is the
the text. Therefore, the appearance of a name without designation as
detailed discussion of radiosurgical procedures for specific pathologies
proprietary is not to be construed as a representation by the publisher
and disorders, such as arteriovenous malformations, meningiomas,
that it is in the public domain.
pituitary adenomas, movement disorders, gliomas, ocular disorders,
and obsessive compulsive disorder. These chapters address the benefits
and limitations of radiosurgical techniques for each indication and
describe the outcomes, possible complications and their management,
and alternative treatments"–Provided by publisher.
ISBN 978-1-62623-032-3 (hardback) – ISBN 978-1-62623-033-0
(eISBN)
I. Lunsford, L. Dade, editor. II. Sheehan, Jason P., editor.
[DNLM: 1. Brain Diseases–surgery. 2. Radiosurgery–methods. 3.
Brain Diseases–radiotherapy. 4. Stereotaxic Techniques. WL 368]
RD594.15
617.4'81059–dc23
2015002877

© 2016 Thieme Medical Publishers, Inc.

Thieme Publishers New York


333 Seventh Avenue, New York, NY 10001 USA
+1 800 782 3488, customerservice@thieme.com

Thieme Publishers Stuttgart


Rüdigerstrasse 14, 70469 Stuttgart, Germany
+49 [0]711 8931 421, customerservice@thieme.de

Thieme Publishers Delhi


A-12, Second Floor, Sector-2, Noida-201301
Uttar Pradesh, India
+91 120 45 566 00, customerservice@thieme.in

Thieme Publishers Rio de Janeiro, Thieme Publicações Ltda.


Edifício Rodolpho de Paoli, 25º andar
Av. Nilo Peçanha, 50 – Sala 2508
Rio de Janeiro 20020-906, Brasil
+55 21 3172 2297 / +55 21 3172 1896

Cover design: Thieme Publishing Group


Typesetting by DiTech Process Solutions
This book, including all parts thereof, is legally protected by copyright.
China by Everbest Printing Co, Ltd 54321 Any use, exploitation, or commercialization outside the narrow limits
set by copyright legislation without the publisher’s consent is illegal and
ISBN 978-1-62623-032-3
liable to prosecution. This applies in particular to photostat reproduc-
Also available as an e-book: tion, copying, mimeographing or duplication of any kind, translating,
eISBN 978-1-62623-033-0 preparation of microfilms, and electronic data processing and storage.
Contents

Foreword. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . viii

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix

Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . x

Part I History

1 The Origins and Birth of the Leksell Gamma Knife . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2


Dan Leksell and L. Dade Lunsford

2 The History of Linac and Proton Beam Radiosurgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11


Laura E. G. Warren, Brian Winey, Jay S. Loeffler, and Helen A. Shih

3 The History of CyberKnife Radiosurgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16


Aditya Iyer, Steven D. Chang, and John R. Adler, Jr.

Part II Radiosurgical Fundamentals

4 The Radiobiology of Radiosurgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20


John C. Flickinger

5 The Physics of Radiosurgery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30


David Schlesinger, Brian Wang, and Stanley H. Benedict

6 Radiosurgical Devices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
Daniel E. Hyer, Frank J. Bova, and John Buatti

7 Critical Structures and Tolerance of the Central Nervous System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52


Siavash Jabbari, Lijun Ma, Young K. Lee, Simon S. Lo, Eric L. Chang, Jimm Grimm, Lance Altenau, Daniel White,
Vikram Udani, Steven J. Goetsch, David Larson, and Arjun Sahgal

8 The Neuropathology of Radiosurgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58


György Szeifert, Douglas Kondziolka, Marc Levivier, José Lorenzoni, and L. Dade Lunsford

Part III Vascular Indications

9 Stereotactic Radiosurgery for Arteriovenous Malformations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68


L. Dade Lunsford, Ajay Niranjan, Hideyuki Kano, Edward A. Monaco III, and John C. Flickinger

10 Stereotactic Radiosurgery for Cavernomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81


Gabor Nagy and Andras A. Kemeny

11 Stereotactic Radiosurgery for Dural Arteriovenous Fistulas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91


David Hung-Chi Pan, Wen-Yuh Chung, Huai-Che Yang, and Cheng-Chia Lee

v
Contents

Part IV Benign Tumor Indications

12 Stereotactic Radiosurgery for Meningiomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102


William Friedman and Frank J. Bova

13 Stereotactic Radiosurgery for Pituitary Adenomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107


Cheng-Chia Lee and Jason P. Sheehan

14 Stereotactic Radiosurgery for Chordomas. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120


Dan Kunaprayoon, Ananth Charya, Jay S. Loeffler, and Helen A. Shih

15 Stereotactic Radiosurgery for Chondrosarcomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128


Hideyuki Kano, Aditya Iyer, and L. Dade Lunsford

16 Stereotactic Radiosurgery for Glomus Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132


Ajay Niranjan, Edward A. Monaco III, Hideyuki Kano, John C. Flickinger, and L. Dade Lunsford

17 Stereotactic Radiosurgery for Nonvestibular Schwannomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137


Edward A. Monaco III, Gurpreet Surinder Gandhoke, Ajay Niranjan, Hideyuki Kano, and L. Dade Lunsford

18 Stereotactic Radiosurgery for Hemangioblastomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146


Hideyuki Kano and L. Dade Lunsford

19 Stereotactic Radiosurgery for Vestibular Schwannomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150


Jean Régis, Romain Carron, Christine Delsanti, Denis Porcheron, Jean-Marc Thomassin, Xavier Muracciole, and
Pierre-Hugues Roche

Part V Functional Indications

20 Stereotactic Radiosurgery for Trigeminal Neuralgia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 160


Michael D. Chan and Stephen B. Tatter

21 Stereotactic Radiosurgery in Movement Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168


Patrick Pezeshkian, Antonio De Salles, and Nader Pouratian

22 Gamma Knife Radiosurgery for Obsessive–Compulsive Disorder . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177


Nrupen Baxi, Andrew Brunswick, Eric Mazel, and Douglas Kondziolka

23 Stereotactic Radiosurgery for Hypothalamic Hamartomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182


Andrew G. Shetter, Heyoung L. McBride, and John F. Kerrigan

24 Stereotactic Radiosurgery for Epilepsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193


Mark Quigg and Nicholas M. Barbaro

Part VI Ocular Disorder and Pediatric Tumor Indications

25 Stereotactic Radiosurgery for Ocular Disorders. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202


Roman Liscak and Gabriela Simonova

vi
Contents

26 Stereotactic Radiosurgery for Pediatric Brain Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210


Christian C. Okoye, Ravi B. Patel, David B. Mansur, Alia Hdeib, Arjun Sahgal, Eric L. Chang, Mitchell Machtay,
John H. Suh, Andrew E. Sloan, and Simon S. Lo

Part VII Malignant Tumor Indications

27 Radiosurgery for Glial Tumors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226


Jason Lee Schroeder, John H. Suh, Michael A. Vogelbaum, and Gene H. Barnett

28 Stereotactic Radiosurgery for the Management of One to Four Brain Metastases . . . . . . . . . . 235
Sten Myrehaug, Simon S. Lo, Siavash Jabbari, Lijun Ma, Sunit Das, Aliaksandr Karotki, Eric L. Chang, and Arjun Sahgal

29 Stereotactic Radiosurgery for the Management of Five or More Brain Metastases. . . . . . . . . . 243
Christopher L. Tinkle, Steve Braunstein, Penny K. Sneed, Igor J. Barani, and David Larson

30 Repeat Radiosurgery for Brain Metastases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251


Aditya Halthore, Ami B. Raval, Michael Schulder, and Jonathan Knisely

Part VIII Radiosurgical Horizon

31 Promising Advances in Radiosurgery: Where Are the Frontiers? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 258


Brian D. Kavanagh

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260

vii
Foreword
The first edition of this book was published in 2009, and I ing those on epilepsy, ocular disorders and pediatric brain
am honored to have been asked again to write the fore- tumors.
word to this second edition of Intracranial Stereotactic As we live through our daily clinical routines we often
Radiosurgery. think that we have reached the outer limits of what we can
In December of 1985 there were three Gamma Knives in do for our patients, only to soon discover, again and again,
the world, one each in Stockholm, Buenos Aires and Shef- that there is more we can do. We now know more about the
field. During Christmas of that year, a few weeks before my use of radiosurgery for conditions such as OCD and hypo-
father Lars suddenly passed away, I asked him how many thalamic hamartomas, both of which are covered in new
more Gamma Knives he thought there would be in the chapters. And the final chapter by Brian D. Kavanagh is
world. His laconic answer was "maybe one or two more." thought-provoking; while I don't see charged particles
He was usually correct in his assessments of things but this striking back in a big way in today’s increasingly cash-
time, in retrospect, he could not have been more wrong! strapped health care systems, I'm a firm believer in his
Between 1968 and 1985 approximately 1,300 patients in other three predictions.
Sweden, Argentina and England underwent Gamma Knife In 1969, in my father’s stead, I travelled to a meeting in
Surgery. In 2009, when I wrote the foreword for the first India to talk about the first 7 or 8 patients we had treated in
edition of this book, the number of patients treated had Stockholm. After the talk there was uproar in the room. It
increased to around 500,000. Six years later, as this second simply could not be true that you could do things in the
edition is published, the number of patients treated by brain without a craniotomy! The chairman of the meeting
Gamma Knives will be approaching one million. An gently but firmly led me out of the lecture hall—maybe for
unknown but likely very large number of additional my own safety?
patients have been treated by other radiosurgery technol- Now thinking back at my personal involvement with
ogies, including protons, Cyberknife and other linac sys- stereotactic radiosurgery in different capacities over the
tems. Similarly, in 1985 there were just 117 published last 46 years, I cannot feel anything but astonishment at the
papers on radiosurgery; a simple search on PubMed today incredible developments we have seen and to which so
yields many thousands of peer-reviewed radiosurgery many have contributed. I'm also both humbled and proud of
papers. the fact that so many patients have been helped by non-
Given this continued growth in radiosurgical procedures invasive brain surgery—despite having been regarded as a
and published science, it is not surprising that in a mere six charlatan at that meeting in India years ago.
years there is so much more that has been learned that it This volume is richly and interestingly illustrated and
warrants an update of the previous excellent edition of this truly a joy to read. The authors of all chapters have made
work. That is what this wonderfully comprehensive tome important contributions to the literature on intracranial
accomplishes with distinction. radiosurgery and, like its predecessor, this volume has been
The growing volume of knowledge is reflected not only elegantly compiled and edited by L. Dade Lunsford and
by the many more pages and chapters in this revised Jason P. Sheehan. We owe them both thanks for bringing
edition, but also by the quantity of new data reported in us this comprehensive update on all aspects of the field.
each section. The history of the development of the field This is a work of reference for students, neurosurgery
radiosurgery is now nicely complemented by a description residents and seasoned radiosurgery practitioners alike. It
of the development of technologies other than the Gamma deserves to occupy a prominent place in our bookshelves—
Knife. The all-important and fundamental topic of the or even better—on our desks!
radiobiology of radiosurgery has been updated, and chap-
ters on radiosurgery physics and critical structures and Dan Leksell, MD
brain tolerance have been added. The chapters on individual Stockholm, Sweden
clinical indications have been revisited and updated, includ-

viii
Preface
Since the first edition of Intracranial Stereotactic Radiosur- nated through national and international research groups.
gery published in 2009, substantial advancements have Scientific dialogue and education have been fostered by
been made in the field. This prompted us to begin work on mature radiosurgical user groups. The user groups have
the current edition as a means of detailing contemporary also created journals and held congresses for timely and
radiosurgery and the many advances that had occurred. By frank discussions of radiosurgical advances.
its very nature, radiosurgery is minimally invasive and Intracranial radiosurgery continues to prove disruptive
relies upon sophisticated technology to aid in its delivery. to existing treatment paradigms (e.g. management of brain
The minimally invasive nature led to a more favorable side metastasis). It has also spawned the application of its
effect profile, and this was one of the core principles which principles to extracranial sites such as the spine. The field
drove Leksell to conceive of stereotactic radiosurgery. of radiosurgery appears to be more vibrant than ever. The
Technological advances in neuro-imaging, medical physics, current work provides a comprehensive assessment of
computer science, and engineering have lead to improve- radiosurgery's past and present, and it affords a glimpse
ments in the speed, accuracy, precision, and comfort of of its bright future.
radiosurgical delivery to single or multiple (10+) targets in
a single session. They have also yielded better clinical L. Dade Lunsford, MD, FACS
outcomes. Pittsburgh, Pennsylvania
Beyond technologic gains, the field has improved as a
result of an increased fund of knowledge. Knowledge in this Jason P. Sheehan, MD, PhD, FACS
field has been acquired through multicenter trials coordi- Charlottesville, Virginia

ix
Contributors
John R. Adler, Jr., MD Steve Braunstein, MD, PhD
Dorothy and TK Chan Professor, Emeritus Assistant Professor
Department of Neurosurgery Radiation Oncology
Stanford University University of California–San Francisco
Stanford, California San Francisco, California

Lance Altenau, MD, FACS Andrew Brunswick, MD


Neurosurgical Medical Clinic Resident Physician
San Diego, California Department of Neurosurgery
New York University Langone Medical Center
Igor J. Barani, MD New York, New York
Associate Professor
Departments of Radiation Oncology and Neurological John Buatti, MD
Surgery Professor and Chair
University of California, San Francisco Department of Radiation Oncology
San Francisco, California Professor
Department of Otolaryngology, Neurosurgery
Nicholas M. Barbaro, MD University of Iowa Carver College of Medicine
Betsey Barton Professor and Chairman of Neurological Iowa City, Iowa
Surgery
Indiana University Romain Carron, MD, PhD
Goodman Campbell Brain and Spine Service de Neurochirurgie Fonctionnelle & Stéréotaxique
Indianapolis, Indiana Aix-Marseille Universite
Hôpital Universitaire La Timone
Gene H. Barnett, MD, MBA Marseille, France
Professor and Director
Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Michael D. Chan, MD
Center Assistant Professor
Department of Neurological Surgery Wake Forest School of Medicine
Cleveland Clinic Winston-Salem, North Carolina
Cleveland, Ohio
Eric L. Chang, MD
Nrupen Baxi, MD Professor and Chairman
Neuromodulation / Functional and Stereotactic Department of Radiation Oncology
Neurosurgery Fellow Keck School of Medicine of USC
New York University Langone Medical Center Los Angeles, California
New York, New York
Steven D. Chang, MD
Stanley H. Benedict, PhD Robert C. and Jeannette Powell Professor
Professor and Vice Chair of Clinical Physics Department of Neurosurgery
Department of Radiation Oncology Stanford University School of Medicine
University of California–Davis, Comprehensive Cancer Stanford, California
Center
Sacramento, California Ananth Charya, MD, MS
The Ohio State University College of Medicine
Frank J. Bova, PhD Columbus, Ohio
Principle Investigator
McNight Brain Institute Radiosurgery/Biology Laboratory Wen-Yuh Chung, MD
Albert E. and Birdie W. Einstein Professor of Computer- Department of Neurosurgery
Assisted Stereotactic Neurosurgery Neurological Institute
Department of Neurosurgery Taipei Veterans General Hospital
University of Florida National Yang-Ming University School of Medicine
Gainesville, Florida Beitou, Taipei, Taiwan

x
Contributors

Sunit Das, MD, PhD Alia Hdeib, MD


Division of Neurosurgery Clinical Instructor
St. Michael's Hospital and Li Ka Shing Knowledge Institute Neurosurgery
Labatt Brain Tumour Research Centre, Hospital for Sick Case Western Reserve University School of Medicine
Children Cleveland, Ohio
Toronto, Ontario, Canada
Daniel E. Hyer, PhD
Christine Delsanti, MD Clinical Assistant Professor
Service de Neurochirurgie Fonctionnelle & Stéréotaxique Department of Radiation Oncology
Aix-Marseille Universite University of Iowa Hospitals and Clinics
Hôpital Universitaire La Timone Iowa City, Iowa
Marseille, France
Aditya Iyer, MD
Antonio De Salles, MD, PhD Neurosurgery Resident
Professor in Residence and Director Stanford School of Medicine
Stereotactic Surgery Program Stanford, California
Co-Director
Radiosurgery Program Siavash Jabbari, MD
Neurosurgery Department Department of Radiation Oncology
University of California–Los Angeles Sharp Healthcare
Los Angeles, California San Diego, California

John C. Flickinger, MD
Hideyuki Kano, MD, PhD
Department of Radiation Oncology
Research Associate Professor
University of Pittsburgh Medical Center
Department of Neurological Surgery
Pittsburgh, Pennsylvania
University of Pittsburgh
University of Pittsburgh Medical Center-Presbyterian
William Friedman, MD
Pittsburgh, Pennsylvania
Professor and Chairman
Department of Neurosurgery
University of Florida Aliaksandr Karotki, PhD
Gainesville, Florida Department of Medical Physics
Odette Cancer Centre
Gurpreet Surinder Gandhoke, MD, MCh Sunnybrook Health Sciences Centre
PGY-4 Resident Toronto, Ontario, Canada
Center for Image-Guided Neurosurgery
Department of Neurological Surgery Brian D. Kavanagh, MD, MPH, FASTRO
Professor and Interim Chair
University of Pittsburgh Medical Center-Presbyterian
Department of Radiation Oncology
Pittsburgh, Pennsylvania
University of Colorado Denver School of Medicine
Anschutz Medical Campus
Steven J. Goetsch, PhD, FAAPM
Director of Medical Physics Aurora, Colorado
San Diego Gamma Knife Center
La Jolla, California Andras A. Kemeny, MD, FRCS
Consultant Neurosurgeon
Jimm Grimm, PhD Thornbury Radiosurgery Centre
Bott Cancer Center BMI Thornbury Hospital
Holy Redeemer Hospital Sheffield, South Yorkshire, United Kingdom
Meadowbrook, Pennsylvania

Aditya Halthore, MD
Resident
Department of Radiation Medicine
Hofstra North Shore LIJ School of Medicine
Manhasset, New York

xi
Contributors

John F. Kerrigan, MD Dan Leksell, MD


Director Chairman
Pediatric Epilepsy and Clinical Neurophysiology Programs Leksell Gamma Knife Society
Division of Pediatric Neurology and Comprehensive Stockholm, Sweden
Epilepsy Center
Hypothalamic Hamartoma Program Marc Levivier, MD, PhD
Barrow Neurological Institute at Phoenix Children’s Professor & Chairman
Hospital Department of Neurosurgery & Gamma Knife Center
Phoenix, Arizona Centre Hospitalier Universitaire Vaudois (CHUV)
Lausanne, Switzerland
Jonathan Knisely, MD
Chief, Division of Radiosurgery and Stereotactic Program, Roman Liscak, MD, PhD
Department of Radiation Medicine Associate Professor
Co-Director, Center for Stereotactic Radiosurgery and Head of the Stereotactic and Radiation Department
Stereotactic Radiation Therapy Na Homolce Hospital
Hofstra North Shore LIJ School of Medicine Prague, Czech Republic
Manhasset, New York
Simon S. Lo, MD, FACR
Associate Professor of Radiation Oncology
Douglas Kondziolka, MD, MSc, FRCSC, FACS
Director of Radiosurgery Services and Neurologic Radiation
Professor
Oncology
Department of Neurosurgery and Radiation Oncology
University Hospitals Seidman Cancer Center
Vice-Chair
Case Western Reserve University
Clinical Research (Neurosurgery)
Cleveland, Ohio
Director
Center for Advanced Radiosurgery Jay S. Loeffler, MD
New York University Langone Medical Center Professor and Chair
New York, New York Chief
Department of Radiation Oncology
Dan Kunaprayoon, MD Herman and Joan Suit Professor
Department of Radiation Oncology Department of Radiation Oncology
University of Cincinnati College of Medicine Massachusetts General Hospital
Cincinnati, Ohio Boston, Massachusetts

David Larson, MD, PhD, FACR, FASTRO José Lorenzoni, MD


Professor Departamento de Neurocirurgia
Departments of Radiation Oncology and Neurological Pontificia Universidad Católica de Chile and Centro Gamma
Surgery Knife de Santiago
University of California-San Francisco Santiago, Chile
San Francisco, California
L. Dade Lunsford, MD, FACS
Cheng-Chia Lee, MD Lars Leksell Professor and Distinguished Professor
Department of Neurosurgery Department of Neurological Surgery
Neurological Institute Director
Taipei Veterans General Hospital Center for Image Guided Neurosurgery
Beitou, Taipei, Taiwan Director
Residency Training Program
Young K. Lee, PhD, MIPEM University of Pittsburgh
Medical Physicist Pittsburgh, Pennsylvania
Department of Medical Physics, Odette Cancer Center
Assistant Professor Lijun Ma, PhD
Department of Radiation Oncology, University of Toronto Professor in Residence
Sunnybrook Health Sciences Centre Department of Radiation Oncology
Toronto, Ontario, Canada University of California, San Francisco
San Francisco, California

xii
Contributors

Mitchell Machtay, MD Christian C. Okoye, MD


Professor and Chair Resident Physician
Department of Radiation Oncology Department of Radiation Oncology
Case Western Reserve University School of Medicine University Hospitals Seidman Cancer Center
University Hospitals Seidman Cancer Center Case Comprehensive Cancer Center
Cleveland, Ohio Cleveland, Ohio

David B. Mansur, MD David Hung-Chi Pan, MD


Associate Professor of Radiation Oncology and Pediatrics Professor
Vice Chairman for Proton Therapy National Yang-Ming University School of Medicine
Department of Radiation Oncology Director
Case Western Reserve University School of Medicine Gamma Knife Center
University Hospitals Seidman Cancer Center Taipei Medical University Shuang-Ho Hospital
Cleveland, Ohio Department of Neurosurgery
Taipei Veterans General Hospital
Eric Mazel, MA, MALS Taipei, Taiwan
Research Assistant
New York University Langone Medical Center Ravi B. Patel, MD, PhD
New York, New York Resident Physician
Department of Radiation Oncology
Heyoung L. McBride, MD, MS University Hospitals Seidman Cancer Center
Associate Professor Case Comprehensive Cancer Center
Barrow Neurological Institute Cleveland, Ohio
University of Arizona College of Medicine-Phoenix
Patrick Pezeshkian, MD
Phoenix, Arizona
Neurosurgeon
Kaiser Permanente
Edward A. Monaco III, MD, PhD
Redwood City, California
Assistant Professor of Neurological Surgery
Center for Image Guided Neurosurgery Denis Porcheron, PhD
Department of Neurological Surgery Service de Neurochirurgie Fonctionnelle & Stéréotaxique
University of Pittsburgh Medical Center Aix-Marseille Universite
Pittsburgh, Pennsylvania Hôpital Universitaire La Timone
Marseille, France
Xavier Muracciole, MD
Service de Radiothérapie Nader Pouratian, MD, PhD
Hôpital Universitaire La Timone Assistant Professor
Marseille, France Department of Neurosurgery
University of California, Los Angeles
Sten Myrehaug, MD Los Angeles, California
Department of Radiation Oncology
Durham Regional Cancer Centre Mark Quigg, MD, MSc, FANA
Oshawa, Ontario, Canada Professor
Department of Neurology
Gabor Nagy, MD, PhD University of Virginia
Department of Functional Neurosurgery Charlottesville, Virginia
National Institute of Clinical Neurosciences
Budapest, Hungary Ami B. Raval, MD
Division of Neurosurgery
Ajay Niranjan, MD, MBA Neuroscience Associates of New York
Associate Professor Staten Island University Hospital
Director of Radiosurgery Research Richmond University Medical Center
Department of Neurological Surgery Staten Island, New York
University of Pittsburgh
Pittsburgh, Pennsylvania

xiii
Contributors

Jean Régis, MD Andrew G. Shetter, MD


Service de Neurochirurgie Fonctionnelle & Stéréotaxique Division of Neurological Surgery
Aix-Marseille Universite Barrow Neurological Institute
Hôpital Universitaire La Timone St. Joseph’s Hospital and Medical Center
Marseille, France Phoenix, Arizona
Chairman
Pierre-Hugues Roche, MD Section of Functional and Stereotactic Neurosurgery
Department of Neurosurgery Co-Director
North hospital, APHM Gamma Knife Center
Aix-Marseille University Phoenix, Arizona
Marseille, France
Helen A. Shih, MD
Arjun Sahgal, MD Chief, CNS & Eye Services
Associate Professor of Radiation Oncology and Surgery Department of Radiation Oncology
Department of Radiation Oncology Massachusetts General Hospital
University of Toronto Associate Medical Director
Sunnybrook Health Sciences Center Francis H. Burr Proton Therapy Center
Toronto, Ontario, Canada Associate Professor of Radiation Oncology
Harvard Medical School
David Schlesinger, PhD Boston, Massachusetts
Associate Professor
Departments of Radiation Oncology and Neurological Gabriela Simonova, MD, PhD
Surgery Department of Stereotactic and Radiation Neurosurgery
University of Virginia Hospital Na Homolce
Charlottesville, Virginia Prague, Czech Republic

Jason Lee Schroeder, MD Andrew E. Sloan, MD, FAANS, FACS


Assistant Professor of Surgery Peter D. Cristal Chair of Neurosurgical Oncology
Department of Surgery Director, Brain Tumor and Neuro-Oncology Center
Division of Neurosurgery Professor & Vice-Chair for Research
University of Toledo Medical Center Department of Neurological Surgery
Toledo, Ohio University Hospitals Case Medical Center & Case
Comprehensive Cancer Center
Michael Schulder, MD, FAANS Cleveland, Ohio
Vice Chair and Residency Program Director
Department of Neurosurgery Penny K. Sneed, MD
Director Professor in Residence and Vice Chair
Brain Tumor Center Department of Radiation Oncology
Co-Director University of California, San Francisco
Center for Stereotactic Radiosurgery and Stereotactic San Francisco, California
Radiation Therapy
Hofstra North Shore LIJ School of Medicine John H. Suh, MD
Manhasset, New York Professor and Chairman
Department of Radiation Oncology
Jason P. Sheehan, MD, PhD, FACS Rose Ella Burkhardt Brain Tumor and Neuro-oncology
Harrison Distinguished Professor Center
Vice Chair of Neurological Surgery Cleveland Clinic
Professor of Radiation Oncology and Professor of Cleveland, Ohio
Neuroscience
Department of Neurological Surgery Gyorgy Szeifert, MD, PhD
Director Péterfy Traumatological Center
UVA Gamma Knife Center Department of Neurotraumatology
University of Virginia School of Medicine Semmelweis University of Budapest
Charlottesville, Virginia Budapest, Hungary

xiv
Contributors

Stephen B. Tatter, MD, PhD Brian Wang, PhD


Professor Associate Professor and Chief of Physics
Department of Neurosurgery Department of Radiation Oncology
Comprehensive Cancer Center University of Louisville
Brain Tumor Center of Excellence Louisville, Kentucky
Medical Center Boulevard
Winston-Salem, North Carolina Laura E. G. Warren, MD, EdM
Resident Physician
Jean-Marc Thomassin, MD Harvard Radiation Oncology Program
Professor Boston, Massachusetts
ENT Department
CHU Timone Daniel White, MD
Marseilles, France Neurosurgery
Sharp Chula Vista Medical Center
Christopher L. Tinkle, MD, PhD Chula Vista, California
Resident Physician
Department of Radiation Oncology Brian Winey, PhD, DABR
University of California, San Francisco Assistant Professor
San Francisco, California Physics Division
Department of Radiation Oncology
Vikram Udani, MD Massachusetts General Hospital
Neurosurgical Medical Clinic Harvard Medical School
San Diego, California Boston, Massachusetts

Michael A. Vogelbaum, MD, PhD, FAANS, FACS Huai-Che Yang, MD


Professor of Surgery (Neurosurgery) Department of Neurosurgery
Associate Director Neurological Institute
Neurosurgical Oncology Taipei Veterans General Hospital
Rose Ella Burkhardt Brain Tumor and Neuro Oncology National Yang-Ming University School of Medicine
Center Beitou, Taipei, Taiwan
Cleveland Clinic
Cleveland, Ohio

xv
1 The Origins and Birth of the Leksell
Part I Gamma Knife 1

History 2 The History of Linac and Proton Beam


Radiosurgery 11

3 The History of CyberKnife Radiosurgery 16

I
The Origins and Birth of the Leksell Gamma Knife

1 The Origins and Birth of the Leksell Gamma Knife


Dan Leksell and L. Dade Lunsford

Key Points
● The background and early development of radiosurgery.
● Building a solid science foundation is a time-consuming
effort.
● The regulatory environment has historically affected
disruptive innovations.

1.1 Background
Cerebral radiosurgery is a relatively new addition to the
armamentarium of neurological surgeons, having come of
age only in the late 1980s and early 1990s. However, the his-
torical background of cerebral radiosurgery goes back much
further to the late 1930s and may not be well known by
many young neurosurgeons today. The possibility of deliver-
ing a high dose of ionizing radiation through the intact skull
to a small and deep-seated cerebral target was a discovery
with roots belonging to the early history of neurosurgery in
Europe. In 1930 Herbert Olivecrona, one of the early Euro-
pean pioneers of neurosurgery, established the first neuro-
surgical department in Scandinavia. A few years later a young
and aspiring neurosurgeon, Lars Leksell, joined the team
training in neurosurgery at the Serafimerlasarettet in Stock-
holm, which at the time was the hospital of the Karolinska
Institute (▶ Fig. 1.1).
Leksell gained firsthand experience in the difficulties fac-
ing Olivecrona and his patients, especially those with brain
tumors and vascular malformations. This experience initi-
ated a lifelong quest to develop and apply less invasive and
less traumatic interventions for these difficult brain pathol-
ogies. As Olivecrona expanded his practice for newly diag- Fig. 1.1 Pioneering Swedish neurosurgeon Herbert Olivecrona with
nosed intracranial disorders, the mortality among his colleagues during surgery at the Serafimerlasarettet, the first
patients approached 60%. Death often occurred in the oper- Scandinavian training center for neurosurgery.
ating room (OR) itself. One of the first procedures witnessed
by Lars Leksell involved a young boy with a tumor of the
posterior fossa. The boy was prone on the operating table thrashing about the way he did.” In the course of his work
and the hospital gatekeeper was acting as anesthetist (a in neurosurgery, Olivecrona developed a stoic personality,
specialty that did not yet exist in Sweden in that era). The no doubt engendered by the challenges of those early years.
gatekeeper/anesthetist sat on the floor while delivering When Leksell joined Olivecrona for training in the 1930s, he
ether into the boy’s face. Olivecrona often told how in the was welcomed by a mixture of Churchillian “blood, sweat
early years of neurosurgery, patients, nurses, and even Oli- and tears” (▶ Fig. 1.2).
vecrona himself were half asleep from the vapors of ether in Dismayed by the level of mayhem in the OR, Leksell began to
the OR. As Leksell recalled the events of this particular ope- believe that there had to be better, less traumatic ways to help
ration, the boy started moving about on the table at a crit- patients requiring neurosurgery. These early training experien-
ical surgical moment in the surgery. Olivecrona began ces became the fulcrum for a life in constant search of mini-
yelling at him to “lie still, lie still!” Moments later a vessel mally invasive ways to treat neurosurgical disorders. He was in
in the boy’s headburst and his life could not be saved. Olive- fact a neurophysiologist at heart, and worked in the laboratory
crona stood up, looked out of the window, and said to his of Ragnar Granit, who in 1967 won the Nobel Prize in Physiol-
scrub nurse: “there is Pettersson in the garden; bring him in ogy or Medicine. Together with Granit, Leksell was the first to
and prepare him for surgery in an hour.” He then left the describe the gamma motor system, which regulates muscle
OR, proceeded to kick off his rubber boots, looked at Leksell, tone. His doctoral degree in this subject was granted by the Kar-
and said about the boy: “It was mostly his own fault, olinska Institute.

2
The Origins and Birth of the Leksell Gamma Knife

stereotactic methodology was based on a three-dimensional


1.2 Stereotactic Surgery as a Cartesian coordinate system, which allowed targeting of any
Method to Improve Patient point within its confines. The intention was to study cerebellar
physiology in monkeys, and it allowed Horsley to reliably deliv-
Outcomes er a probe to a deep target in the animal brain. The paper was
Almost 30 years before Leksell’s training at the Karolinska Insti- soon forgotten and interest in stereotactic techniques did not
tute, Sir Victor Horsley and Robert Clark had in 1908 published resurface until the 1940s, more than 10 years after Leksell’s first
a report describing an instrument for localizing structures in experiences with Olivecrona. Although it was common in the
the brain that they named a stereotactic device.1 This first use of 1930s and 1940s for trainees from the United States to travel to
Europe for additional training, it was rather rare for Europeans
to seek additional training in the United States. After the con-
clusion of the Second World War, Leksell arranged a fellowship
for training under the team of Ernst Spiegel, a neurologist, and
Henry Wycis, a neurosurgeon, who were working at Temple
University in Philadelphia. Spiegel and Wycis were the first gen-
erally recognized pioneers to apply stereotactic principles to
human patients,2 and over the years they developed several
models of their stereotactic instruments, including those
mounted to plaster casts on the head and others attached
directly to the vertex of the skull to anchor a probe holder. Lek-
sell returned to Stockholm and by 1949 had published his
article describing a prototype stereotactic guiding device using
a rectilinear coordinate system and an arc-centered principle to
deliver a probe to an intracranial target defined by radiographs
of the skull.3 During this same era, several other clinical investi-
gators designed their own stereotactic instruments. These con-
temporaries of Leksell included Jean Tallairach in Paris, Traugott
Riechert in Freiburg, and Irving Cooper in New York City. The
only instrument to use rectilinear Cartesian coordinates was
the Leksell instrument, which was used on a patient for the first
time in 1949. The design of the instrument, based on the arc-
centered principle, was intuitive and made the instrument easy
to use (▶ Fig. 1.3).
Two years later, in 1951, Leksell conceived the idea of replac-
ing the probe that was delivered surgically with his stereotactic
guiding device with multiple, narrow, cross-fired beams of ion-
izing radiation capable of traversing the intact skull. 4 In his first
report of this he outlined how this might be accomplished. By
attaching an X-ray tube to the arc of the guiding device, the
tube could be rotated in the anteroposterior (AP) direction and
also moved laterally along the arc to simulate an indefinite
number of X-ray beams, all aimed toward a common point of
Fig. 1.2 Olivecrona’s operating room in Stockholm during the dawn of
beam intersection. The point of intersection was selected as the
Scandinavian neurosurgery.
target for surgery (▶ Fig. 1.4). Leksell anticipated that the best

Fig. 1.3 The basic principle of the arc-centered


Leksell stereotactic instrument. The intersection
of three planes (X, Y, and Z) defined the target
point for treatment, with this point being located
at the center of an arc. (a) By knowing the radius
of the arc (19 cm), the surgeon could deliver a
probe to the target point after choosing an
anatomically safe trajectory (left). (b) The first
Leksell frame (1949) is shown on the right.

3
The Origins and Birth of the Leksell Gamma Knife

Fig. 1.4 The X-ray tube of an orthovoltage dental X-ray device was
mounted on the semicircular arc of the prototype Leksell frame. The
delivery of multiple converging beams to a common point of beam
intersection created the field of radiosurgery.

type of ionizing radiation for use with his device would be X- Fig. 1.5 The cyclotron in Uppsala that generated cross-fired proton
rays or gamma rays. He also stated that the available orthovolt- beams guided by a version of the Leksell stereotactic methodology.
age energy of 200 kV was too low to sufficiently penetrate tis-
sue and protect skin. With this landmark paper, the term
“stereotactic radiosurgery” was coined. Of historical interest is
duplicated but made much sturdier so as to mate it with the
the journal editor’s initial rejection of this seminal paper with
cyclotron. The set-up was such that the patient undergoing
the notation that “Everyone knows there is no such thing as
stereotactic radiosurgery was positioned on a couch that piv-
radiosurgery.”
oted around the exit point of the fixed beam of the cyclotron,
thus again simulating an indefinite number of beams intersect-
1.3 Initial Clinical Experience ing at the center of the stereotactic arc, which was the target to
be irradiated.11,16
In 1953, Leksell put into practice the hypothetical suggestions A long series of animal experiments were then undertaken to
made in his 1951 report when two patients with trigeminal better understand the effects of single-dose radiation on the
neuralgia were treated with his technique of stereotactic radio- brain, to determine the dose necessary to obtain a lesion in nor-
surgery.5 The original stereotactic instrument lent itself well to mal brain tissue, and to refine the methodology of stereotactic
these initial applications. The target was the gasserian ganglion, radiosurgery. The initial confirmatory animal work was per-
a structure whose treatment had previously required a subtem- formed on goats, which were allowed to roam freely on Lars-
poral craniotomy in an era long before the introduction of the son’s farm until they died of natural causes. Their brains were
operating microscope and safe neuroanesthesia. The first pa- then harvested and studied to assess the lesion created by the
tient treated with Leksell’s technique had a gradual reduction radiation they had been given.
of pain until its complete disappearance at 5 months postoper- Following extended study of the effects of stereotactic radia-
atively. The second patient became pain-free after a few days, tion on the brains of goats and other animals, a third patient
and both patients remained pain-free at 17 years after their was treated at Uppsala in 1961 with the proton radiosurgical
surgery. The orthovoltage X-ray tube that was attached to the technique (▶ Fig. 1.5). Nevertheless, the cyclotron used for the
arc of Leksell’s instrument was heavy and its energy was low, at treatment, although a good source of cross-fired proton radia-
280 kV. The entire setup was awkward and took too long to tion (the proton Bragg peak effect was not used), was not prac-
deliver the intended dose of radiation. However, the prelimi- tical from a clinical standpoint. The method was cumbersome
nary results warranted continued efforts in stereotactic radio- and relied on very expensive equipment that required signifi-
surgery, and the search began for a better source of radiation. cant technical support and support in physics.
In the mid-1950s Leksell made contact with the radiobiolo- After Leksell succeeded Olivecrona as Professor of Neurosur-
gist Börje Larsson, who was on the Faculty of Medicine at gery at the Karolinska Institute and returned to Stockholm, his
Uppsala University and eventually became dean of the Gustav commute to Uppsala became easier. Patients undergoing ster-
Werner synchrocyclotron laboratory in Uppsala, an hour north eotactic radiosurgery had their stereotactic frames applied in
of Stockholm. Larsson was evaluating the effect of proton radia- Stockholm and were then driven in the back seat of Leksell’s
tion on cell biology.6–15 Leksell at the time was chairman of Daimler to Uppsala to undergo the radiosurgical procedure
the Department of Neurosurgery at the University of Lund, a itself. The search for a radiosurgical technology that could be
city in southern Sweden 700 km south of Uppsala. Despite the used in the daily routine of a hospital therefore continued. A
distance, he soon established a very close collaboration with linear accelerator in the Department of Radiation Oncology at
Larsson. This led to Leksell’s stereotactic instrument being Uppsala was adapted for stereotactic irradiation and a small

4
The Origins and Birth of the Leksell Gamma Knife

Fig. 1.6 Lars Leksell attempted to use an early-generation linear


accelerator for stereotactic radiosurgery, but was worried about the
wobble of the beam caused by the excessive weight of the gantry.

Fig. 1.7 The design of the first Gamma Knife, based on the
series of patients with arteriovenous malformations (AVMs) specifications established in 1963. This prototype instrument used 179
were treated with this methodology (▶ Fig. 1.6). The results sources of cobalt 60, which emitted gamma rays that converged on a
target at the focal point of treatment.
were disappointing. The wobble of the gantry caused by the
very heavy head of the photon delivery device provided unac-
ceptable uncertainty in accurate dose delivery.
Leksell did not like the complexity of the linear accelerator
and its dependence on physics and engineering support to
remain operational. The search began for a device that was
extremely precise, reliable, and capable of operation by the sur-
geon without significant need for additional technical person-
nel. The device also had to be designed so that it would leave as
little room for surgeon error as possible. Knowing that sur-
geons, like everyone else, can make mistakes, Leksell often
quoted Norman Brown’s aphorism that “Fools with tools are
still fools.”
In 1963, the technical specifications for what was to become
the prototype Gamma Knife were finalized.17 Despite Leksell’s
personal interests in functional brain surgery and his plan to
use it to create radiodestructive lesions in the brain for unre-
sponsive behavioral,18 nociceptic,5,16 or movement disorders,
the first patient treated had a craniopharyngioma. The patient
Fig. 1.8 Erik-Olof Backlund explains the thermoplast Gamma Knife
was treated at the factory in Studsvik, Sweden, where the first
head-fixation system to a pioneering early patient. The thermoplast
Leksell Gamma Knife was built and loaded with 179 sources of system was used for fixation of the patient’s head because the
cobalt 60.17 In early 1968, this prototype machine was installed stereotactic frame on which the Gamma Knife was mounted could not
at the Sophiahemmet in Stockholm, one of very few private fit within the helmet used with the first Gamma Knife. The thermoplast
hospitals in Sweden because it was built under the auspices of cast of the first treated patient was given by Professor Backlund to L.
then Swedish Queen Sophia (▶ Fig. 1.7). Meticulous, cautious, Dade Lunsford when Lunsford was inaugurated as the first Lars Leksell
Professor of Neurosurgery at the University of Pittsburgh in Pennsyl-
and exceedingly slow clinical exploration ensued. This was a
vania.
hallmark of Leksell’s philosophy: Don’t treat large numbers of
patients until you have convinced yourself and your colleagues
that what you do makes a difference without undue complica-
tions. In fact, between 1968 and 1986 an average of less than secondary collimators were designed to focus the beams and
three patients per month were treated by Gamma Knife surgery contained a small slit rather than a circular opening. It was
in Stockholm. designed to create a discoid rather than oblate spheroidal radio-
The term Gamma Knife derived from its early description as surgical field, as if to “section” a target. The helmet containing
strålkniven, or “radiation knife” in Swedish; the concept was to the secondary collimators was too small to accommodate the
use the photon beam intersection as a surgical sectioning tool frame itself. As a result, after the frame had been used in deter-
within the closed skull. Since the 179 cobalt 60 sources pro- mining the target for surgery, the frame was removed from the
duced gamma rays, the new technology eventually came to be plaster or Surgiplast cast that had been applied to the patient’s
known as the “Gamma Knife.” The prototype Gamma Knife had head (▶ Fig. 1.8). Bearings were seared into the plaster at the Z
two collimators, of 3 × 5 and 3 × 7 mm, respectively. These and Y coordinates of the three-dimensional field of treatment

5
The Origins and Birth of the Leksell Gamma Knife

with the Gamma Knife, and the patient was moved to the left or emerging indications treated with the Gamma Knife. The cross-
right manually, using trunnions to reach the X coordinate of the fired proton experiments in Uppsala had revealed that 140 Gy
treatment site. were needed to produce a radiodestructive lesion in the brain.
Most of the early patients treated with the Gamma Knife had In the early years, the photon doses that the Gamma Knife
functional disorders,19 including intractable pain, trigeminal delivered were therefore were very high, often around 200 Gy.
neuralgia, and refractory obsessive–compulsive/anxiety disor- Today such doses are never used, and in fact, most patients do
ders for which neither medication or psychiatric treatment pro- not receive even 10% of that dose. Little was it known at the
vided any benefit. The first reported patient to be treated for an time that the appropriate radiobiological response for many
AVM underwent radiosurgery in 1971.20 As usual, Leksell indications of stereotactic radiosurgery could be achieved with
authorized a single patient to be treated, with an ensuing 2- nonnecrotizing doses of radiation. Today some sources profess
year period of observation. The necessary obliterative dose of that nonnecrotizing doses of radiation delivered in radiosurgery
gamma radiation was unknown and the effect unpredictable. can modulate brain function, much like deep brain stimulation.
Fortunately, follow-up angiography showed obliteration of the In contrast to Leksell’s early experience in the ORs of Olive-
patient’s AVM, which stimulated Ladislau Steiner to proceed crona, the new, noninvasive Gamma Knife procedure was
with the Gamma Knife treatment of additional cases. Imaging undramatic. There was no blood, no sweat, and absolutely no
for target localization consisted primarily of pneumoencepha- tears. When the patient had been positioned in the focus of the
lography, cisternography, and cerebral angiography. Because collimator helmet of the Gamma Knife, the treatment team
this occurred in an era before computers were available, both called the kitchen and ordered sandwiches and Swedish beer
target localization through X-ray films and dose planning were for lunch. At the end of the procedure, the premises did not
done manually. Dose determination was done by means of a look like any other OR. When the frame was removed from the
nomogram handcrafted by the physicist21 (▶ Fig. 1.9). patient’s head, everyone was happy. The patients were happy
The wavy lines on the nomogram would probably not inspire because they were going home on the day of brain surgery, and
confidence today, and it is possible that patients received either the doctors were also happy, perhaps because they had had too
a larger or a smaller dose of radiation than intended in those much beer for lunch (▶ Fig. 1.10).
early years. This did not matter very much because at the time
nobody really knew the effective and safe dose for any of the

Fig. 1.10 (a) The prototype noninvasive Leksell Gamma knife OR, with
Lars Leksell at the console and surrounded by colleagues, including
Fig. 1.9 (a) Manual target localization and (b) dose determinations Erik-Olof Backlund (far left) and Bengt Jernberg, his engineer (far right).
were performed using special nomograms developed by Lars Leksell (b) At the end of the procedure, celebratory starköl (Swedish beer) was
and medical physicists. routine.

6
The Origins and Birth of the Leksell Gamma Knife

While the original prototype remained at the private Sophia- In 1979, Lars Leksell was persuaded by his friend Robert
hemmet in Stockholm, Leksell arranged for the construction of Rand, Professor of Neurosurgery at the University of California–
a second Gamma Knife that was installed in the basement of Los Angeles (UCLA), to donate the prototype first Gamma Knife
the Radiumhemmet on the grounds of the Karolinska Hospital to him. As the story goes, the fully loaded (but with low-
in Stockholm. This unit entered service in 1975 and was rede- strength cobalt 60, now in its third half-life) unit was shipped
signed to produce a more oblate spheroid field that was suit- from Stockholm to Long Beach, California, in a special container
able for morphological and pathological targets in the brain. marked “radioactive.” During the long trek from Stockholm
Disciples of Leksell who were given special projects began addi- around the world to California, the freighter carrying the unit
tional work: Ladislau Steiner and Christer Lindquist were stopped periodically to unload cargo at various ports. The
assigned to treat AVMs20; George Noren to treat acoustic neuro- inspecting harbormaster invariably told the captain to move on
mas22; Tiit Rähn to treat pituitary tumors23; and Erik-Olof Back- after finding the “radiation-containing” cargo in the freighter’s
lund to treat craniopharyngiomas.17,24 hold. The story reminded Leksell of the saga of the Flying
Seven years after the prototype Gamma Knife was installed in Dutchman, who was allowed to enter port every 7 years to look
Stockholm, the first computed tomography (CT) scanner in the for true love. Eventually, the freighter carrying the prototype
country was installed at the Karolinska Hospital in 1974.25 Sud- Gamma Knife docked in Long Beach, and shortly thereafter in
denly, it became possible to directly visualize cerebral pathologies 1980, the knife miraculously appeared in a vault at UCLA. It
and the surrounding brain without having to rely on indirect remains unclear how many patients Rand treated using this
signs of pathology such as the displacement of a ventricle or a prototype Gamma Knife, although an early report showed its
vessel. The Leksell frame was redesigned to make it compatible potential in a rabbit model of ocular melanoma. 27 After some
with CT imaging, and was further modified in the 1980s to make years, the unit was unloaded and shipped back to Stockholm to
it compatible with magnetic resonance imaging (MRI).26 potentially rest at the Stockholm Technological Museum.

1.4 Spreading the Gospel 1.5 The Next Gamma Knife Units
Leksell much preferred working in Stockholm to traveling to With such abundant skepticism surrounding its efficacy, radio-
meetings. This meant that his son Dan (a coauthor of this chap- surgery did not catch on quickly as a treatment modality. Lars
ter), long before finishing medical school, was asked to travel in Leksell was not particularly interested in the economic poten-
his stead to give talks all over the world. Dan Leksell spoke tial of this remarkable discovery. Three of Leksell’s trainees
about the early experiences with the radically new, noninvasive eventually persuaded him to authorize the construction of two
form of neurosurgery made possible with the Gamma Knife in additional Gamma Knife units. They were constructed in Swit-
Stockholm. At the time, the possibility of curing brain disease zerland by an authorized agent, Scanditronix. Based on the per-
without a craniotomy was considered incredible. Like most dis- sistent intercessions of Roberto Chescotta and Hernan Bunge,
ruptive innovations, descriptions of the method and roles of also prior students of Leksell, a third Gamma Knife was built in
radiosurgery were for many years met with great skepticism if 1984 and shipped to Buenos Aires for installation in the Clinica
not outright hostility. Dan Leksell usually started his talks by del Sol. Unit 4 was delivered to Sheffield, England, where it
showing an engraving done by the Renaissance German artist entered service under the direction of David Forster
Matthias Greuter (▶ Fig. 1.11). He then began by telling his audi- (▶ Fig. 1.12). In a remarkable prediction of the direction of the
ence that he was not going to talk about anything new, and that
what was done in Stockholm had already been done during the
Renaissance. At this, everybody smiled and laughed. But by the
time he was finished with his lecture, nobody was laughing!

Fig. 1.11 This Renaissance-era lithograph from the annals of Mathias


Greuter was often used to introduce talks about the Gamma Knife and
its roles to audiences around the world. Fig. 1.12 David Forster and Gamma Unit 4 in Sheffield, England.

7
The Origins and Birth of the Leksell Gamma Knife

Gamma Knife technology, the initial design of these third and


fourth units required moving the patient’s positioning on the
treatment table to accurately focus the 179 beams of radiation
delivered by the instrument (rather than the surgeon manually
moving the patient’s head and treatment target into the focus
of the selected beams). Unfortunately, this radical idea foun-
dered because of insufficiently reliable table-positioning tech-
nologies in that era. Both the Buenos Aires and the Sheffield
units were subsequently reconfigured to again have the sur-
geon manually position the patient’s head at the focus of these
instruments’ radiation beams.
With the passage of time, the tireless efforts of a few pioneers
eventually brought about grudging acknowledgment of the
potential of radiosurgery. Leksell’s disciples diligently evaluated
the indications for radiosurgery to which each of these pioneers
was assigned. During the 1980s other radiosurgical pioneers Fig. 1.13 Gamma Unit 5 (the first installation to have 201 sources of
emerged: Juan Barcia Salorio in Madrid,28 Volker Sturm in Hei- cobalt 60 and a new, 18-mm secondary collimator helmet) was
installed at Presbyterian University Hospital at the University of
delberg, Federico Colombo in Vicenza, Italy,29 and Osvaldo Betti
Pittsburgh Medical Center (UPMC), and began clinical operation on
in Buenos Aires and Paris, all of whom adapted linear accelera- August 14, 1987. Since then, more than 12,600 patients have
tors to emulate the principle of the Gamma Knife. Using fractio- undergone Gamma Knife radiosurgery at UPMC, with different models
nated techniques with accelerated helium ions, John H. of the Gamma Knife.
Lawrence30 and then Jacob Fabrikant, working at the Berkeley
cyclotron, began to create pituitary lesions and to obliterate
AVMs. Raymond Kjellberg, working in the Cambridge cyclotron
the rate at which the Pittsburgh group published their data was
unit in Cambridge, Massachusetts, that had been donated to
very high, and has continued to be very high since they began
Harvard by the Navy in thanks for its collaboration in the Man-
their reporting. In relatively short order, several more installa-
hattan Project during the Second World War, used the Bragg
tions of the Gamma Knife in the United States followed its debut
peak principle of proton beam therapy to treat a large number
at Pittsburgh, and in 1990 the first Gamma Knife Center in Asia
of patients with AVMs or pituitary tumors. 31
became operational at Tokyo University under the supervision
Despite all the efforts of these early enthusiasts, it would take
of Kintomo Takakura. Other countries would subsequently fol-
almost 20 years for the development of sufficient traction to
low suit.
advance the field of radiosurgery. After studying with Lars Lek-
sell and Erik-Olof Backlund at the Karolinska institute in 1979
and for a year in 1980 and 1981, L. Dade Lunsford returned to
Pittsburgh, Pennsylvania, with a two-part plan: to establish a
1.6 A Final Perspective
dedicated OR for image-guided neurosurgery (installing the In December of 1985, a few weeks before Leksell died and when
first dedicated CT scanner)32 and to develop CT-compatible there were still only four Gamma Knives in the world, his son
technologies for radiosurgery.33 Beginning in 1982, a 5-year Dan asked him, “How many more machines do you think the
effort pushed the concept of stereotactic radiosurgery with the world might absorb or need?” His answer was “Maybe two or
Gamma Knife in the United States through many regulatory three more.” As of the time of this writing, more than 300 Gam-
hurdles, including those of the U.S. Food and Drug Administra- ma Knife centers have appeared in the world and the number
tion (FDA) and the U.S. Nuclear Regulatory Commission (NRC).27 continues to grow annually.
The greatest issue in implementing use of the knife was devis- During the first two decades after its introduction, the Gam-
ing a method for the on-site loading of the new 201-source ma Knife existed at only four treatment centers in the world,
Gamma Knife with its four focusing collimator helmets (with whereas the ensuing two decades have seen the installation of
apertures of 4, 8, 14, and 18 mm, respectively) because the NRC more than 300 of these instruments. New generations of the
would not allow moving a loaded unit across any distance. Gamma Knife have been introduced at regular intervals, each
The Pittsburgh site initiated a new explosion of interest in bringing a higher level of sophistication to stereotactic radio-
radiosurgery as a method and in the Gamma Knife as a radio- surgery. The latest generation (Perfexion; Elekta AB, Stockholm,
surgical tool (▶ Fig. 1.13). After installation of the Gamma Knife Sweden) incorporates several significant improvements, not
at Pittsburgh in 1987, the Pittsburgh team, under the leadership least of which are shorter treatment times, improved patient
of L. Dade Lunsford, began publishing not only their early clini- treatment efficiency, and vastly more sophisticated treatment
cal results with the new instrument, but also cost-effectiveness planning.
data and other information relating to its use.34 It did not take One very important contributing factor to all of this must be
long for the information from Pittsburgh to spread and kindle recognized. The rate of development of neuroimaging since Sir
international interest. Many have often wondered why the 20 Geoffrey Hounsfield was awarded the Nobel Prize for inventing
years of painstaking work done in Stockholm never by itself led CT in 197635 has been astonishing and all-important for the
to the greater acceptance of stereotactic radiosurgery. The development of radiosurgery. The subsequently developed of
answer is that independent corroboration is a key factor for the MRI has become instrumental to neurosurgeons’ daily work.26
credibility of any new technology. It should also be added that Gamma Knife radiosurgery represents the treatment-delivery

8
The Origins and Birth of the Leksell Gamma Knife

Fig. 1.14 The fifth-generation Leksell Gamma Knife represented the therapeutic correlate to the (a,b) early generation of computed tomography
scanners. (c,d) The most recent Gamma Knife device (the Perfexion instrument) corresponds to new imaging methods with 3-Tesla magnetic
resonance imaging.

technology that was needed to mirror the revolution in diag- medical approaches and technologies are regarded with
nostic imaging of the past half century (▶ Fig. 1.14). In the cur- appropriate skepticism and must prove their value before
rent regulatory environment it is unlikely that this history being widely adopted.
could be repeated. The way in which Lars Leksell, his collabora-
tors, and his disciples could work in the 1950s to 1970s would
not be possible today. Had the current regulatory environment References
existed in that era, it is unlikely that radiosurgery would have [1] Horsley V, Clarke RH. The structure and functions of the cerebellum exam-
emerged. ined by a new method. Brain 1908; 31: 45–124
A great source of pride for radiosurgery providers [2] Spiegel EA, Wycis HT, Freed H, et al. Stereoencephalotomy. Proc Soc Exp Biol
throughout the world is the sheer number of patients who Med 1948; 69: 175–177
[3] Leksell L. A stereotaxic apparatus for intracerebral surgery. Acta Chir Scand
have benefited from the hard work done by so many in this
1949; 99: 229–233
field. Statistics reported by about 80% of all Gamma Knife [4] Leksell L. The stereotaxic method and radiosurgery of the brain. Acta Chir
centers indicate that as of 2014, more than a million Scand 1951; 102: 316–319
patients had undergone Gamma Knife surgery, and approxi- [5] Leksell L, Backlund EO. Stereotaxic gamma capsulotomy. In: Hitchcock ER,
Ballantine HT, Meyerson BD, eds. Modern Concepts in Psychiatric Surgery.
mately 60,000 new patients undergo such surgery every
Amsterdam: Elsevier/North-Holland Biomedical Press;1979:213–216
year. Between 1968 and 1988 approximately 80 papers on [6] Andersson B, Larsson B, Leksell L, et al. Histopathology of late local radiole-
Gamma Knife surgery were published; over the next 15 sions in the goat brain. Acta Radiol Ther Phys Biol 1970; 9: 385–394
years, more than 2,000 such papers were published in the [7] Dahlin H, Larsson B, Leksell L, Rosander K, Sarby B, Steiner L. Influence of ab-
peer-reviewed literature. This development would probably sorbed dose and field size on the geometry of the radiation-surgical brain le-
sion. Acta Radiol Ther Phys Biol 1975; 14: 139–144
have both perplexed and amused Lars Leksell. The introduc-
[8] Dahlin H, Sarby B. Destruction of small intracranial tumours with 60Co gam-
tion of new methods and disruptive technologies in medi- ma radiation. Physical and technical considerations. Acta Radiol Ther Phys Bi-
cine is not easy. It requires perseverance and patience. New ol 1975; 14: 209–227

9
The Origins and Birth of the Leksell Gamma Knife

[9] Larsson B. Blood vessel changes following local irradiation of the brain with [23] Thorén M, Rähn T, Hall K, Backlund EO. Treatment of pituitary dependent
high-energy protons. Acta Soc Med Ups 1960; 65: 51–71 Cushing’s syndrome with closed stereotactic radiosurgery by means of 60Co
[10] Larsson B, Leksell L, Rexed B. The use of high-energy protons for cerebral sur- gamma radiation. Acta Endocrinol (Copenh) 1978; 88: 7–17
gery in man. Acta Chir Scand 1963; 125: 1–7 [24] Backlund EO, Rähn T, Sarby B, De Schryver A, Wennerstrand J. Closed stereo-
[11] Larsson B, Leksell L, Rexed B, Sourander P, Mair W, Andersson B. The high-en- taxic hypophysectomy by means of 60-Co gamma radiation. Acta Radiol Ther
ergy proton beam as a neurosurgical tool. Nature 1958; 182: 1222–1223 Phys Biol 1972; 11: 545–555
[12] Mair W, Rexed B, Sourander P. Histology of the surgical radiolesion in the hu- [25] Leksell L, Jernberg B. Stereotaxis and tomography. A technical note. Acta Neu-
man brain as produced by high-energy protons. Radiat Res Suppl 1967; 7 rochir (Wien) 1980; 52: 1–7
(Suppl): 384–389 [26] Leksell L, Leksell D, Schwebel J. Stereotaxis and nuclear magnetic resonance. J
[13] Rexed B, Main W, Sourander P, et al. Effect of high energy protons on the Neurol Neurosurg Psychiatry 1985; 48: 14–18
brain of the rabbit. Acta Radiol 1960; 53: 289–299 [27] Rand RW, Khonsary A, Brown WJ, Winter J, Snow HD. Leksell stereotac-
[14] Sarby B. Cerebral radiation surgery with narrow gamma beams; physical ex- tic radiosurgery in the treatment of eye melanoma. Neurol Res 1987; 9:
periments. Acta Radiol Ther Phys Biol 1974; 13: 425–445 142–146
[15] Wennerstrand J, Ungerstedt U. Cerebral radiosurgery. II. An anatomical study [28] Barcia-Salorio JL, Broseta J, Hernandez G, et al. Radiosurgical treatment in
of gamma radiolesions. Acta Chir Scand 1970; 136: 133–137 huge acoustic neuromas. In: Szikla G, ed. Stereotactic Cerebral Irradiation. IN-
[16] Leksell L. Cerebral radiosurgery. I. Gammathalanotomy in two cases of in- SERM Symposium, No. 12. Amsterdam: Elsevier/North-Holland Biomedical
tractable pain. Acta Chir Scand 1968; 134: 585–595 Press; 1979: 245–249
[17] Backlund E-O. The history and development of radiosurgery. In: Lunsford LD, [29] Colombo F, Benedetti A, Pozza F, et al. External stereotactic irradiation by lin-
ed. Stereotactic Radiosurgery Update. Philadelphia, PA: Elsevier; 1992: 3–9 ear accelerator. Neurosurgery 1985; 16: 154–160
[18] Mindus P, Bergström K, Levander SE, Norén G, Hindmarsh T, Thuomas KA. [30] Lawrence JH, Tobias CA, Linfoot JA, Born JL, Chong CY. Heavy-particle therapy
Magnetic resonance images related to clinical outcome after psychosurgical in acromegaly and Cushing disease. JAMA 1976; 235: 2307–2310
intervention in severe anxiety disorder. J Neurol Neurosurg Psychiatry 1987; [31] Kjellberg RN, Koehler AM, Preston WM, Sweet WH. Stereotaxic instrument
50: 1288–1293 for use with the Bragg peak of a proton beam. Confin Neurol 1962; 22: 183–
[19] Leksell L. Sterotaxic radiosurgery in trigeminal neuralgia. Acta Chir Scand 189
1971; 137: 311–314 [32] Lunsford LD, Rosenbaum AE, Perry J. Stereotactic surgery using the “thera-
[20] Steiner L, Leksell L, Greitz T, Forster DM, Backlund EO. Stereotaxic radiosur- peutic” CT scanner. Surg Neurol 1982; 18: 116–122
gery for cerebral arteriovenous malformations. Report of a case. Acta Chir [33] Lunsford LD, Leksell L, Jernberg B. Probe holder for stereotactic surgery in the
Scand 1972; 138: 459–464 CT scanner. A technical note. Acta Neurochir (Wien) 1983; 69: 297–304
[21] Arndt J, Backlund EO, Sahlin H, et al. A computer controlled dose planning [34] Lunsford LD, Flickinger J, Lindner G, Maitz A. Stereotactic radiosurgery of the
system for irradiation of small volumes of the brain with a multi-cobalt unit. brain using the first United States 201 cobalt-60 source gamma knife. Neuro-
Paper presented at: the Third Congress of the European Society of Radiology; surgery 1989; 24: 151–159
June 22–27, 1975; Edinburgh, Scotland [35] Hounsfield GN. Computerized transverse axial scanning (tomography). 1. De-
[22] Leksell L. A note on the treatment of acoustic tumours. Acta Chir Scand 1971; scription of system. Br J Radiol 1973; 46: 1016–1022
137: 763–765

10
The History of Linac and Proton Beam Radiosurgery

2 The History of Linac and Proton Beam Radiosurgery


Laura E. G. Warren, Brian Winey, Jay S. Loeffler, and Helen A. Shih

not superior to those with X-ray therapy. Stone’s work was dis-
Key Points continued in 1942, when the cyclotron at Berkeley was reallo-
cated to other purposes.
● Particle-beam experiments in physics laboratories and the In 1946, Robert Wilson, who had worked on the Manhattan
development of stereotactic neurosurgical techniques in the Project, first proposed that accelerated protons and heavier ions
early 20th century provided the foundations for stereotactic could be utilized for treating human patients.7 His work intro-
radiosurgery. duced the therapeutic concept of the Bragg peak, a distinct and
● Lars Leksell, a Swedish neurosurgeon, is widely credited with localized region of high-dose radiation that was characteristic
the introduction of charged-particle radiosurgery to the in- of proton beam irradiation. Soon thereafter, Wilson and Edward
ternational medical community with his famous 1951 paper Creutz, who had joined the Department of Physics at the Carne-
on stereotactic radiosurgery of the brain. gie Institute of Technology in Pittsburgh, Pennsylvania, after
● Linear accelerator–, as compared with cyclotron-based, ra- having worked on the Manhattan Project, described the meth-
diosurgery developed in the 1980s and 1990s. odology by which they were able to produce monoenergetic
● Advances in imaging for diagnosis and treatment planning, protons from a cyclotron, further raising the potential for the
patient-positioning technologies, and the construction of clinical application of proton beam irradiation. 8 On the other
dedicated clinical facilities have resulted in increasing utiliza- side of the United States, in Berkeley, California, Ernest Orlando
tion of stereotactic radiosurgery and the expansion of its Lawrence founded the Radiation Laboratory at the University of
clinical applications over the past two decades. California, a physics laboratory centered around a newly devel-
oped cyclotron that had been built with his student Niels Edlef-
sen.9 Animal studies were soon underway, with the ultimate
goal of oncologic applications. Studies focusing on pituitary
2.1 The Early Years irradiation were undertaken on the basis of prior work demon-
strating that hypophysectomy might be of benefit in the treat-
The Swedish physician Lars Leksell introduced the world to the ment of various diseases, including metastatic breast cancer. 10,
stereotactic utilization of therapeutic X-radiation in 1951.1 His 11 John Lawrence, Ernest Lawrence’s brother, left Yale to join his

published paper entitled “The Stereotaxic Method and Radio- brother at Berkeley and was integral to these initial therapeutic
surgery of the Brain” is often cited as the introduction to the efforts.12 Cornelius A. Tobias, another prominent member of the
technical and practical aspects of clinical stereotactic radiosur- Radiation Laboratory at Berkeley, which later became the Don-
gery. In subsequent years, Leksell continued to have a fruitful ner Laboratory, described the value of high-energy particles in
collaborative relationship with Borje Larsson, a physicist and 1958 when he stated that “[These] particles, when focused,
radiobiologist in the cyclotron unit at Uppsala University in travel in a straight beam with little divergence and may be
Sweden. Leksell advocated the use of proton radiosurgery pre- directed to any portion of the body.” Moreover, noted Tobias,
dominantly for benign brain tumors. This was motivated by his “As the particles penetrate tissue, their scattering is very small
concern that the biology of malignant neoplasms was such that compared with electrons, and for practical purposes negligibly
radiosurgery might prove ineffective for treating them, and that small amounts of radiation fall outside the main beam.”13
the combination of the great incidence of benign tumors and After promising animal studies showing tumor regression or
limited capacity of his radiosurgery unit would make the the arrest of tumor growth, Lawrence treated 26 metastatic
expansion of radiosurgery to the treatment of malignant dis- breast cancer patients with pituitary proton irradiation using a
ease logistically challenging.2 Leksell understood that the inva- 340-MeV proton beam from a 184-inch synchrocyclotron at the
sive nature of the contemporary neurosurgery of his time Donner Laboratory in Berkeley, California. Lawrence and his
resulted in high perioperative morbidity and mortality, and was colleagues described decreased pituitary function and gross
therefore enthusiastic about minimally invasive methods for microscopic damage to the pituitary gland.13,14 A good clinical
treating intracranial lesions. response in a few of the patients was also seen. In 1957, proton
Although Leksell can certainly be credited for his clinical beam experiments in the Donner Laboratory were discontinued
applications of radiosurgery, the work of other international in favor of experiments with 910-MeV helium ions and then
centers in the decades before Leksell’s work helped to provide with heavier ions.15
its foundational underpinnings. Ernest Rutherford, the 1908 Internationally, Lars Leksell and his colleagues at the Univer-
Nobel laureate in chemistry, demonstrated the existence of and sity of Uppsala were equally interested in destroying small,
named the proton in 1919.3,4 In 1932, James Chadwick discov- selected regions within the central nervous system with high-
ered neutrons under Rutherford’s guidance.5 Chadwick’s dis- energy heavy particles.16 They utilized a 185-MeV proton beam
covery sparked considerable interest in the clinical utilization from a 230-cm synchrocyclotron. Animal studies showed that it
of neutron beams. Robert Stone experimented with fast-neu- was possible to produce sharply delineated lesions in any re-
tron radiotherapy in Berkeley, California,6 treating approxi- gion of choice in the central nervous system.17,18 In 1960, Lek-
mately 250 patients in this way between 1937 and 1942. sell and his colleagues performed their first stereotactic proton
Unfortunately, his experiments demonstrated the potential for beam “operation.”17 However, following these experiments,
severe late tissue injury with such therapy, and the results were Leksell stated that “Although the synchrocyclotron is a valuable

11
The History of Linac and Proton Beam Radiosurgery

Fig. 2.2 Dr. Lee Davenport, a research fellow and physics professor,
Fig. 2.1 Harvard University Physics Professors Jabez C. Street (left, and Dr. Leo Lavatelli, then a doctoral-degree candidate, in the second
standing) and Kenneth Bainbridge (right, standing), with then Harvard University Cyclotron control room in about 1950. (Used with
graduate student Dr. Roger W. Hickman (kneeling), examining the first permission from Professor Richard Wilson, from Wilson R. A Brief
Harvard Cyclotron in about 1940. Construction of the cyclotron had History of the Harvard University Cyclotrons. Cambridge, MA: Harvard
been completed in 1938. (Used with permission from Professor University Press; 2004. Available at: http://users.physics.harvard.edu/
Richard Wilson, from Wilson R. A Brief History of the Harvard ~wilson/cyclotron/history.html. Updated June 16, 2003. Accessed May
University Cyclotrons. Cambridge, MA: Harvard University Press; 2004. 14, 2014.)
Available at: http://users.physics.harvard.edu/~wilson/cyclotron/his-
tory.html. Updated June 16, 2003. Accessed May 14, 2014.)

Unfortunately, despite significant initial tumor shrinkage, the


patient died several years later.21 Thereafter, the work at the
research tool, it has proved too complicated for general neuro- Harvard cyclotron was predominantly focused on pituitary
surgical application.”19 This was particularly relevant in Swe- adenomas and AVMs.22,23,24
den, where patients had to be transported from Stockholm to The treatment of pituitary adenoma was an early clinical
Uppsala, a distance of over 70 km, for proton beam treatment. application of the Harvard cyclotron because of the ability to
Given these limitations, Leksell and his collaborators in medical localize the sella turcica, and therefore the pituitary that was
physics developed the Gamma Knife, which utilizes cobalt-60 the treatment target, on plain orthogonal radiographs. A 1968
as a radioactive source and had multiple channels through New England Journal of Medicine article describes 14 acrome-
which to deliver radiation therapy.20 Between 1968 and 1982, galic patients who were followed after stereotactic radiother-
Leksell treated 762 patients with arteriovenous malformations apy, of whom 9 experienced a reduction in size of their hands,
(AVMs) or benign tumors with Gamma Knife-based stereotactic feet or facial features and 8 demonstrated a striking decrease in
radiosurgery.20 The history of Gamma Knife radiosurgery is dis- their level of human growth hormone.24 Kjellberg and col-
cussed in-depth in Chapter 1 of this book. leagues also described their experience with 75 consecutive
At Harvard University, the first cyclotron was built in 1937 patients treated with proton beam therapy for AVMs.25 Lesions
(▶ Fig. 2.1). It was primarily used to produce radioactive iso- were localized using pretreatment angiography and stereotactic
topes for medical purposes. However, the cyclotron was sent techniques. Patients were selected based on inoperability of
from Harvard (Cambridge, Massachusetts) to Los Alamos (New their lesions, most often because these lesions were large, cen-
Mexico) in 1943 to assist in the Manhattan Project, with the trally located, or within the speech areas of the patient’s domi-
promise that the U.S. Government would replace the unit at the nant cerebral hemisphere. Sixty-six of these patients were
conclusion of the Second World War. In 1956 a new, 160-MeV treated without complications. Kjellberg and his colleagues also
cyclotron was built at Harvard with funds from the U.S. Depart- proposed dose-effect curves for radiation necrosis of the brain.
ment of Defense (▶ Fig. 2.2). This was commissioned under the This relationship was the basis for later dosimetry in radiosur-
direction of Robert Wilson and was intended for research in gery both with the linear accelerator and Gamma Knife.26,27
medical physics. However, its uses in physics research waned
over the next 5 years and clinical applications for it became of
increasing interest. Dr. Raymond Kjellberg, a pituitary neuro- 2.2 Fundamental Stereotactic
surgeon at Massachusetts General Hospital in Boston, visited
Stockholm in 1959; inspired by his visit and supported by his
Principles
departmental chief, Dr. William Sweet, Kjellberg began a pro- Although the development of instruments capable of precisely
gram in therapeutic stereotactic proton beam irradiation. In delivering ionizing radiation was critical to the emergence of
1961, the neurosurgical staff at Massachusetts General treated stereotactic radiosurgery, so too was the development of ster-
the first patient to undergo “small-field” irradiation, a 2-year- eotactic methods in the neurosurgical arena. In 1908, Sir Victor
old girl with an inoperable glioma of the diencephalons. A. H. Horsley and his colleague Robert H. Clarke at University

12
The History of Linac and Proton Beam Radiosurgery

College London developed a stereotactic device using a set of


three coordinates in an orthogonal frame of reference by which
to precisely identify the coordinates of an intracranial structure.
They initially utilized this technology to study a desired loca-
tion within the brain of a monkey. The coordinate system used
with their device was relative to bony landmarks of the skull,
including the interior orbital rim and internal auditory canal.
Horsley and Clarke then suggested potential clinical applica-
tions of their stereotactic device,28,29 but their recommenda-
tions were largely ignored by contemporary neurosurgeons.
Nearly three decades later, after modifications for the use of
intracerebral reference structures rather than landmarks on the
skull for the localization of treatment targets, Spiegel and Wycis
introduced stereoencephalotomy and destroyed selected areas
of the thalamus in the treatment of psychiatric disorders
and pain.30,31 Intraoperative radiographs allowed for localiza-
tion of intracranial structures. Soon thereafter, as described
above, Leksell and others developed closed radiosurgical meth-
ods for treating intracranial abnormalities with stereotactically
directed ionizing radiation. Critical subsequent technologic
advances included mechanical X-ray coupling.19 Target localiza-
tion improved dramatically with diagnosis based on computed
Fig. 2.3 A patient undergoing treatment with the stereotacic align-
tomography (CT) and treatment planning within a stereotactic ment for radiosurgery (STAR) device at the Francis H. Burr Proton
coordinate space referenced to the treatment system. 32,33 Mag- Therapy Center at Massachusetts General Hospital in Boston. Patients
netic resonance imaging (MRI) was subsequently used to treated with the device are rotated around a fixed-beam line.
increase the specificity of target localization.34
However, despite these imaging advances in the delineation
of treatment targets, techniques had to be developed for using delivery, in the 1980s linac-based delivery was pioneered.
target information gathered in three-dimensional (3D) imaging Multiple international groups contributed to the development
in conventional X-ray stereotactic space.35 This approach was of linac-based stereotactic radiosurgery. Neurosurgeon Osval-
felt to be less accurate than methods based on CT/MRI imaging. do Betti and engineer Victor Derechinsky in Buenos Aires, and
More recent developments in this field have included in-room neurosurgeon Federico Colombo in Vicenza, Italy, coupled
imaging options such as the two-dimensional (2D)/3D image stereotactic devices to available medical linear accelerators. 38,
registration provided by sophisticated stereotactic treatment- 39,40 On the basis of these investigators’ initial reports, Ken R.

planning software and cone-beam CT devices attached to linear Winston and Wendell Lutz adapted a stereotactic apparatus
accelerator (linac) based treatment systems. and developed a special collimator to ensure positional accu-
Given the desire for improved accuracy of patient positioning, racy for the dependable provision of linac-based radio-
techniques evolved to allow reproducible patient positioning surgery.41,42 This technology was the basis for modern
and treatment with stereotactic radiosurgery. The team at the radiotherapy with linac-based systems. The use of linear
Harvard Cyclotron Laboratory developed a patient-positioning accelerators in radiosurgery remained controversial during
system known as the Stereotactic Alignment for Radiosurgery the 1990s based on the assertion by some clinicians that
(STAR) system.36 Target-centered stereotaxis was the basis for Gamma Knife units had better accuracy than linac units due
the system, with the stereotactic coordinates obtained directly to fewer moving parts. This fueled the development of linac-
from CT, MRI, or angiographic imaging. As with other accelera- based technologies for specific use in radiosurgery. In 1992,
tor systems used in physics research, the STAR system was the first commercially built linear accelerator dedicated to
designed with a fixed-direction, horizontal beam (▶ Fig. 2.3). radiosurgery was installed in Boston. 2,43 Researchers at the
Patients were rotated into different positions in a precise fash- University of Florida in Gainesville also reported their early
ion such that the beam was always aimed at the same target experiences with linac-based stereotactic radiosurgical sys-
within the patient’s head, allowing treatment to be fully isocen- tems.44,45 Subsequent critical developments in radiosurgery
tric. Implantation of radiopaque fiducial markers before treat- included “frameless” technologies, which were initially devel-
ment planning further improved the efficiency of the treatment oped to facilitate neurosurgical applications. 46 Fiducial
system.37 markers on the patient’s scalp were used to translate spatial
data provided by a surgical probe onto preoperative CT or
MRI scans. Several refinements were made to the initial tech-
2.3 Introduction of Linear nology for linac-based radiosurgery, 47,48 which has since has
Accelerator–Based Radiosurgery become commonly used as a result of significant improve-
ments in patient comfort, the increased availability of
The 1980s were a time of rapid growth in the development of advanced in-room imaging options, and reduced need for
stereotactic radiosurgery. Whereas the early years of radio- departmental staff resources as compared with those required
surgery predominantly focused on heavy particle-based for frame-based systems.

13
The History of Linac and Proton Beam Radiosurgery

[3] Rutherford E. Collisions of alpha particles with light atoms. III. Nitrogen and
2.4 Migration from Clinical Labo- oxygen atoms. Philos Mag 1919; 37: 571–580

ratories to Hospital-Based Facilities [4] Rutherford E. Collisions of alpha particles with light atoms. IV. An anomalous
effect in nitrogen. Philos Mag 1919; 37: 581–587
[5] Chadwick J. Possible existence of a neutron. Nature 1932; 129: 312
Early facilities for particle therapy were housed either within [6] Stone RS. Neutron therapy and specific ionization. Am J Roentgenol Radium
national laboratories or in freestanding structures. The Lawrence Ther 1948; 59: 771–785
Berkeley Laboratory was an example of the former and the Har- [7] Wilson RR. Radiological use of fast protons. Radiology 1946; 47: 487–491
vard Cyclotron Laboratory of the latter. However, as the thera- [8] Creutz EC, Wilson RR. Mono-energetic protons from a cyclotron. Rev Sci Ins-
trum 1946; 17: 385–388
peutic utilization of particle beams assumed greater importance,
[9] Lawrence EO, Edlefsen NE. On the production of high-speed protons. Science
there was a need for the deliberate construction of facilities 1930; 72: 376–377
intended to provide optimal patient care. In 1990 the first hospi- [10] Tobias CA, Anger HO, Lawrence JH. Radiological use of high energy deuterons
tal-based proton facility opened at the Loma Linda Medical Cen- and alpha particles. Am J Roentgenol Radium Ther Nucl Med 1952; 67: 1–27
[11] Tobias CA, Van Dyke DC, Simpson ME, Anger HO, Huff RL, Koneff AA. Irradia-
ter in Loma Linda, California.49,50 All clinical treatments provided
tion of the pituitary of the rat with high energy deuterons. Am J Roentgenol
through the Massachusetts General Hospital took place at the
Radium Ther Nucl Med 1954; 72: 1–21
Harvard Cyclotron Laboratory in Cambridge, Massachusetts, [12] Williams JE. Donner Laboratory: the birthplace of nuclear medicine. J Nucl
until 2001, when a new proton beam facility was constructed Med 1999; 40: 16N–20N, 18N, 20N
and opened at the hospital’s main campus in Boston with fund- [13] Lawrence JH, Tobias CA, Born JL, et al. Pituitary irradiation with high-energy
proton beams: a preliminary report. Cancer Res 1958; 18: 121–134
ing from the U.S. National Cancer Institute. Hundreds of centers
[14] Lawrence JH. Proton irradiation of the pituitary. Cancer 1957; 10: 795–798
around the world are now using linear accelerators, proton [15] Suit HD, Chu W. History of charged particle radiotherapy. In: DeLaney TF,
beam accelerators, or the Gamma Knife to deliver stereotactic Kooy HM, eds. Proton and Charged Particle Radiotherapy. 1st ed. Philadelphia,
treatment to hundreds of thousands of patients a year. PA: Lippincott Williams & Wilkins; 2008:1–7
[16] Larsson B, Leksell L, Rexed B, Sourander P, Mair W, Andersson B. The high-en-
ergy proton beam as a neurosurgical tool. Nature 1958; 182: 1222–1223

2.5 Future Directions [17] Leksell L, Larsson B, Andersson B, Rexed B, Sourander P, Mair W. Lesions in
the depth of the brain produced by a beam of high energy protons. Acta Ra-
As linac-based therapy expanded and more proton beam-based diol 1960; 54: 251–264
[18] Larsson B, Leksell L, Rexed B, Sourander P. Effect of high energy protons on
treatment centers opened, stereotactic radiation expanded from
the spinal cord. Acta Radiol 1959; 51: 52–64
the treatment solely of intracranial lesions to that of other ana- [19] Leksell L. Stereotaxis and Radiosurgery: An Operative System. 1st ed. Spring-
tomical sites, including the spine, chest, abdomen, and pelvis. field, IL: Charles C. Thomas; 1971
The fundamental principles of intracranial stereotactic radio- [20] Leksell L. Stereotactic radiosurgery. J Neurol Neurosurg Psychiatry 1983; 46:
797–803
therapy, precise anatomical localization and high dose per frac-
[21] Kirn TF. Proton radiotherapy: some perspectives. JAMA 1988; 259: 787–788
tion treatment, form the basis of stereotactic body radiotherapy. [22] Kjellberg RN, Koehler AM, Preston WM, Sweet WH. Stereotaxic instrument
The development of the CyberKnife (Accuray Inc., Sunnyvale, for use with the Bragg peak of a proton beam. Confin Neurol 1962; 22: 183–
CA), a linac-based system, obviated the need for rigid fixation of 189
the anatomical site to be treated.51,52 Intracranial applications of [23] Kjellberg RN, Sweet WH, Preston WM, Koehler AM. The Bragg peak of a pro-
ton beam in intracranial therapy of tumors. Trans Am Neurol Assoc 1962; 87:
radiosurgery have also significantly expanded since the original
216–218
treatments of AVMs and pituitary adenomas. In addition to the [24] Kjellberg RN, Shintani A, Frantz AG, Kliman B. Proton-beam therapy in acro-
evolution of treatment technologies and clinical applications for megaly. N Engl J Med 1968; 278: 689–695
stereotactic radiotherapy, the antitumor effects of single-dose [25] Kjellberg RN, Hanamura T, Davis KR, Lyons SL, Adams RD. Bragg-peak proton-
radiotherapy and accurate radiobiological modeling are active beam therapy for arteriovenous malformations of the brain. N Engl J Med
1983; 309: 269–274
areas of research and are becoming better understood.
[26] Flickinger JC. An integrated logistic formula for prediction of complications
In 1993, Charles B. Wilson, Professor of Neurosurgery at the from radiosurgery. Int J Radiat Oncol Biol Phys 1989; 17: 879–885
University of California, San Francisco, and founder of its Brain [27] Saunders WM, Winston KR, Siddon RL, et al. Radiosurgery for arteriovenous
Tumor Research Center, stated in the foreword to the first edi- malformations of the brain using a standard linear accelerator: rationale and
technique. Int J Radiat Oncol Biol Phys 1988; 15: 441–447
tion of Stereotactic Radiosurgery that “Ultimately, the results
[28] Horsley V, Clarke R. The structure and functions of the cerebellum examined
and complications in comparable groups of patients treated by a new moethod. Brain 1908; 31: 45–124
with radiosurgery must be judged against those obtained with [29] Clarke R, Horsley V. On a method of investigating the deep ganglia and tracts
other therapeutic approaches.... Objective comparisons are the of the central nervous system (cerebelleum). BMJ 1906; 2: 1799–1800
ultimate yardstick of therapeutic superiority.”53 More than two [30] Spiegel EA, Wycis HT. Mesencephalothalamotomy for relief of intractable
pain. Am J Med 1949; 6: 504
decades later, Dr. Wilson’s counsel has been followed, and peer-
[31] Spiegel EA, Wycis HT, Marks M, Lee AJ. Stereotaxic apparatus for operations
reviewed data support the ever-expanding utilization of radio- on the human brain. Science 1947; 106: 349–350
surgery. His wisdom regarding the metrics by which radiosur- [32] Bergström M, Greitz T. Stereotaxic computed tomography. AJR Am J Roent-
gery should be evaluated must be kept in mind as this field of genol 1976; 127: 167–170
medicine continues to evolve and grow in the decades to come. [33] Boëthius J, Bergström M, Greitz T, Ribbe T. CT localization of stereotactic sur-
gery. Appl Neurophysiol 1980; 43: 164–169
[34] Leksell L, Herner T, Leksell D, Persson B, Lindquist C. Visualisation of stereo-

References tactic radiolesions by nuclear magnetic resonance. J Neurol Neurosurg Psy-


chiatry 1985; 48: 19–20
[35] De Salles AA, Asfora WT, Abe M, Kjellberg RN. Transposition of target infor-
[1] Leksell L. The stereotaxic method and radiosurgery of the brain. Acta Chir
mation from the magnetic resonance and computed tomography scan images
Scand 1951; 102: 316–319
to conventional X-ray stereotactic space. Appl Neurophysiol 1987; 50: 23–32
[2] Lunsford LD, Alexander E, Loeffler JS. General introduction: history of radio-
[36] Chapman P, Ogilvy C, Butler W. A new stereotactic alignment system for
surgery. In: Alexander E, Loeffler JS, Lunsford LD, eds. Stereotactic Radiosur-
charged-particle radiosurgery at the Harvard Cyclotron Laboratory, Boston.
gery. New York, NY: McGraw-Hill; 1993:1–4

14
The History of Linac and Proton Beam Radiosurgery

In: Alexander E, Loeffler JS, Lunsford LD, eds. Stereotactic Radiosurgery. 1st [46] Roberts DW, Strohbehn JW, Hatch JF, Murray W, Kettenberger H. A frameless
ed. New York. NY: McGraw-Hill; 1993:105–108 stereotaxic integration of computerized tomographic imaging and the oper-
[37] Gall KP, Verhey LJ, Wagner M. Computer-assisted positioning of radiotherapy ating microscope. J Neurosurg 1986; 65: 545–549
patients using implanted radiopaque fiducials. Med Phys 1993; 20: 1153– [47] Kato A, Yoshimine T, Hayakawa T, et al. A frameless, armless navigational sys-
1159 tem for computer-assisted neurosurgery. Technical note. J Neurosurg 1991;
[38] Betti OO, Galmarini D, Derechinsky V. Radiosurgery with a linear accelerator. 74: 845–849
Methodological aspects. Stereotact Funct Neurosurg 1991; 57: 87–98 [48] Tan KK, Grzeszczuk R, Levin DN, et al. A frameless stereotactic approach to
[39] Colombo F, Benedetti A, Pozza F, et al. Stereotactic radiosurgery utilizing a lin- neurosurgical planning based on retrospective patient-image registration.
ear accelerator. Appl Neurophysiol 1985; 48: 133–145 Technical note. J Neurosurg 1993; 79: 296–303
[40] Colombo F, Benedetti A, Pozza F, et al. External stereotactic irradiation by lin- [49] Slater JM, Miller DW, Archambeau JO. Development of a hospital-based pro-
ear accelerator. Neurosurgery 1985; 16: 154–160 ton beam treatment center. Int J Radiat Oncol Biol Phys 1988; 14: 761–775
[41] Lutz W, Winston KR, Maleki N. A system for stereotactic radiosurgery with a [50] Slater JM, Archambeau JO, Miller DW, Notarus MI, Preston W, Slater JD. The
linear accelerator. Int J Radiat Oncol Biol Phys 1988; 14: 373–381 proton treatment center at Loma Linda University Medical Center: rationale
[42] Winston KR, Lutz W. Linear accelerator as a neurosurgical tool for stereotactic for and description of its development. Int J Radiat Oncol Biol Phys 1992; 22:
radiosurgery. Neurosurgery 1988; 22: 454–464 383–389
[43] Kooy HM, Nedzi LA, Loeffler JS, et al. Treatment planning for stereotactic ra- [51] Adler JR Jr, Chang SD, Murphy MJ, Doty J, Geis P, Hancock SL. The Cyberknife:
diosurgery of intra-cranial lesions. Int J Radiat Oncol Biol Phys 1991; 21: a frameless robotic system for radiosurgery. Stereotact Funct Neurosurg
683–693 1997; 69: 124–128
[44] Friedman WA, Bova FJ, Spiegelmann R. Linear accelerator radiosurgery at the [52] Chang SD, Adler JR. Robotics and radiosurgery—the cyberknife. Stereotact
University of Florida. Neurosurg Clin N Am 1992; 3: 141–166 Funct Neurosurg 2001; 76: 204–208
[45] Friedman WA, Bova FJ. The University of Florida radiosurgery system. Surg [53] Alexander E III, Loeffler JS, Lunsford LD, eds. Stereotactic Radiosurgery. 1st ed.
Neurol 1989; 32: 334–342 New York, NY: McGraw-Hill; 1993

15
The History of CyberKnife Radiosurgery

3 The History of CyberKnife Radiosurgery


Aditya Iyer, Steven D. Chang, and John R. Adler, Jr.

In the late 1940s, Ernest Spiegl, a neurosurgeon at Temple


Key Points University in Philadelphia, combined Horsley’s principles of
frame-based localization with plain-film radiography and cre-
● The CyberKnife is capable of delivering large doses of fo- ated a Cartesian atlas of the human head, naming the method
cused ionizing radiation to well-defined targets. stereoencephalotomy.2 Given the high intraoperative mortality
● It consists of a lightweight linear accelerator mounted on a in open cranial surgery for movement disorders at the time,
mobile industrial robotic arm. Spiegl and colleagues initially used frame-based localization to
● No rigid immobilization of the patient is required to undergo target the extrapyramidal system.3 However, it was not until
CyberKnife treatment. Swedish neurosurgeon Lars Leksell, the father of modern ster-
● The CyberKnife can be used for both isocentric and noniso- eotactic radiosurgery, combined the principles of image guid-
centric treatment ance with the nascent practice of therapeutic radiation that
● The CyberKnife can target lesions throughout the body. radiation was successfully used to treat neurological disorders.
In 1947 Leksell developed the first arc-based stereotactic appa-
ratus for inserting electrodes and needles into desired locations
within the brain with submillimeter accuracy.4 During Leksell’s
3.1 Introduction time, medical particle accelerators (such as the linac used in the
CyberKnife, and the cyclotron) were too expensive or cumber-
Over the past several decades, the CyberKnife (Accuray Inc.,
some for medical purposes. Working alongside Swedish physi-
Sunnyvale, CA) has become an important tool for treating
cists Kurt Liden and Börje Larsson of Uppsala University, Leksell
lesions with maximal precision through the entire body.
concluded that gamma rays were the most practical source of
Since its introduction into clinical use in 1994, the Cyber-
therapeutic radiation at the time and proceeded to develop the
Knife, which can be used to treat lesions anywhere in the
Gamma Knife, which has become the most widely used radio-
body (in contrast to the Gamma Knife, which is used solely
surgical device.5,6
for treating intracranial lesions), has been used to treat more
than 100,000 patients around the world. The technology that
underlies the CyberKnife derives from the basic principles
that motivate stereotactic radiosurgery as a field, and the his-
3.3 Linear Accelerator Technology
tory of the CyberKnife is therefore inextricably linked to the From the 1920s until the 1950s, linacs remained within the
history of image guidance, therapeutic radiation, and radio- realm of theoretical physicists. The technology was initially
surgery. More specifically, the CyberKnife combines the tech- developed to accelerate charged particles through oscillating
nology of the lightweight linear accelerator (linac) with electrical fields in vacuum-sealed tubing so that the energy
advances in image guidance and robotics. As with all radio- transfer upon collisions of the accelerated particles with other
surgery, the purpose of the CyberKnife is to administer large particles would result in a stream of subatomic particles that
doses of ionizing radiation to destroy well-defined targets physicists could study. During the Second World War, Russell
within the body while minimizing damage to surrounding and Sigurd Varian of Stanford University developed, in their
structures. high-energy physics laboratories, a linear beam vacuum tube
named the klystron that generated X-rays with mega-electron
voltage and served as a high-frequency amplifier.7 Notably, X-
3.2 Image Guidance and rays of this energy were sufficient to penetrate and ablate
biological tissue. The Varians patented this technology and
Radiosurgery in the Decades founded Varian Associates, which ultimately became Varian
before CyberKnife Medical Systems (Palo Alto, CA). The technology was popular-
ized in 1956 after Stanford University radiologist Henry Kaplan
The beginnings of stereotaxis and image guidance in neurosur- used this medical linac to successfully treat a 2-year-old boy
gery date back to the origin of neurosurgery as a medical field with retinoblastoma.8,9
itself. Working alongside mathematician Robert Clark, neuro- Over the next several decades, both the multileaf collimator
surgeon Sir Victor Horsley developed the first tool to localize and achromatic bending magnets were added to the linac,
intracranial structures in three dimensions in the early 20th respectively allowing the more precise contouring of X-ray
century.1 Using a rigid head frame that provided a Cartesian beams and energy stabilization. Additionally, with introduction
coordinate system, Horsley postulated that any point within of the computed tomography (CT) scanner in the 1970s, linac
the cranial vault could be assigned three coordinates relative to technology became more practical for targeting intracranial
the frame, and that clinicians could therefore reliably describe lesions, as described in Chapter 2. Betti and Derechinsky 10
and target intracranial structures. However, this technology described one of the earliest clinical experiences with the linac,
never progressed beyond animal models, and for the few deca- with promising results. Lutz and Winston11 subsequently
des after its origin, stereotaxis remained a concept rather than defined the dosimetry of this method, and the modern linac
an approach integrated into clinical practice. system became commercially available in the 1980s. However,

16
The History of CyberKnife Radiosurgery

as in the case of the Gamma Knife, the original linac relied on


rigid immobilization of the patient to achieve submillimeter
accuracy. Patient movement, in addition to the more subtle
motion of respiration, the beating heart, and blood flow
through arteries and veins, necessitated a rigid frame screwed
into patients’ bone structures to ensure submillimeter accuracy.

3.4 The Early Years of Frameless


Radiosurgery
In 1985, while studying with Lars Leksell at the Karolinska
Institute in Sweden, the American neurosurgeon John Adler
envisioned a way to expand on Leksell’s vision of minimally
invasive neurosurgery by completely abandoning the use of a
head frame altogetherfor such surgery. Although several groups
had in the early 1990s proposed approaches to frameless radio-
surgery, none of these was clinically practical. Bova et al at the
University of Florida, Gainesville, used an infrared tracking sys-
tem that was calibrated with an external frame that was held to
Fig. 3.1 The original Neurotron 1000 designed by John Adler from U.S.
the patient’s body with a bite block.12 However, given the lim- Patent 5,207,223, entitled “Apparatus and Method for Performing
ited computing power of the time, it would often take days to Stereotaxic Surgery.”
weeks for the system to recalculate new targets to account for
patient movement.
The technological opportunities and innovations in the San
Francisco Bay Area’s burgeoning Silicon Valley facilitated the 3.5 The Modern CyberKnife
advent of the CyberKnife. In the early 1990s, while working
The modern CyberKnife consists of a compact, 120-lb, 6-MeV
at Stanford University, John Adler collaborated with Professor
linac capable of providing 6 Gy of radiation per minute. The
Tom Bradford and his graduate student at the Stanford School
linac is mounted to a robotic arm with six degrees of freedom
of Engineering to develop more efficient computer algorithms
(Kuka GmbH, Augsburg, Germany), allowing up to 1600 direc-
for accurately correlating X-ray and computed tomography
tions in which the instrument’s beam can be aimed to reach a
(CT) scans in real time.7 These algorithms would ultimately
lesion. Two orthogonally mounted X-ray generators, attached to
be used to identify and correlate the motion of set anatomical
the ceiling and the wall, respectively, are registered to X-ray
landmarks so that radiation beams could be redirected to
images that have been extracted from a CT scan done for the
remain on a treatment target despite movement. Additional-
initial planning of a patient’s treatment. Any movements of the
ly, in the 1980s, robots used in the manufacturing industry
target region in the patient’s body are corrected both by read-
were becoming increasingly portable and offering an
justing the linac and rotating the patient on a mobile treatment
expanded range of motion, making them ideal platforms for
bed.15 This provides submillimeter accuracy comparable to that
holding a mobile radiation source such as the linac. However,
of frame-based systems.16
the size and weight of the linacs of that the time made this
Given the maneuverability of its robotic arm, the CyberKnife
unfeasible.
system can be used to treat both isocentrically (in which all of
A small Silicon Valley-based company named Schonberg En-
the instrument’s beams converge at a single point) and noniso-
gineering was working on miniaturizing the linear accelerator,
centrically; the instrument’s beams are not constrained to pass
and by the late 1980s had developed an ultra-lightweight linac
through the geometric center of a target lesion, but rather
weighing less than 300 pounds. This gave way to Adler’s first
through any part of the lesion. This allows more uniform radia-
design for a frameless stereotactic radiosurgery system
tion dosage throughout the entire region of a target, as opposed
(▶ Fig. 3.1). His design included a linac mounted on a mobile
to a maximum dose of radiation at the geometric center and a
industrial robot. As patients moved either voluntarily or invol-
declining dose to the remainder of the target. Additionally,
untarily, two X-ray generators, oriented orthogonally to one
frameless radiosurgery makes fractionation and hypofractiona-
another, continuously generated plain-film images that were
tion possible both for intra- and extracranial structures because
then subjected to software processing to correlate the motion
there is no need to apply a rigid frame to the patient. Before the
of fixed, radiographically visible landmarks. The robotically
introduction of the CyberKnife, no method existed for fractio-
mounted linac would then readjust the direction of its beams
nating the treatment of extracranial targets with stereotactic
so that they remained on target. If submillimeter accuracy was
precision.
not achieved, the treatment would stop until the system could
readjust. With this idea, Adler and the Schonberg brothers were
awarded U.S. Patent 5,207,223 in 1993, and Accuray, Inc., was
founded. Originally named the Neurotron 1000,13 the initial
3.6 Extracranial Radiosurgery
CyberKnife was a 300-lb, 6-MeV linac attached to an industrial Until recently, the applications of radiosurgery were limited to
robot designed by GMF.14 the brain and upper cervical spine. However, over the past

17
The History of CyberKnife Radiosurgery

decade, the CyberKnife has been used to treat other spinal


lesions. Traditional spine radiosurgery had relied on surgically
References
implanted fiducials to account for nonrigid deformation of the [1] Clarke RH, Horsley V. THE CLASSIC: on a method of investigating the deep
spine as well as for voluntary and involuntary patient motion of ganglia and tracts of the central nervous system (cerebellum). Br Med J
1906:1799–1800. Clin Orthop Relat Res 2007; 463: 3–6
the types usually encountered in such surgery. In the early
[2] Spiegel EA, Wycis HT, Thur C. The stereoencephalotome (model III of our ster-
2000s, Accuray, Inc., of Sunnyvale, California, developed a fidu- eotaxic apparatus for operations on the human brain). J Neurosurg 1951; 8:
cial-free system for CyberKnife that continuously refers to 452–453
radiopaque skeletal structures and is capable of targeting [3] Spiegel EA, Wycis HT, Marks M, Lee AJ. Stereotaxic apparatus for operations
on the human brain. Science 1947; 106: 349–350
lesions with great precision.
[4] Leksell L. The stereotaxic method and radiosurgery of the brain. Acta Chir
The CyberKnife can also be used to treat lesions anywhere in Scand 1951; 102: 316–319
the visceral organs of the body. Lesions in the chest, abdomen, [5] Larsson B, Lidén K, Sarby B. Irradiation of small structures through the intact
and pelvis move significantly more than those in the brain and skull. Acta Radiol Ther Phys Biol 1974; 13: 512–534
spinal cord during treatment, particularly with respiration. [6] Leksell L. Stereotactic radiosurgery. J Neurol Neurosurg Psychiatry 1983; 46:
797–803
Treatment begins when gold fiducial markers are inserted per-
[7] Adler JR. Accuray, Incorporated: a neurosurgical business case study. Clin
cutaneously within the target, which the CyberKnife’s orthogo- Neurosurg 2005; 52: 87–96
nal X-ray system detects. Additionally, infrared cameras track [8] Ginzton EL, Mallory KB, Kaplan HS. The Stanford medical linear accelerator. I.
light-emitting diodes (LEDs) attached to the patient’s skin. The Design and development. Stanford Med Bull 1957; 15: 123–140
[9] Kaplan HS, Bagshaw MA. The Stanford medical linear accelerator. III. Application
Synchrony system for the CyberKnife continuously correlates
to clinical problems of radiation therapy. Stanford Med Bull 1957; 15: 141–151
the position of the infrared-emitting LEDs with the implanted [10] Betti O, Derechinsky V. Multiple-beam stereotaxic irradiation] Neurochirur-
gold markers, and the instrument’s radiation beams are gie 1983; 29: 295–298
adjusted accordingly.17 [11] Winston KR, Lutz W. Linear accelerator as a neurosurgical tool for stereotactic
In summary, the CyberKnife allows the safe and effective radiosurgery. Neurosurgery 1988; 22: 454–464
[12] Bova FJ, Buatti JM, Friedman WA, Mendenhall WM, Yang CC, Liu C. The Uni-
treatment of lesions throughout the body with minimal com-
versity of Florida frameless high-precision stereotactic radiotherapy system.
plications. The technology that underlies the CyberKnife is Int J Radiat Oncol Biol Phys 1997; 38: 875–882
the culmination of more than a quarter century of advances [13] Cox RS. Positioning accuracy of the Neurotron 1000. Radiother Oncol 1995;
in image guidance, radiation therapy, robotics, and software 32 971: 301
engineering. As radiosurgery evolves, new technologies will [14] Adler JR, Cox RS. Preliminary experience with the CyberKnife: image-guided
stereotactic radiosurgery. In: Alexander III E, Kondziolka D, Loeffler JS, eds.
emerge to allow shorter treatment times with even greater
Radiosurgery. Basel, Switzerland:Karger; 1996:316–326
precision and less patient discomfort. The CyberKnife repre- [15] Romanelli P, Adler JR Jr. Technology Insight: image-guided robotic radiosur-
sents a major step in this evolution of radiosurgical devices. gery—a new approach for noninvasive ablation of spinal lesions. Nat Clin
The history of the CyberKnife demonstrates that multidisci- Pract Oncol 2008; 5: 405–414
[16] Chang SD, Main W, Martin DP, Gibbs IC, Heilbrun MP. An analysis of the accu-
plinary collaboration is essential to advance the practice of
racy of the CyberKnife: a robotic frameless stereotactic radiosurgical system.
medicine, and that industry and academia can successfully Neurosurgery 2003; 52: 140–146, discussion 146–147
work together to better the lives of patients throughout the [17] Ozhasoglu C, Saw CB, Chen H, et al. Synchrony—cyberknife respiratory com-
world. pensation technology. Med Dosim 2008; 33: 117–123

18
4 The Radiobiology of Radiosurgery 20
Part II
5 The Physics of Radiosurgery 30
Radiosurgical Fundamentals 6 Radiosurgical Devices 42

7 Critical Structures and Tolerance of the


Central Nervous System 52

8 The Neuropathology of Radiosurgery 58

II
The Radiobiology of Radiosurgery

4 The Radiobiology of Radiosurgery


John C. Flickinger

4.2 Categories of Normal Tissue


Key Points
Response to Radiation
● The linear-quadratic equation cannot reliably extrapolate
from data for conventional fractionated radiotherapy beyond Normal tissue is typically categorized as being either early
dose-fractions of 6–10 Gy to estimate equivalent radiation responding or late responding in its response to radiation.
effects for the high-dose (12–25 Gy) single-fraction radiosur- Early-responding tissues have faster-growing cells as in the case
gery. Consistently high negative values were found for alpha/ of epithelial or mucosal layers that are continuously replaced by
beta ratios calculated from radiosurgery responses that fast-growing stem cells. This occurs in the oral mucosa, skin,
conflict with values seen with conventional fractionated hair follicles, and intestinal crypts where the responses can be
radiotherapy. mucositis, skin redness, hair loss, and diarrhea. Late-responding
● Radiation injury models need to take into account contribu- or slow-growing tissues include that of the brain and spinal
tions from the target or tumor affecting radiation injury cord where the late responses of concern are radiation necrosis
reactions in the surrounding normal tissue. of brain or radiation myelopathy.
● Much of the radiation response of a radiosurgical target and Another way to categorize the responses of organs or tissues
surrounding tissue appears to be mediated by the to radiation is based on whether they are serial or parallel
supporting vasculature. organs. Organs with a parallel structure have redundancy as the
● Areas of tumor hypoxia in heterogeneously enhancing key defense against radiation injury. Organs such as lung, kid-
tumors may lead to radioresistance that can be addressed ney, and liver are relatively sensitive to irradiation of the whole
through different approaches including fractionated organ, but can withstand very high doses to portions of the
radiotherapy. organ (with loss of function in those high-dose regions) with-
out significant loss of overall function if the radiation dose to a
reasonable portion of the organ is kept below a certain rela-
tively low threshold. Organs with a serial structure—such as the
4.1 Introduction spinal cord or esophagus—can generally withstand higher doses
to small portions of the organ without loss of function of that
Radiobiology is the study of tumor and normal tissue res- portion (or the overall organ); unfortunately, when injury to
ponse to ionizing radiation with the goal of isolating these even a small portion of that organ occurs, considerable loss of
responses to promote more tumor-cell death with less nor- function develops as a result.
mal tissue injury. In comparison with standard large-field
radiotherapy techniques (without image guidance), radiosur-
gery techniques change this balance first and foremost by 4.3 What Is the Rationale for
eliminating the need to irradiate a 1–2 cm margin around a
target or tumor volume to full treatment doses that allows Fractionation in Conventional
for positional errors in the radiation target definition and
delivery. Radiosurgery techniques also exploit the ability of
Radiotherapy?
small volumes of normal organs to tolerate much higher radi- Prior to the introduction of radiosurgery, almost all clinical
ation doses without injury than large volumes of the same radiation therapy of intracranial targets was fractionated. From
organ. This dose-volume effect for radiation becomes highly the reaction of skin that received much higher doses with
pronounced at very small volumes such as those treated in external beam orthovoltage radiotherapy used before the intro-
radiosurgery. By using stereotactic fixation and localization duction of megavoltage radiotherapy sources (cobalt 60 and the
techniques, radiosurgery allows clinicians to eliminate the linear accelerator), it became obvious that early and late normal
need to expand radiation treatment volumes by adding mar- tissue reactions could be limited through fractionation while
gins around well-defined target/tumor volumes to correct for maintaining high response rates in treating malignant tumors.
the localization and targeting errors seen with conventional The radiobiological analysis of a few cell culture lines of fast-
radiotherapy setups. The highly conformal radiation-dose dis- growing malignant tumors and clinical experience with tumor
tributions delivered through multiple beams using radiosur- control and complications from common fast-growing malig-
gery techniques reduce the amount of normal tissue nant tumors commonly treated with fractionated radiotherapy
subjected to full treatment doses within the intracranial strongly supported this observation. These studies showed that
treatment volume and also reduce the radiation dose falloff increased fractionation (reducing daily radiation dose-fractions
into surrounding normal tissue outside of the target volume. to the range of 1.8 to 2.0 Gy per fraction [/fr] and treating with
Treatment can therefore change from multiple small dose- more fractions to equal or slightly higher total doses) preferen-
fractions that a large volume of surrounding normal tissue tially spared normal tissue from radiation injury without much
can tolerate to high-dose single-fraction radiation doses that change in tumor control. With the widespread use of 1.8- to
markedly smaller, thin rims of normal tissue adjacent to 2.0-Gy fractions, radiation oncologists became comfortable
radiosurgical targets can tolerate. treating all tumors with this fraction formula (both rapidly

20
The Radiobiology of Radiosurgery

growing malignant and slow-growing benign tumors), espe- dominant mode of cell death in tumors seems to be mediated
cially because most information about normal tissue tolerance through radiation effects on endothelial cells in the supporting
was garnered using these fractionation schemes. vasculature.4 These effects on the vasculature seem to become
Slow-growing benign tumors have been notoriously difficult pronounced with dose-fractions at 8 to 12 Gy or higher. Patho-
to study in cell cultures or animal models. Their radiobiological logical studies of tumors resected after radiosurgery also indi-
responses—and particularly how these responses change with cate that vascular endothelium may be the primary target
different fractionation—are poorly defined. Prior to the advent responding to radiosurgery.5 This vasculature effect seems to
of radiosurgery, few radiation oncologists had more than lim- explain why single-fraction radiosurgery provides similarly
ited experience treating with high doses of single-fraction radi- high tumor rates for very different tumors found to differ
ation. Stereotactic radiosurgery provided clinicians with the widely by being either highly resistant or highly sensitive to
ability to administer high single doses of radiation to intracrani- radiation whether in cell culture models or in clinical experi-
al targets with relative safety. Radiosurgery ushered in a new ence with conventional fractionated radiotherapy.
era of understanding how different approaches to radiation Optimizing the outcome of any clinical intervention (radia-
treatment planning and radiobiology may be modified in the tion, drug therapy, or surgery) usually requires striking a bal-
clinic to achieve goals previously thought unreachable. ance between maximizing the desired outcome of the
intervention (tumor cure, vascular malformation obliteration,
etc.) while minimizing undesired sequelae of the intervention
4.4 What Exactly Happens When a (complications). This balance can be represented in many clini-
Tissue Is Radiated? cal situations by a pair of dose-response curves representing
cure (tumor control or arteriovenous malformation [AVM]
When a person undergoes X-ray treatment or cobalt irradiation, obliteration) and complications (radiation injury) as shown in
the target region is exposed to a high-energy photon beam. The (▶ Fig. 4.1). The separation between the curves has been
beam interacts with the tissue at the target, producing a cas- referred to as the therapeutic window, whereas the ratio of cure
cade of ionizing electrons that result in biochemical damage to complication risk is referred to as the therapeutic ratio.
within the cells exposed to the radiation. Both single-stranded Radiosurgery exploits the radiobiological fact that smaller vol-
and double-stranded DNA breaks may occur. Depending on umes of normal tissue can withstand much higher doses of
how much biochemical damage occurs and how much of that radiation than larger volumes of normal tissue. Reducing the
damage can be repaired by the cell, the consequences lead to volume of tissue irradiated to high doses reduces the complica-
necrosis (rapid cell death), apoptosis (slow programmed cell tion risk for any dose given to the target, shifting the complica-
death), or cell survival (with or without survival of abilities such tion dose-response curve for the treatment to the right, further
as further mitosis, hormonal secretion, etc.). The exact biology away from the complication curve, thereby improving the ther-
of these injury and repair responses is being unraveled to show apeutic ratio. The effect on both curves must be adequately
increasing complexity with potential points of modification. 1,2,3 assessed to evaluate potential radiation dose-response modifi-
Tumor control by radiation treatment comes not only through cations (such as altered fractionation; radiation protectors; or
radiation directly affecting tumor cells, but also through effects radiosensitizing drugs, such as some chemotherapy or hypoxic
on the adventitia, most notably the supporting vasculature. 4 radiation sensitizers) to find if they improve the therapeutic
Within the range of doses commonly used for radiosurgery, the ratio.

Fig. 4.1 Typical sigmoid dose-response curves for


tumor control and complications and the effect
of reducing treatment volume on the complica-
tion curve.

21
The Radiobiology of Radiosurgery

4.5 How Do We Estimate Effects one dose/fr d1 and total dose D1 to a different fractionation
scheme with dose/fr d2 and total dose D2 according to a given
of Fractionating Radiation? alpha/beta (α/β) ratio as follows:6
 
The linear-quadratic (LQ) formula is the most widely accepted Gy
Total dose D1 Gy  d 1 þ
way to represent fractionation effects in radiotherapy. 6–11 It fr 
Gy
relates the log of surviving cells after a dose-fraction of ionizing ¼ total dose D2 Gy  d 2 þ ð4:2Þ
fr
radiation as being proportional to a linear function of the dose/
fr (consisting of the dose/fr × a coefficient, alpha [α]) plus a sec-
ond quadratic function (consisting of the dose/fr2 × a second Another formula useful in the clinic is the expression for NTD 2,
coefficient, beta [β]). This LQ formula forms a power series the normalized total dose at 2-Gy/fr, which is the equivalent
modeling radiation cell killing with a combination of linear, total dose administered with daily 2-Gy fractions using a given
single-hit kinetics (which could mathematically represent dou- alpha/beta value.6 NTD2 for a given alpha/beta ratio α/β can be
ble-stranded DNA breaks leading to cell death) and quadratic, calculated for a course of radiotherapy given with a dose/fr of d
double-hit kinetics (which could mathematically represent a Gy/fr with the following equation:6,11
critical accumulation of single-stranded DNA breaks leading to h i
    d Gy
cell death). The LQ formula represents the chances of either Gy fr þ
NTD2 ¼ Total dose at d h i ð4:3Þ
tumor cure or normal tissue injury, P(cure or complication), fr 2 Gy þ fr
from a single fraction of radiation of dose, d, by the following
probabilistic double-exponential equation:
  ▶ Table 4.1 shows the 2-Gy/fr equivalent doses predicted for
Pðcure or complicationÞ ¼ EXP K  EXP d  d2
different single-fraction radiosurgery doses with the LQ formu-
ð4:1Þ
la using different alpha/beta ratios.
Equations 4.2 and 4.3 ignore the effect of overall treatment
where EXP represents the number “e” (2.7183, used in natural time, which should be negligible for central nervous system tis-
logarithms) raised exponentially to the power of the terms that sue and benign tumors in normal circumstances. A time-
follow, K represents the number of target clonogens, whereas α dependent repopulation term should be added for protracted
and β are the linear and quadratic coefficients. fractionated treatment of fast-growing malignant tumors.
This mathematical expression describes sigmoid-shaped Repopulation of malignant tumors accelerates 2 to 3 weeks
dose-response curves with the probability of cure or complica- after the start of radiotherapy. Because of clinical studies that
tions increasing with dose from 0% to a limit of 100% as the related poor tumor control from head and neck cancer radio-
dose is increased (▶ Fig. 4.1). Although used in research, this therapy with increasing overall treatment time, split-course
expression of the LQ formula is virtually never used in day-to- radiotherapy with planned 2-week breaks in the middle to alle-
day clinical radiotherapy practice. viate mucositis are no longer routinely used.
A much more useful calculation in the clinic is the formula to The ratio of the alpha coefficient divided by the beta coeffi-
equate the effect of a fractionated course of radiotherapy with cient (the alpha/beta ratio) provides a useful parameter to

Table 4.1 Calculation of radiobiologically equivalent dose (normalized total dose) for fractionated radiotherapy with 2-Gy fractions (NTD 2) from single-
fraction radiosurgery according to the linear-quadratic equation using different values of alpha/beta (and no time correction)
NTD2 (α/β = 10) equivalent NTD2 (α/β = 2) equivalent NTD2 (α/β = 1) equivalent NTD2 (α/β = 0) equivalent
dose (Gy) for early-reacting dose (Gy) for late-reacting dose (Gy) with a lower dose (Gy) for model with no
Single-fraction dose (Gy) tissue tissue (brain, etc.) value of α/β α component

4 4.7 6 6.7 8

6 8.0 12 14.0 18

8 12.0 20 24.0 32

10 16.7 30 36.7 50

12 22.0 42 52.0 72

14 28.0 56 70.0 98

16 34.7 72 90.7 128

18 42.0 90 114.0 162

20 50.0 110 140.0 200

22 58.7 132 168.7 242

24 68.0 156 200.0 288

NTD2 equivalent values of 52-72 Gy from α/β values of 1 to 0 appear to reflect observed risks of optic neuropathy for a single fraction dose of 12 Gy.

22
The Radiobiology of Radiosurgery

quantify fractionation effects in target tissues and tumors. With limited extrapolations from one course of fractionated radio-
the alpha/beta ratio for a given organ or tumor, we can estimate therapy to another with different-sized dose-fractions, particu-
the total dose and number of fractions needed to change from larly within a dose-fractions range of 1 to 8 Gy. Extrapolating
treatment with one dose per fraction to another dose per frac- from 1.8- to 2.0-Gy fractions used in conventionally fractio-
tion with the same effect on that tissue or tumor. Because dif- nated radiotherapy to the high single-fraction doses commonly
ferent kinds of tumors, different organs, and even different used in radiosurgery stretches the formula beyond limits of
types of injury reactions in the same organ (e.g., skin redness accuracy.
vs. late skin fibrosis) have different alpha/beta ratios, changing ▶ Table 4.1 takes commonly used single-fraction radiosur-
to a different fractionation scheme with an equivalent effect gery doses and relates predicted equivalent doses for fractio-
calculated for one organ will lead to a different effect on organs nated radiotherapy with 2-Gy fractions (NTD2) using alpha/beta
or tumors with different alpha/beta ratios. This became appa- values of 10 for early-reacting tissues and 2 for late-reacting tis-
rent when early radiation therapists tried switching to cheaper sue (which is the accepted value for brain tissue from large
and more convenient hypofractionated radiation schemes with studies of fractionated radiotherapy data). The last column pro-
larger dose-fractions producing similar acute skin reactions and vides dose-equivalent dose calculations using an alpha/beta val-
found increased late complications. ue of zero (specifically, α = 0), making it a quadratic-exponential
Different tissues can be lumped into categories by their alpha/ formula rather than a true LQ formula. A conservative estimate
beta ratios as either early-responding tissues (fast-growing with of the NTD2 equivalent for brain injury is easily calculated using
injury responses such as mucositis or skin erythema occurring an alpha/beta value of zero because it is simply one-half the sin-
relatively short time after radiation) or late-responding tissues gle-fraction dose squared. Using an alpha/beta value of zero is
(slow-growing tissues that take a longer time after radiation quite a stretch from the theoretical basis of the formula, which
exposure for injury reactions to become apparent). Studies of presupposes some contribution with single-hit kinetics (the
fractionation effects in radiotherapy clinics and in animal mod- alpha component) corresponding to direct double-stranded
els found brain and spinal cord to be late-responding tissues, rel- DNA breakage. From what is known about optic nerve tolerance
atively more sensitive to large dose-fractions, with alpha/beta (discussed in the next section), an alpha/beta ratio somewhere
ratios around 2. Faster-growing, faster-responding tissues such between 0 and 1 seems to fit the data for optic nerve injury risk
as skin erythema or mucositis reactions have alpha/beta values with radiosurgery doses near 12 Gy.
in the intermediate range of 5 to 8, whereas many fast-growing
malignant tumors have alpha/beta values closer to 10.6 Smaller
radiation dose-fraction schedules cause proportionally less
injury to normal tissues (or a limited number of tumors) with a
4.7 What Are the Optic Nerve
low alpha/beta ratio (a smaller alpha or single-hit component to Tolerance Doses for Radiosurgery?
radiation cell-kill kinetics) compared with malignant tumors (or
An early analysis of optic nerve complications in the combined
faster-responding tissues) with higher alpha/beta ratios.
Harvard University and University of Pittsburgh experience rec-
Because most malignant tumors have higher alpha/beta
ommended 8 Gy as a safe dose limit for the optic nerves/
ratios than adjacent normal tissue, increasing fractionation for
chiasm.12 The lowest optic chiasm dose quoted for radiation-
conventional large-field radiotherapy improves the therapeutic
induced optic injury in that study was 9.7 Gy. As shown in
window most of the time. This is not the case for all tumors that
▶ Table 4.1, the LQ formula with values of 0 and 2 for α/β pre-
can be treated by radiation and may not hold for radiosurgery
dicts that a 10 Gy single-fraction dose to the optic chiasm
dose distributions. First, there is a wide variation of alpha/beta
should have the equivalent effect of 32 Gy or 50 Gy at 2 Gy per
ratios found in malignant tumors. Some malignant tumors such
fraction, respectively, for α/β = 0 or 2, respectively. Using an α/β
as melanoma or prostate cancer (or at least some strains of
value of 0, the equivalent dose at 2 Gy per fraction for the 9.7-
them) have lower alpha/beta ratios than surrounding normal
Gy radiosurgery dose that causes optic neuropathy is only
tissues (skin or rectum/bladder), supporting the use of larger
47 Gy. From clinical experience with fractionated radiotherapy
dose-fractions for treatment.10 Second, benign tumors should
of pituitary adenomas and other parasellar tumors, the risk of
have lower alpha/beta ratios than malignant tumors. Third, and
optic neuropathy for 46 to 48 Gy at 2 Gy per fraction should be
possibly most important for radiosurgery, alpha/beta ratios for
approximately 1/300.13
the tumor cells themselves do not describe the radiation res-
Stafford et al reported a later analysis of the Mayo Clinic
ponse of the supporting adventitia such as the endothelial cells
experience with four cases of optic neuropathy occurring out
within the tumor vasculature, which appear to be responsible
of 215 radiosurgery patients receiving a median optic chiasm
for most of the response with high-dose single fractions and
dose of 10 Gy.14 One case of optic injury developed after a
appear to have low alpha/beta ratios.4
dose of 12.8 Gy with radiosurgery alone, where the risk
seemed to be approximately 3%. The other cases developed
4.6 What Are the Limitations of after prior fractionated radiotherapy (7 Gy preceded by
58.8 Gy fractionated, 9 Gy after 45 Gy fractionated, and one
Using the Linear-Quadratic case that followed two radiosurgery procedures delivering 9
and later 12 Gy to the optic system after 50.4 Gy of fractio-
Formula in Radiosurgery? nated radiotherapy).
The LQ formula provides a reasonably good fit to postradiation Leber et al from Graz, Austria, analyzed optic neuropathy
single-cell culture survival data and is useful in the clinic for risks in 50 patients after 24- to 60-month follow-up (median 40

23
The Radiobiology of Radiosurgery

months) post–Gamma Knife radiosurgery for benign skull base occur. The long-term failure rate for vestibular schwannoma
tumors.15 They found optic neuropathy risks of 0% with < 10 Gy, radiosurgery is in the range of 1 to 2%. Tumors that have been
27% with 10 to 15 Gy, and 78% with > 15 Gy. No cavernous sinus previously resected have higher marginal failure rates, but
nerve injury developed with doses of 5 to 30 Gy. The optic there are more difficulties with defining the true target vol-
nerve injury risks seen in the Graz and Mayo Clinic series are ume because of difficulty distinguishing gross tumor from
what would be expected with 2-Gy fraction equivalent values postoperative effects and inability to define microscopic
calculated with alpha/beta ratios between 0 and 1 Gy/fr, rather areas of residual tumor that can lead to marginal recurrence.
than the value of 2 Gy/fr expected from a large body of data for Unresected and unbiopsied meningiomas defined by imaging
fractionated radiotherapy.6 criteria provide more clearly defined targets for defining a
dose response without a biopsy, but they also have at least a
2% chance of misdiagnosis.16 After excluding known cases of
4.8 What Are the Problems misdiagnosis in the Pittsburgh unbiopsied meningioma expe-
Assessing Radiobiological rience, it was not possible to define a significant dose res-
ponse by prescription dose. Tumor contours were lacking to
Parameters from Radiosurgery test dose-response relationships for absolute minimum dose;
any assessment would be limited by the lack of failures to
Data? analyze (because of the 93% tumor control rate). Genetic dif-
Analyzing clinical radiosurgery data to delineate dose-res- ferences in tumors and/or host adventitia (particularly endo-
ponse relationships and define radiobiological parameters is thelial cells) can lead to radiobiological heterogeneity that
full of difficulties. Radiosurgery plans typically use inhomo- flattens or blurs tumor dose responses. That could make
geneous dose distributions with doses prescribed to 50 to alpha/beta ratios more difficult or impossible to define from
80% isodose-treatment volumes covering from 90 to 100% of the data—the same way that misdiagnosis could.
the target volume. Absolute minimum doses within the tar- Malignant tumors, particularly brain metastases, have higher
get volume typically run 5 to 30% lower than the prescription failure rates, which should make dose responses easier to
dose, and they are highly dependent on how accurately tar- define. The chief problem with assessing radiobiological param-
gets are contoured. Contours of the same tumor/target vol- eters from dose responses for brain metastasis radiosurgery is
ume or critical structures can vary slightly from one clinician reliably distinguishing recurrence from radiation injury reac-
to another. With the steep dose falloff at the edge of the tions before patients die from advanced metastatic cancer. In
tumor contour in most radiosurgery plans, there can be sig- other problems such as tumor/adventitial heterogeneity, possi-
nificant variation in absolute minimum dose with subtle dif- ble slight variations in tumor definitions remain.
ferences in tumor contouring, much more than the variation
seen for average dose, percent coverage, or other parameters.
Early treatment-planning programs for Gamma Knife radio- 4.9 What about Neuropathy
surgery were not integrated with imaging and tumor-con-
touring tools. After starting with that approach, many Models for Acoustic Schwannoma
Gamma Knife centers such as Pittsburgh did not routinely Radiosurgery?
contour all target volumes until many years later. Single-frac-
tion radiosurgery dose-response data for AVM obliteration The LQ formula runs into problems in modeling dose-res-
and radiation injury to brain parenchyma and cranial nerves ponse curves for cranial neuropathies after single-fraction
can be used to calculate alpha/beta ratios; however, as shown radiosurgery of acoustic schwannomas. We should be able
in ▶ Table 4.2, they yield values of −30 to −60 Gy/fr rather to calculate values of alpha, beta, and the alpha/beta ratio
than 2 to 3 Gy/fr, as would be expected from conventional from clinical data for the single-fraction dose-response
fractionated radiotherapy data.6,9,10 curve. Data for injury to the facial, acoustic, and auditory
Attempts to calculate alpha/beta ratios for benign tumor nerves after acoustic schwannoma radiosurgery should
control are further limited by the difficulty defining any clear allow the calculation of alpha/beta ratios for radiation
dose response using the very small number of failures that injury to these nerves. We analyzed 218 acoustic schwanno-
ma patients who underwent radiosurgery at the University
of Pittsburgh from 1987 to 1997 with more than 2 years of
follow-up.17 Higher doses used in our early experience at
Table 4.2 Calculated alpha/beta ratios for different end points after ra- Pittsburgh resulted in higher rates of facial and trigeminal
diosurgery at the University of Pittsburgh neuropathy and poorer hearing preservation during that
Clinical end point n α/β ratio time than are seen with marginal doses of 12 to 13 Gy cur-
rently in use. This analysis assumed that the dose to the
Facial weakness 31/218 acoustic −60.8 ± 81.4
facial and auditory nerves matched the marginal doses pre-
schwannomas
scribed at the time of radiosurgery because these nerves
Hearing loss 57/138 acoustics with −39.6 ± 9.2 invariably lie along the capsule of the tumor. This assump-
hearing tion is not as reliable for the trigeminal nerve so that data
In-field AVM obliteration 293/355 AVMs obliterated −49.3 ± 5.3 will be excluded, although the findings were similar. We
found small, negative values for the beta coefficients for
Post-RS imaging changes 87/307 AVMs with changes −29.7 ± 2.4 facial and auditory neuropathy, with best-fitting alpha/beta

24
The Radiobiology of Radiosurgery

ratios in the range of −30 to −55 (▶ Fig. 4.2 and ▶ Table 4.2). patients and with magnetic resonance imaging (MR) alone in
Not only does this clash with the expected value of α/β = 2 75/87 (86%) AVM patients who had refused angiographic fol-
from an analysis of conventional fractionated radiotherapy low-up. The overall corrected obliteration rate (assuming 96%
data, the negative values for beta and the alpha/beta ratios, accuracy for MR) was 75%.
which mathematically describe the empirically best-fitting Because part of the AVM nidus may be unintentionally omit-
dose-response curves for this data, should be disallowed by ted from the treatment volume due to poor visualization at
the theoretical rationale for the LQ formula. radiosurgery, it makes more sense to define a radiobiological
dose-response analysis for in-field nidus obliteration (whether
or not there was obliteration within the treatment volume).
4.10 What about Radiobiological The best-fitting values of alpha/beta ratios for in-field oblitera-
Analyses of AVM Radiosurgery? tion for empirically derived logistic regression models were α/
β = −44.4 ± 12.5 and α/β = −45.3 ± 9.8, for angiographic only and
We found similar problems using the LQ formula to fit dose- for MR plus angiographic obliteration, respectively. We derived
response data for AVM radiosurgery. We analyzed oblitera- similar values with standard LQ Poisson models of α/β = −48.1 ±
tion after Gamma Knife radiosurgery in 351 AVM patients 5.9 for angiographic obliteration only and α/β = −49.3 ± 5.3 for
with 3 to 11 years of follow-up imaging after Gamma Knife MR or angiographic obliteration. ▶ Fig. 4.3 shows the dose-res-
radiosurgery at Pittsburgh.18 The median marginal-dose ponse curve for in-field MRI or angiographically defined oblit-
range was 12 to 30 Gy (median 20 Gy) to treatment volumes eration. The negative alpha/beta ratio was necessary to
that varied from 0.26 to 4.0 cc (median 5.7 cc). We docu- mathematically describe the plateau in the in-field dose-res-
mented obliteration with angiography in 193/264 (73%) AVM ponse curve (at approximately 23 Gy).

Fig. 4.2 Linear-quadratic (Poisson) dose-response


curves for the development of auditory and facial
neuropathies according to marginal (minimum
tumor) dose. These curves and their corre-
sponding alpha/beta ratio values were obtained
from nonlinear regression analysis of 218 acous-
tic schwannoma patients who underwent radio-
surgery at the University of Pittsburgh from 1987
to 1997 with more than 2 years of follow-up.

Fig. 4.3 Standard linear-quadratic (Poisson) dose-


response curve for postradiosurgery, magnetic
resonance, or angiographic arteriovenous mal-
formation obliteration of 297 patients who
underwent Gamma Knife radiosurgery at the
University of Pittsburgh from 1987 to 1997
without embolization. Each square and associ-
ated error bar represents the percent obliteration
rate with the respective 95% confidence interval
for each marginal-dose groups at the mean
marginal-dose value for each group.

25
The Radiobiology of Radiosurgery

We also derived an alternative maximum obliteration rate received by the normal tissue surrounding a radiosurgery
model for the dose response of AVM obliteration using data target volume, the same tissue that manifests that injury.
for overall obliteration rather than in-field obliteration as the Normal tissue complication probability models calculate
end point. This model added an empirically fitted maximum chances of radiation injury from dose distribution to normal
obliteration rate to the dose-response function to account for tissue outside the target/tumor. If this is true, then there
marginal misses or any unknown factor that could limit the should be no difference in complications between different
maximum obliteration rate from reaching 100%. Because it targets (AVM, benign or malignant tumor) after accounting
required only overall obliteration data, it avoided any poten- for the effects of dose and volume. To test this, we modeled
tial error in determining whether failures with persistent postradiosurgery imaging changes (symptomatic or asympto-
AVM nidus were inside or outside the original radiosurgery matic edema) that developed in 27 of 307 AVM patients and
volume. We found a maximum obliteration rate for cases compared them with those developing in 14 of 291 meningi-
treated without embolization of 87.9 ± 4.9% (p < 0.0001). The oma patients, all of whom had 2 or more years of follow-up.
additional failure rate due to embolization (i.e., probable ▶ Table 4.3 shows the results of the multivariate logistic
increased marginal miss) was 16.4 ± 7.6% (p = 0.03). This regression model comparing postradiosurgery imaging
model had less sensitivity to the alpha/beta value, so its value sequelae for AVM versus meningioma while controlling for
could not be precisely defined aside from seeing a poor fit the effects of dose (in a LQ expression with marginal dose
with any positive values. A negative value for the alpha/beta and marginal dose squared) and for treatment volume in the
ratio could not be precisely defined when negative values model. The probability of postradiosurgery imaging sequelae
were allowed. The alpha/beta ratio value converged to zero was significantly higher (p < 0.0001) for AVM patients by a
when constrained to positive values, but unlike the data for factor of 7.5 (95% confidence interval [CI]: 3.4–16.4). The
in-field obliteration, an adequate fit was achieved at that ratio of the regression coefficients for marginal dose and
value. marginal dose squared gave an alpha/beta ratio of −42.2 ±
Radiation injury reactions after AVM radiosurgery also pro- 17.2 for these postradiosurgery imaging changes for the com-
vide excellent data to test radiobiological models. New areas of bined data set.
increased signal develop on T2-weighted MR sequences in brain Reanalyzing these data using symptomatic postradiosurgery
adjacent to AVM in approximately 30% of patients within 2 injury as the end point gave similar results, despite the fact that
years after radiosurgery. These postradiosurgery imaging location was not incorporated into the analysis to keep it as
changes represent a radiation injury reaction involving sur- simple as possible. An AVM had a significantly higher (p = 0.04)
rounding brain. These signal changes are symptomatic in a third risk of symptomatic postradiosurgery sequelae by a factor of
of the patients developing this reaction (10% of all AVM radio- 2.58 (95% CI: 1.04–6.41). The approximate alpha/beta value cal-
surgery patients). The chance that these changes produce culated using symptomatic postradiosurgery sequelae as an
noticeable symptoms varies dramatically with location within end point was −42.0 ± 26.7.
the brain. Because modeling the influence of location on symp- These comparisons of postradiosurgery imaging changes and
tomatic sequelae is extremely complex,19 postradiosurgery of symptomatic sequelae in AVM versus meningioma radiosur-
imaging changes (that do not seem to vary with location) repre- gery targets demonstrate that the target tissue can have a sub-
sent a better end point for radiobiological analysis of parenchy- stantial effect on the chance of developing a radiation injury
mal brain injury. reaction and should not be ignored in radiobiological models.
We tested the fit of the LQ equation with data for postradio- The higher risk of an injury reaction in treating an AVM com-
surgery imaging changes in 307 AVM patients with more than 2 pared with meningioma suggests that the injury reaction of the
years of follow-up after radiosurgery at Pittsburgh. We identi- target vasculature may affect the surrounding normal tissue
fied postradiosurgery imaging changes in 87 of these patients; and its supporting vasculature and contribute to radiation
32 were symptomatic. We used marginal dose (minimum AVM injury reaction outside the target.
nidus dose prescribed) to represent dose. As shown in
▶ Table 4.2, the best-fitting alpha/beta value with nonlinear
regression analysis using a Poisson model (Equation 4.1) was α/
Table 4.3 Multivariate logistic regression models comparing postradio-
β = −29.7 ± 2.4. Although marginal dose is a simplified way to
surgery imaging changes (symptomatic or asymptomatic edema) in 307
represent the inhomogeneous dose distribution to the sur- AVM patients and 291 meningioma patients with 2 or more years of fol-
rounding normal tissue, we obtained a negative alpha/beta val- low-up
ue that was consistent with those for AVM obliteration, facial,
Variable p value Odds ratio 95% confidence
and auditory neuropathies. interval

AVM vs. menin- < 0.0001 7.502 3.43–16.40


4.11 Do Radiation Injury Reactions gioma

Come Only from Tissue Outside Marginal dosea 0.0143 2.457 per Gy 1.197–5.043

0.9789 per Gy2


the Target/Tumor? Marginal dose
squareda
0.0148 0.9624–0.9958

Except for functional radiosurgery, where the target is nor- Volume < 0.0001 1.142 per mL 1.217–3.431
mal tissue, it seems intuitive that radiation injury reactions
aApproximate value of α/β = −42.21 ± 17.22.
should depend only on the dose distribution of radiation

26
The Radiobiology of Radiosurgery

(such as 6–8 Gy). Guckenberger et al tested the fit of the


4.12 What Was Learned from the standard LQ model compared with the LQ model in a large
RTOG Dose-Escalation Study for multicenter lung cancer radiosurgery database and found
no significant difference in the fit of the clinical data
Radiosurgery? between the two models in assessing tumor control. 25 It is
The Radiation Therapy Oncology Group (RTOG) Phase 1 possible that any improvement in fit could have been
Radiosurgery Dose-Escalation Study (95-05) was a landmark blurred out by heterogeneity in tumor radioresponsiveness
study that provided a number of important findings. 20 One and heterogeneity due to variations in target definition
hundred fifty-six patients who failed prior conventional and treatment delivery between institutions and individual
radiotherapy for either brain metastasis or primary tumors patients.
were enrolled and treated according to a dose-escalation
protocol stratified by lesion diameter. 20 Brainstem tumors
were excluded from the study because of higher risks of 4.14 Tumor Hypoxia
symptomatic injury expected that could have confounded
Several authors have documented poorer control after radio-
the analysis. Starting with marginal doses of 18, 15, and
surgery of heterogeneously enhancing brain metastases com-
12 Gy for diameters ≤ 20, 21–30, and 31–40 mm, respec-
pared with homogeneously enhancing brain metastases.26,27
tively, the study escalated doses in 3-Gy intervals in succes-
This is thought to be from tumor hypoxia occurring in areas of
sive patient cohorts until unacceptable toxicity was
poor contrast enhancement that confers relative radioresist-
reached, defined as irreversible RTOG grade 3, 4, or 5 neu-
ance to those areas of tumor. Radiation resistance due to
rologic toxicity within 3 months of radiosurgery in > 30% of
tumor hypoxia may be addressed by fractionating radiation
patients. They defined maximum tolerated dose (MTD) as
treatments to allow for reoxygenation of hypoxic regions after
the next lowest dose level before reaching unacceptable
more fully oxygenated regions are more easily killed off by the
(> 30%) toxicity. The cohort with tumors ≤ 20 mm never
initial fractions of radiation. Other approaches to address
exceeded 30% grade 3 or greater toxicity despite dose esca-
radioresistance due to tumor hypoxia include surgical resec-
lation to 24 Gy, and investigators balked at escalating to
tion before or after radiation treatment, use of selective
higher doses. ▶ Fig. 4.4 shows the reported late neurologic
hypoxic sensitizing drugs, or chemotherapy (such as mitomy-
toxicity data at each size category fitted to logistic dose-
cin C) with greater toxicity to hypoxic cells. ▶ Fig. 4.5 shows a
response curves.
radiosurgery dose plan for 20 Gy to a brain metastasis with a
higher dose (30 Gy) specified to a central nonenhancing por-
4.13 Other Models tion of the tumor.

Several authors have suggested modifying the standard LQ


model because laboratory and clinical studies have shown
more of a linear dose response with doses above 6 Gy/fr due
4.15 Future Developments
to the quadratic component overestimating the response. 21, Different radiobiological strategies continue to be explored to
22,23,24 They have suggested using alternative models such as
better control tumors with radiosurgery with less toxicity. All
a mixed LQ model that switches from a LQ response at low- radiobiological strategies need to be evaluated through careful
dose fractions to a linear response after a break point dose laboratory and clinical trials.

Fig. 4.4 Logistic dose-response curves fit to the


late toxicity data reported for the RTOG (Radia-
tion Therapy Oncology Group) Radiosurgery
Dose-Escalation Study. The numbers refer to the
number of tumors treated at each dose level for
each size category (filled diamonds, < 20 mm;
squares, 21–30 mm; open diamonds, 31–
40 mm). The doses where 10% late toxicity occurs
in the fitted curves are shown next to the legend.
MTD, maximum tolerated dose.

27
The Radiobiology of Radiosurgery

Fig. 4.5 Radiosurgery treatment plan for a brain metastasis with 20 Gy prescribed to the tumor margin and 30 Gy to the central nonenhancing portion
of the tumor thought to have greater radioresistance from tumor hypoxia.

[14] Stafford SL, Pollock BE, Leavitt JA, et al. A study on the radiation tolerance of
References the optic nerves and chiasm after stereotactic radiosurgery. Int J Radiat Oncol
Biol Phys 2003; 55: 1177–1181
[1] Richard DJ, Bolderson E, Cubeddu L, et al. Single-stranded DNA-binding pro-
[15] Leber KA, Berglöff J, Pendl G. Dose-response tolerance of the visual pathways
tein hSSB1 is critical for genomic stability. Nature 2008; 453: 677–681
and cranial nerves of the cavernous sinus to stereotactic radiosurgery. J Neu-
[2] Ayoub N, Jeyasekharan AD, Bernal JA, Venkitaraman AR. HP1-beta mobiliza-
rosurg 1998; 88: 43–50
tion promotes chromatin changes that initiate the DNA damage response. Na-
[16] Flickinger JC, Kondziolka D, Maitz AH, Lunsford LD. Gamma knife radiosur-
ture 2008; 453: 682–686
gery of imaging-diagnosed intracranial meningioma. Int J Radiat Oncol Biol
[3] Greenberger JS, Epperly MW. Review. Antioxidant gene therapeutic ap-
Phys 2003; 56: 801–806
proaches to normal tissue radioprotection and tumor radiosensitization. In
[17] Flickinger JC, Kondziolka D, Lunsford LD. Dose and diameter relationships for
Vivo 2007 21:141–146
facial, trigeminal, and acoustic neuropathies following acoustic neuroma ra-
[4] Garcia-Barros M, Paris F, Cordon-Cardo C, et al. Tumor response to radiother-
diosurgery. Radiother Oncol 1996; 41: 215–219
apy regulated by endothelial cell apoptosis. Science 2003; 300: 1155–1159
[18] Flickinger JC, Kondziolka D, Maitz AH, Lunsford LD. An analysis of the dose-
[5] Szeifert GT, Massager N, DeVriendt D, et al. Observations of intracranial neo-
response for arteriovenous malformation radiosurgery and other factors af-
plasms treated with gamma knife radiosurgery. J Neurosurg 2002; 97
fecting obliteration. Radiother Oncol 2002; 63: 347–354
(Suppl): 623–626
[19] Flickinger JC, Kondziolka D, Lunsford LD, et al. Arteriovenous Malformation
[6] Barendsen GW. Dose fractionation, dose rate and iso-effect relationships for
Radiosurgery Study Group. Development of a model to predict permanent
normal tissue responses. Int J Radiat Oncol Biol Phys 1982; 8: 1981–1997
symptomatic postradiosurgery injury for arteriovenous malformation pa-
[7] Dale RG. The application of the linear-quadratic dose-effect equation to frac-
tients. Int J Radiat Oncol Biol Phys 2000; 46: 1143–1148
tionated and protracted radiotherapy. Br J Radiol 1985; 58: 515–528
[20] Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treatment of recur-
[8] Fowler JF. The linear-quadratic formula and progress in fractionated radio-
rent previously irradiated primary brain tumors and brain metastases: final
therapy. Br J Radiol 1989; 62: 679–694
report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys 2000; 47: 291–
[9] Hall EJ, Brenner DJ. The radiobiology of radiosurgery: rationale for different
298
treatment regimes for AVMs and malignancies. Int J Radiat Oncol Biol Phys
[21] Iwata H, Shibamoto Y, Murata R, et al. Estimation of errors associated with
1993; 25: 381–385
use of linear-quadratic formalism for evaluation of biologic equivalence be-
[10] Brenner DJ, Hall EJ. Fractionation and protraction for radiotherapy of prostate
tween single and hypofractionated radiation doses: an in vitro study. Int J Ra-
carcinoma. Int J Radiat Oncol Biol Phys 1999; 43: 1095–1101
diat Oncol Biol Phys 2009; 75: 482–488
[11] Flickinger JC, Kalend A. Use of normalized total dose to represent the biolog-
[22] Shibamoto Y, Otsuka S, Iwata H, Sugie C, Ogino H, Tomita N. Radiobiological
ical effect of fractionated radiotherapy. Radiother Oncol 1990; 17: 339–347
evaluation of the radiation dose as used in high-precision radiotherapy: effect
[12] Tishler RB, Loeffler JS, Lunsford LD, et al. Tolerance of cranial nerves of the cav-
of prolonged delivery time and applicability of the linear-quadratic model. J
ernous sinus to radiosurgery. Int J Radiat Oncol Biol Phys 1993; 27: 215–221
Radiat Res (Tokyo) 2012; 53: 1–9
[13] Flickinger JC, Rush SC. Linear accelerator radiotherapy of pituitary adenomas.
[23] Iwata H, Matsufuji N, Toshito T, Akagi T, Otsuka S, Shibamoto Y. Compatibility
In: Landolt AM, Reilly V, eds. Pituitary Adenomas—Biology, Diagnosis and
of the repairable-conditionally repairable, multi-target and linear-quadratic
Treatment. Edinburgh, UK: Churchill Livingstone; 1996:475–483

28
The Radiobiology of Radiosurgery

models in converting hypofractionated radiation doses to single doses. J Radi- dose per fraction stereotactic body radiotherapy for early stage non-small cell
at Res (Tokyo) 2013; 54: 367–373 lung cancer. Radiother Oncol 2013; 109: 13–20
[24] Otsuka S, Shibamoto Y, Iwata H, et al. Compatibility of the linear-quadratic [26] Shiau CY, Sneed PK, Shu HK, et al. Radiosurgery for brain metastases: rela-
formalism and biologically effective dose concept to high-dose-per-fraction tionship of dose and pattern of enhancement to local control. Int J Radiat On-
irradiation in a murine tumor. Int J Radiat Oncol Biol Phys 2011; 81: 1538– col Biol Phys 1997; 37: 375–383
1543 [27] Peterson AM, Meltzer CC, Evanson EJ, Flickinger JC, Kondziolka D. MR imaging
[25] Guckenberger M, Klement RJ, Allgäuer M, et al. Applicability of the linear- response of brain metastases after gamma knife stereotactic radiosurgery.
quadratic formalism for modeling local tumor control probability in high Radiology 1999; 211: 807–814

29
The Physics of Radiosurgery

5 The Physics of Radiosurgery


David Schlesinger, Brian Wang, and Stanley H. Benedict

surface area, (2) accurate and precise target localization, and (3)
Key Points the delivery of small fields to high doses.

● The three basic principles of stereotactic radiosurgery (SRS)


are (1) generation of high-dose gradients, (2) accurate and 5.3 Generation of High-Dose
precise target localization, and (3) delivery of small fields to
high doses.
Gradients
● High-dose gradients are created by spreading the energy of Stereotactic radiosurgery devices, regardless of modality, all fol-
the treatment delivery over a large surface area. low a similar guiding principle to create the steep dose gra-
● Accurate and precise target localization is achieved through dients required by radiosurgery; they each spread the total
the use of immobilization and image-guidance techniques. energy delivered to the target out over a wide surface area. In
● The physics of small-field delivery poses particular challenges the case of Gamma Knife radiosurgery, this is achieved through
for SRS and stereotactic radiotherapy (SRT) and requires the use of many (either 201 or 192) widely distributed beams
great care when measuring and modeling. all collimated so they intersect at a focal spot.4 CyberKnife
● The specialized nature of SRS/SRT requires equally specialized radiosurgery also makes use of large numbers of widely distrib-
quality assurance and credentialing procedures. uted beams that intersect at the targeted tissue (although not
always a distinct focal spot).5 Linear accelerator (linac)-based
radiosurgery makes use of either a relatively large number of
non-coplanar modulated fields or a series of non-coplanar arcs
5.1 Introduction that intersect isocentrically at a point within the targeted tis-
sue.6,7 The fields themselves may be shaped using small circular
The technical aspects of stereotactic radiosurgery (SRS) have
cones, or micro-multileaf collimators (MLCs)8,9—the latter of
profound differences as compared with those used for tradi-
which may be static for each field, may modulate during deliv-
tional intracranial radiotherapy. Traditional intracranial radia-
ery of each field, or in the case of arcs may modulate over the
tion involves the irradiation of large volumes of tissue,
course of the arc (volume-modulated arc therapy [VMAT]).10
including both normal and abnormal tissue. A large number
Because the incoming photons are widely distributed as they
(often 20–30) of small doses (~ 2–3 Gy) are given, with a rest
enter the patient, the intensity of each individual radiation
period (usually 1–3 days) in between each treatment fraction.
beam is relatively low and causes minimal damage to normal
The treatment relies on the differential ability of normal and
tissue distant from the focus point. However, the sum total
pathological tissue to repair DNA damage induced by the radia-
intensity of all of the beams at the target is quite high. The large
tion.1 In contrast, SRS relies less on differential repair and more
spatial distribution achieves a very steep gradient away from
on differential targeting for its treatment effect. A large dose of
the target.
radiation, delivered in one to at most five fractions, is directed
towards ideally only the targeted pathological tissue. The sur-
rounding normal tissue would receive no dose, but in actuality
SRS treatments do not quite reach this idealized goal. The
5.4 Precise and Accurate
resulting realized tumoricidal efficacy is likely due to both DNA Localization
damage as well as biological effects that occur only after a
threshold dose has been absorbed.2,3 The ability to aim large numbers of beams or arcs at a small
The differences in requirements between traditional radia- point in three-dimensional (3D) space is not in itself sufficient
tion oncology and SRS and stereotactic radiotherapy (SRT) led for radiosurgery. To be useful, that small point in space has to
to the development of specialized equipment and procedures coincide with the targeted tissue as accurately and precisely as
for the latter. In this chapter, we discuss the technical underpin- possible throughout the course of treatment. In SRS, this is
nings of SRS/SRT, including physics, immobilization techniques, achieved through a combination of modern 3D-imaging techni-
treatment planning, image guidance, quality assurance, and ques to permit visualization of the target and surrounding tis-
credentialing. We place special emphasis on the features that sue, methods to define the 3D spatial relationships between the
develop as a response to the specific technical demands of ster- targeted tissue and the treatment device, and methods for
eotactic radiosurgery. immobilization to prevent the targeted tissue from moving out
of the treatment beams during the procedure.

5.2 Basic Physical Principles of 5.5 Small Fields, High Doses


Stereotactic Radiosurgery The prior principles—spreading out the energy, and precise and
Stereotactic radiosurgery relies on three basic principles to accurate localization—then permit the final principle: delivering
achieve its goal of differential targeting: (1) generation of high- high doses of radiation to small volumes of tissue. Radiosurgical
dose gradients by spreading out the radiation dose over a large targets are generally quite small in the broad scope of radiation

30
The Physics of Radiosurgery

oncology—hard limits vary, but a good rule of thumb is that tar- systems, the back of the patient’s head is supported by an insert
gets should be less than 30 mm in largest diameter. Targets larg- and a plastic mask is stretched over the patient’s face. The plas-
er than this treated to high doses increase the risk of adverse tic of the mask is flexible when heated, but becomes rigid when
treatment outcomes. Doses are also quite large, typically in the cooled. Mask systems have the advantage that they can be
range of 10 to 150 Gy point-dose maximum within the target— easily removed and reapplied to a patient, making multiple
with the largest doses of 80 to 150 Gy usually reserved for func- fraction treatments practical. The disadvantage of mask sys-
tional lesioning cases.11,12 tems is that they are not as robust in terms of immobilization,
with patient motion routinely greater than 1 mm reported
within the mask.19 Because of their immobilization uncertainty,
5.6 Technical Challenges for mask systems are typically used only for SRT and traditional
Stereotactic Radiosurgery hyperfractionated treatment schemes.

Developing technical solutions that can achieve the above goals


is no small task, and much of the development of SRS/SRT has 5.7.3 Relocatable Frames
been in refinements to help reach them.13 Accurately placing Relocatable frame systems have been developed that attempt to
the targeted tissue at the intersection of the beams creates chal- improve on the immobilization capability of mask systems yet
lenges for imaging, localization, and immobilization. Accurately maintain the ability to reputably remove and replace the frame.
computing dose to small fields, which depart from well-investi- The TALON frame20 is a hybrid system that involves implanting
gated reference standards, creates challenges for radiation two titanium base screws into the patient’s skull. An adjustable
metrology and dosimetric modeling. In the remainder of this Nomogrip/TALON device is attached to the base screws and
chapter, we describe how techniques adopted for radiosurgery locked into a patient-specific position. The patient can then be
address these issues. removed and reattached to the TALON device.
Several systems have been developed that use dental fixation
to assist in immobilization of the head. In these systems, a den-
5.7 Immobilization Techniques tal mold of the patient’s upper palate is created. The dental
applicator is then attached to a frame system.21 The patient
5.7.1 Frames position within the frame system is determined either through
Radiosurgery evolved directly from the practice of stereotactic measurement22 or on-board imaging.23 In some systems, the
neurosurgery, which involved the placement of an invasive intratreatment position of the patient can be monitored using
frame system over a patient’s head both to immobilize the pa- optical tracking24 or a vacuum monitoring system.25 The uncer-
tient and to create a coordinate system, allowing neurosurgeons tainties of using these systems have been shown to approach
to consistently target any desired point in the brain. Radiosur- uncertainties reported with frame systems.
gery replaced the neurosurgery probe with a focal point of con-
verging beams, and therefore had similar requirements for
immobilization and localization.
5.7.4 Fully Frameless
Several frames have been and continue to be in use in radio- The development of on-board 3D-imaging, 6-degree-of-free-
surgery, including the Leksell G-Frame,14 Gill-Thomas-Cosman dom robotic couches, and intradelivery imaging and position-
(GTC) frame,15 and the BRW frame.15,16 Although the details monitoring systems have made feasible the idea of frameless
vary slightly depending on the design, the basic principles of radiosurgery: In these systems, no frame is required.
each frame are similar. The frame defines a targeting coordinate
system that encompasses the volume of the target and sur-
rounding tissue (usually the entire head). The patient is imaged 5.8 Problems Posed by Small and
with the frame and associated external fiducials, which create a
coordinate transformation between the native image coordi-
Nonstandard Treatment Fields
nate system and the frame coordinate system. The frame is rig- Medical physicists have historically defined a “small field” as
idly mounted to the treatment table, which creates a fixed any field smaller than conventional field sizes: usually smaller
transformation between the frame coordinates and the delivery than 3 cm × 3 cm.26 A slightly less subjective definition is a field
machine coordinates. The advantages of stereotactic frames are size “smaller than the lateral range of charged particles that
their mechanical stability, accuracy,17 and their relative simplic- deposit dose at a point along the central axis.”27,28 Small fields
ity. The disadvantages of stereotactic frames are that they are or very small fields can also be defined in terms change in out-
mildly invasive, practically limit radiosurgery to a single-frac- put factor (OF) as a function of change in field size or detector
tion treatment, and create a procedural time constraint in that position (e.g., a very small field is any field whose OF changes
treatment planning, imaging, and delivery must all occur on the by more than 1.0% given a 1.0-mm change in field size or detec-
same day, as adjusting the frame after imaging for treatment tor position).28 Stereotactic radiosurgery tends to fall within
planning would alter the targeting coordinate system. any of the clinically relevant definitions of small field size.
In addition, many specialized radiosurgery devices have geo-
metries that are incompatible with standardized calibration
5.7.2 Masks protocols such as TG-51 and IAEA TRS-398, which are based on
To overcome some of the disadvantages of invasive stereotactic broad (10 × 10 cm) reference fields and are specified to use ion
frames, thermoplastic mask systems were developed.18 In these chambers to measure dose using water as a medium to achieve

31
The Physics of Radiosurgery

33 The emergence of high-dose-rate flattening-filter-free (FFF)


a direct conversion from collected charge to absorbed dose in
water.27 For example, neither a Gamma Knife nor CyberKnife modes on recent SRS/SRT-capable linear accelerators may cre-
has the ability to create a 10 cm × 10 cm reference field. It is also ate another departure from standardized protocols.34
not practical to create a water-filled phantom for a hemispheri-
cal Gamma Knife geometry, although attempts at this have been
made.29 For the reasons outlined above, for these devices an ion 5.10 Detectors for Stereotactic
chamber may not be the appropriate choice for conducting
measurements.
Radiosurgery and Stereotactic
Clinical dosimetric measurements required for commission- Radiotherapy
ing an SRS machine, modeling SRS performance, or providing
quality assurance must take into account the effects small fields A large variety of detectors are now available to the clinical
can have on different measurement devices and techniques. physicist, including “Farmer”- type ion chambers, microioniza-
Serious consequences garnering attention from prominent tion chambers, solid-state detectors, radiochromic film, gel dos-
news organizations have been reported in cases where this has imeters, diamond detectors, etc. It is essential for SRS/SRT that
not been the case.30 an appropriate choice of detector be made depending on the
dosimetric task at hand (e.g., absolute dose output measure-
ments or relative dose profile measurements).
5.9 Effects of Small and A recent addendum to the American Association of Physicists
in Medicine (AAPM) TG51 protocol34 maintains the recommen-
Nonstandard Fields dation that cylindrical ionization chambers be used for refer-
When radiation fields are collimated to small sizes approaching ence dosimetry for treatment machines that can conform to the
the size of the detector, a variety of assumptions used in relat- field-size requirements and water-phantom requirements of
ing observed readings to dose start to break down. the protocol. Very small (volume < 0.05 cm3) chambers are not
Below a threshold field size, the radiation source as viewed recommended due to perturbations from high-Z electrodes, sig-
from the perspective of the detector is partially obscured by the nificant polarity effects, and recombination behavior that can
collimation. This has the effect of blurring and widening the pe- be difficult to model.
numbra of the beam and lowering the output at the position of For devices that do not conform to standard protocols, work
the detector. Measurements not taking this into account could is ongoing within both the AAPM and International Atomic
overestimate the field size and underestimate the output dose Energy Agency (IAEA) to create a complementary protocol that
rate.26 can encompass these machines while maintaining traceability
When field sizes are smaller than the range of charge par- to reference standards. A recent summary of the proposed
ticles liberated in the medium, the assumption of charged par- approach defines a system of intermediate reference fields,
ticle equilibrium (CPE) that underpin measurements with including a machine-specific reference (msr) field for machines
detectors such as ionization chambers start to break down. The that can produce a static field, but not of standard reference
average energy in the spectrum of electrons reaching the cen- dimensions, and a plan-class-specific reference (pcsr) field for
tral axis of the beam rises as compared with the spectrum with machines that are composed of many small fields (e.g., Gamma
CPE in place.31 In addition, the loss of CPE makes the presence Knife, CyberKnife, tomotherapy, step-and-shoot intensity-
of the detector itself a significant source of measurement uncer- modulated radiotherapy [IMRT]). Correction factors will be
tainty as it perturbs the particle fluence in the medium. The used to correct for differences between these intermediate
conversion from collected charge to absorbed dose in the me- reference fields and standard reference fields.27
dium relies on standardized protocols that are a function of Beam profiles in the setting of SRS/SRT are best measured
geometric setup, and beam quality. The lack of CPE increases using high-resolution detectors such as microionization cham-
the uncertainty of these protocols. Lack of consideration for bers, stereotactic diode detectors, diamond detectors, or radio-
small-field measurements can affect absolute output measure- chromic film. Errors due to volume-averaging effects can be
ments, as well as dose ratios such as OFs, percent depth dose/ minimized using deconvolution35,36,37 or extrapolation35,38
tissue maximum ratios (PDDs/TMRs), and off-axis ratios techniques, which can separate out the detector response from
(OARs).26 the underlying signal.
Finally, the absorbed dose gradient near a field edge is not
correctly measured by finite-sized detectors such as ion cham-
bers due to volume averaging of signal over the volume of the 5.11 Physics Considerations for
chamber. The small sizes and blurred penumbras in SRS fields
enhance this effect, and can lead to increased uncertainty when
Proton SRS/SRT
measuring beam profiles. The physics of proton interaction in tissue make them a theo-
The major problem for machine designs that are incompati- retically attractive alternative to photons in an SRS/SRT setting.
ble with standard reference dosimetry protocols are that they Protons have a predictable, finite depth of penetration that
break the traceability those protocols maintain with national depends on the energy of the protons and the density of the
and international standards laboratories.27 Machines that can- material. Protons deposit minimal energy in tissue near the sur-
not provide standard reference (i.e., 10 cm × 10 cm) fields or face and for most of the proton’s path. However, near the end of
cannot be measured in water are left to cobble together modifi- the path there is a steep rise in ionization density, creating
cations to the standards that often vary from center to center. 32, what is known as a Bragg Peak. Beyond the peak, dose is

32
The Physics of Radiosurgery

negligible. To achieve complete coverage of the tumor in the 5.12.1 Isocentric versus Nonisocentric
longitudinal direction, the energy of the proton beam is modu-
lated, either with compensators or by summing pencil beams of The isocenter is a virtual point in space where two axes cross
varying energy to “spread out” the Bragg Peak to cover the tar- and around which the treatment device and couch rotate.
get. For typical radiosurgery geometry of many converging When treating a spherical-shaped target, one single isocenter is
beams, the use of protons would mean the beams could be generally placed at the center of the target. Gamma Knife and
delivered to the target, where they would stop. The exit dose other treatment machines with conical collimators use this so-
from each beam would be negligible, greatly reducing the inte- called isocentric planning technique. For an irregular-shaped
gral dose to normal tissue.39 target, Gamma Knife planning places several isocenters inside
However, in practice, achieving the benefits of protons can be the target with associated different collimator sizes to generate
challenging. Tissue inhomogeneity can create significant range a conformal plan (▶ Fig. 5.1). This is referred as ball packing.46
uncertainty for protons. Any deviation in the patient’s position For an irregular-shaped target, CyberKnife planning aims some
relative to the simulation position accentuates this range uncer- beams at the peripheral region of the target, not passing the
tainty. Range uncertainty limits the use of beam arrangements isocenter to patch dose to this area. Such planning is called an
where the distal edge of a beam abuts a critical structure nonisocentric technique (▶ Fig. 5.2).
(which is often the case in SRS/SRT scenarios). Passive-scattered
proton systems have poor beam-shaping capabilities relative to 5.12.2 Forward versus Inverse
modern X-ray systems and can lead to higher integral doses.40
In the early days of radiosurgery, the planning computer was
Scanned protons perform better, but are more sensitive to
not powerful enough to perform optimization with multiple
imaging and motion uncertainties. Many currently active pro-
iterations and forward planning was predominantly utilized for
ton centers lack the kind of on-board 3D imaging that is avail-
intracranial radiosurgery. In contemporary practice, inverse
able on X-ray accelerators, which contributes to these
planning techniques are increasingly used with the develop-
uncertainties.41
ment of IMRT. In forward planning, treatment planners choose
various parameters such as collimator size, number and loca-
tion of isocenter, beam angle, arc angle, couch angle, and beam
5.12 Treatment Planning weights. This technique relies on the experience of the planner,
Treatment planning for intracranial radiosurgery has its unique and the developed plan may not be optimal. In inverse plan-
features and challenges compared with conventional fractio- ning, dosimetric goals and constraints are first set by planners
nated radiotherapy and treatment to other body sites. Radiation for both the treatment target and normal structures. A com-
dose is very high, and critical structures are usually located puter program then optimizes all the variable parameters to
near the target. Therefore, a steep dose falloff from the target is meet these goals and constraints. For specialized CyberKnife,47
required and planning target volume (PTV) margins are small. Axesse, Novalis, Edge, and tomotherapy48 devices, the number
Despite these demanding challenges, several unique features of beam parameter combinations is enormous and most cases
can make the treatment planning for intracranial radiosurgery can only be planned by an inverse technique. Although inverse
easier because of the location and tissue composition of the cra- planning has become an important technique for intracranial
nium. First, many noncoplanar beams can be used to spread an radiosurgery, a forward technique still remains a fast and effi-
intermediate or low-dose bath to a larger region to avoid an cient way for many cases.
ablative dose to normal structures. Specifically, vertex beams
are often used to smear the dose in a cranial–caudal direction
while maintaining the required target dose. This cannot be
5.13 Beam Shaping and Delivery
easily achieved at other body sites because of potential collision The Gamma Knife was developed more than half a century ago,
concerns between the treatment machine and the patient or and it is still a popular way to deliver radiosurgery treat-
treatment couch. Second, treatment targets are usually located ments.49,50 The Gamma Knife uses many intersecting circular
away from inhomogeneity interfaces of tissue and bone or air. beamlets from different directions to create a spherical-shaped
Dose distributions can be calculated accurately by simple fast high-dose region around the treatment target. It is not surpris-
algorithms.4 In contrast, targets in lungs demand a more ing that the initial application of a linac for radiosurgery has
sophisticated, time-consuming Monte Carlo dose-calculation used a conical collimator to shape the radiation beam by mim-
algorithm to accurately represent dose distribution. Treatment- icking the Gamma Knife’s circular beamlets.51,52 Plans are gen-
planning algorithms have evolved tremendously in the past erated by a forward technique, often with templates of
decades thanks to the advances of computational hardware. predefined couch and beam angles. The next development,
Current calculation algorithms generate more accurate dose spearheaded by Brainlab (Munich, Germany) and Varian (Palo
distributions that have been verified by measurements. 42,43 The Alto, CA), utilizes a MLC for a conformal arc delivery technique.
largest inaccuracy from previous simple algorithms occurs at In this approach, an MLC is shaped continuously to match the
inhomogeneity interfaces such as the target in lungs. When beam-eye-view shape of the target at all delivery arc angles.53,54
comparing with the treatment outcome of historical data, the One advantage of the conformal arc technique over a cone-
plan needs to be recalculated with a current algorithm to accu- based technique is its ability to deliver more efficiently to a
rately represent the dose distribution. 44,45 Treatment planning nonspherical-shaped target. A conformal arc is still a forward-
for intracranial radiosurgery suffers less from this issue because planning process that requires a manual selection of arcs and
of its relative homogenous tissue composition. 4 adjustment of the weighing. In challenging cases where the

33
The Physics of Radiosurgery

Fig. 5.1 Gamma Knife plan of a vestibular schwannoma using multiple isocenters to achieve conformality.

Fig. 5.2 Cyber Knife plan of four targets with 74


nodes and 107 beams, MU 12564.

dose needs to be shaped to avoid nearby critical structures, an combination of inverse IMRT and arc delivery techniques. Simi-
IMRT technique is often utilized to inversely optimize beams to lar to a conformal arc approach, VMAT spread a small dose to a
generate a high dose at the target while keeping the dose to larger region by delivering the dose in several arcs. Unlike a
critical structures within tolerance. An IMRT technique uses conformal arc, VMAT can inversely optimize the plan to avoid
many fixed beams to avoid irradiating ablative dose from critical structures by modulating MLC shape, gantry rotation
any single gantry angle. The next evolution of VMAT was a speed, and dose rate. Another benefit of VMAT is its ability to

34
The Physics of Radiosurgery

Fig. 5.3 Eclipse plan of single isocenter with four arcs for two targets, MU 5029.

treat multiple targets with one isocenter and multiple non- frame-based method where a localization frame is screwed into
coplanar arcs, providing a unique efficient treatment-delivery the patient’s skull. Even though the frameless approach is more
approach (▶ Fig. 5.3).10,55 convenient and comfortable, a patient could move up to 2 mm
inside an immobilization mask during treatment, causing addi-
tional uncertainty.57 Third, a nontrivial discrepancy remains
5.14 Margins and Issues with between the two isocenters of the treatment-delivery and the
Margin Assumption image-guidance systems, which is on the order of a submillime-
ter.58,59 Fourth, setup localization images are registered to the
A PTV margin is typically added from clinical target volume treatment-planning image set before treatment delivery to an
(CTV) for most radiotherapy to account for setup uncertainties. individual patient, and any registration mismatch is not
A PTV margin is typically assumed to be zero for intracranial accounted for. Last but not the least, possible tumor cells may
radiosurgery plans.56 This is probably because historically a lie outside of the lesion as defined by imaging contrast. This
frame is always used to invasively attach the skull to the treat- uncertainty is arguably the margin from gross tumor volume
ment couch for all intracranial radiosurgery treatments. Unlike (GTV) to CTV,56,60 and some physicians account this indirectly
other body parts, the brain is not subject to internal motion; by placing the prescription isodose line several millimeters
thus, a lesion stays fixed in space during treatment. However, beyond the visual tumor. All these assumptions should be char-
several issues need to be considered for potential violation of acterized to start an intracranial radiosurgery program and an
this zero PTV margin assumption. First, errors from multimo- end-to-end test can evaluate the overall geometric uncertain-
dality image registration can propagate to target contour uncer- ties by simulating the whole process with a phantom.
tainty. Most intracranial targets are delineated on functional
image datasets such as magnetic resonance imaging (MRI) and
then registered to a planning computed tomography (CT) data- 5.14.1 Dose Inhomogeneity and
set. Any mismatch between the two registered image sets
would cause target contour uncertainty. Second, intrafractional
Conformity
motion during radiation treatment delivery causes target Dose inhomogeneity and conformity are two competing met-
uncertainty. Frameless immobilization has become more com- rics for intracranial radiosurgery planning, similar to other
mon for intracranial radiosurgery thanks to recent advances in radiotherapy planning. Inhomogeneity is evaluated as the
image-guidance technology. Such a frameless approach utilizes percentage of maximum dose to the prescription dose. The
less invasive masks and offers an alternative to the traditional location of the maximum dose is called a hot spot and should

35
The Physics of Radiosurgery

occur inside the treatment target. Conformity measures how resonance (MR) spectroscopic imaging,65,66 and single-photon
well the prescription isodose surface matches the target out- emission computed tomography (SPECT).67 The geometric
line. A common conformity metric is defined as the product uncertainty should be quantified and corrected for MRI distor-
of two ratios. One measures the amount of prescription iso- tion. Inherent system distortion is caused by inhomogeneities
dose spillover into normal tissue—defined as the ratio of tar- in the main magnetic field and nonlinearity of gradient fields.
get volume encompassed by the prescription isodose volume Such system distortion increases with distance from the mag-
to the prescription isodose volume. The other measures how net center; it can be corrected by the use of phantoms. 68,69
much target is not covered by the prescription isodose— However, patient-related distortions cannot be easily corrected,
defined as the ratio of target volume encompassed by the which is mainly a concern for a high magnetic field.
prescription isodose volume to the target volume. This con-
formity metric ranges from 0 to 1, with 1 being the best
case.61 As shown in ▶ Fig. 5.4, an additional 5-mm cone was 5.15 Image Guidance
used to avoid a cold spot at the corner of this irregular-
Traditional intracranial radiosurgery procedures utilized
shaped target. Because the isodose clouds overlap from the
invasive frames together with localization boxes to immobi-
two cones, this plan will generate a higher hot spot than a
lize patients and to align the target for treatment delivery.
single isocenter plan. In this case, conformity is achieved at
The advancements in image guidance have been one of the
the sacrifice of inhomogeneity. Dose inhomogeneity and con-
most valuable contributions to radiosurgery. Image guidance
formity varies greatly with the delivery approach and are a
has facilitated the application of noninvasive frameless
function of the prescription isodose surface. For example,
immobilization, thus enabling multiple fraction treatments
Hazard and colleagues proposed a uniform method to choose
and the flexibility of separate days for simulation and treat-
a prescription isodose surface to balance target coverage
ment delivery. In addition to these benefits of patient con-
and conformity for the dynamic conformal arc delivery
venience and a less invasive procedure, image guidance has
approach.62
provided direct visualization of the treatment target or skull
surrogate before or during the ablative high-dose delivery. Of
5.14.2 Multimodality Imaging course, all of these advantages come with an additional imag-
ing dose to patients for some techniques and associated qual-
Registration ity assurance procedures required to verify the coincidence
A primary CT image dataset is often required for dose calcula- of the imaging and treatment isocenters. Common image-
tions and image guidance, but intracranial radiosurgery targets guidance techniques include stereoscopic two-dimensional
often cannot be easily visualized on the CT dataset. Multimo- (2D) X-ray, volumetric 3D X-ray, infrared or optical guidance,
dality imaging registration is required to map the target delin- and MR guidance. They can be used either for setup guidance
eation onto the primary CT dataset. Such modalities include before treatment delivery or for intrafractional guidance dur-
MRI,63 positron emission tomography (PET),64 proton magnetic ing treatment.

Fig. 5.4 Eclipse plan of two cones and four arcs


each, MU 4644.

36
The Physics of Radiosurgery

5.15.1 Stereoscopic 2D X-Ray Imaging field. In this case, a robotic couch is necessary to correct set-
up rotational errors. When the required rotation is outside
In this technique, a set of digital reconstructed radiographs the limits of the robotic couch, the immobilization and setup
(DRRs) are first generated from a planning CT dataset. Two pla- image-guidance process should be repeated. During treat-
nar X-ray images are then acquired at an oblique or orthogonal ment delivery, intrafractional guidance should monitor both
angle with patients at the treatment position. Then, the X-ray the rotational and shift changes.
images are registered to the DRRs to calculate image-guidance
shifts. This technique includes room-mounted and gantry-
mounted imaging systems. In the room-mounted setup, kilo- 5.15.3 Infrared and Optical Guidance
voltage (kV) X-ray tubes and digital detecting panels are X-ray image-guidance techniques expose patients with addi-
installed oppositely at the ceiling and on the floor with image tional imaging dose that is not trivial, especially when used
beams passing through targets obliquely. Typical commercial for intrafractional guidance. Infrared 71 and optical72,73 guid-
systems with this technique are Novalis54 and CyberKnife.47 For ance are two popular techniques to solve this issue. Infrared
gantry-mounted systems, a kV X-ray tube and detecting panel technique uses ceiling-mounted cameras to detect either
are installed orthogonally to the treatment beam on the gantry. reflection of passive markers or active light-emitting
Two kV images can be acquired orthogonally at two gantry markers. The markers are attached to a bite block that is
angles for image guidance. An alternative technique is to formed to the patient’s dentition. Infrared guidance techni-
acquire a pairing of a kV image and a megavoltage (MV) image ques can be used for either initial setup prior to treatment
from the portal imager without the need of a rotating gantry. delivery or intrafractional guidance. The major limitation is
Gantry-mounted systems are available for most modern linac the potential movement of markers relative to the patient
machines. skull. In the past few years, optical guidance techniques have
The room-mounted setup separates the hardware compo- emerged for intracranial radiosurgery applications. Several
nents of the image-guidance system from the treatment-deliv- groups have investigated an open mask immobilization with
ery system. The associated advantages include a more-stable a commercial AlignRT system.72,73 In this optical guidance
imaging isocenter that is not subject to the changes of the gan- technique, a reference image is first generated from the skin
try position, and shorter imaging time compared with gantry- rendering of the planning CT dataset. Three ceiling-mounted
mounted systems. Room-mounted systems can also be used for camera pods capture facial landmarks of the patient and then
intrafractional guidance during treatment delivery in addition compare them with the reference image. One major advant-
to setup guidance prior to treatment. On the other hand, the age of this optical guidance technique is the ability of moni-
gantry-mounted system can image the target at any angle with- toring any movement during treatment delivery without
out the concerns that gantry blocks images at certain angles. additional imaging dose. In addition, patients are more com-
fortable and compliant under the less restrictive immobiliza-
tion masks. The optical guidance technique is typically used
5.15.2 Volumetric 3D X-Ray Imaging as an intrafractional guidance method after setup guidance
Another popular method is to use image guidance with volu- by an X-ray–based technique. Some issues may cause local-
metric 3D cone-beam CT (CBCT), which is reconstructed from a ization errors using the optical technique, for example, its
number of projections acquired with the gantry-mounted kV dependence on room lighting and region-of-interest (ROI)
imager. The CBCT image set is registered to the planning CT selection.73 In addition, skin deformations can occur when
dataset to derive the shifting and rotational corrections.70 Com- patients change weight or take medication, causing a shift
pared with the 2D technique, volumetric 3D imaging can dis- between the positions of treatment target and monitored
play the anatomy at all three view planes: axial, sagittal, and facial surface.
coronal. This feature is especially attractive when treating mul-
tiple targets with a single isocenter setup. One limitation is the
use of CBCT for setup guidance for non-coplanar beams because
5.15.4 MR-Based Guidance
of potential collision between the couch and gantry. Another Magnetic resonance-based image-guidance systems are recent
major limitation of volumetric 3D technique is its inability for exciting developments that could introduce paradigm change
intrafractional guidance because the whole process of image for intracranial radiosurgery. Two types are explored for an
acquisition and reconstruction takes approximately 1 minute, integrated MRI-linac system74,75 and MRI with radioactive sour-
which is too long for real-time guidance. ces such as the ViewRay system. The advantages of MR image
There is an increasing interest to plan and treat multiple guidance include nonradiation imaging dose, fast 3D volumetric
intracranial targets with one single isocenter and several data, and high soft-tissue contrast. They can provide both pre-
non-coplanar arcs55 due to the high efficiency of treatment treatment and intrafractional guidance with a 3D dataset that
delivery. The shorter treatment time is advantageous for pa- can be viewed in many planes. The MR-based image guidance
tient comfort and associated less variation of target position. can provide direct target visualization, which is a major advant-
However, uncorrected rotational discrepancy between treat- age over other image-guidance techniques that relies on skull,
ment and planning positions could introduce nontrivial dosi- skin, or markers as a surrogate. For intracranial radiosurgery
metric errors. For example, a rotation discrepancy of 1 applications, MR images are already used in most cases for tar-
degree causes a displacement of 1.7 mm at a distance of get delineation during treatment-planning process; therefore,
10 cm from the isocenter. Such displacement could shift an MR-based technique provides unique same image modality
treatment target partially or even totally outside of treatment guidance.

37
The Physics of Radiosurgery

given the overall uncertainties of various aspects of the com-


5.16 Quality Assurance and Safety plete treatment.76
The safety of the patient and the treatment team are critical
concerns in any radiotherapy procedure, but perhaps especially
so with SRS/SRT. Doses are high and the number of fractions is 5.16.2 Winston-Lutz and Hidden
small, meaning there is little or no room to correct for mistakes Target/End-to-End Tests
in treatment delivery as can be done in conventional radiother-
apy. Modern treatment devices increasingly rely on complex The recognition early on that traditional quality assurance pro-
simulation imaging, on-board imaging, and intradelivery treat- cedures for linear accelerators were inadequate in the setting of
ment-monitoring techniques to ensure correct beam delivery. SRS/SRBT led to the development of specialized procedures to
Certain devices used for SRS (the Gamma Knife being a promi- ensure accuracy of the procedure. Perhaps the most prominent
nent example) rely on radioactive material (RAM) as an energy of these tests is the Winston-Lutz test, which assesses the coin-
source, and thus special care and custodianship of the RAM. A cidence of the mechanical and radiation isocenters of the linear
robust quality assurance, risk management, and training regime accelerator.51 The classic Winston-Lutz test involves placing a
are essential to the safe and effective operation of a radiosur- small spherical target at the presumed isocenter of the treat-
gery program. ment machine (usually using the room lasers). A suitable colli-
mator is set on the machine (often a circular collimator in the
case of SRS) and a radiographic film is placed perpendicular to
5.16.1 Quality Assurance Needs for SRS/ the collimator downstream of the target sphere. The film is
exposed, resulting in a radiation spot with the superimposed
SRT shadow of the target sphere. The offset between the center of
A large selection of published studies and reports are available the target shadow and the center of the field can be used to
from national and international organizations, which outline determine the difference between mechanical and radiation
best practices for commissioning and quality assurance of gen- isocenter. The test is repeated at various gantry, table, and colli-
eral aspects of beam delivery devices, in-room imaging equip- mator angles to evaluate the stability of isocenter coincidence.83
ment,76 MLCs,77 treatment-planning systems,78 and secondary Over time, the Winston-Lutz test has been modified as tech-
dosimetry check software.79 The authors refer the reader to nology has evolved. Methods have been developed to make use
these reports for guidance on these aspects of SRS/SRT treat- of electronic portal imaging devices (EPIDS) to replace film as a
ment delivery. The scope of a quality assurance program is also detector. On-board CBCT systems are used to align to the test
beginning to move beyond standardized, one-size-fits-all pro- target to test the coincidence of the on-board imaging system
cedures and instead is based on a formal, risk-based analysis with the mechanical and radiation isocenters. Targets have
of the relative risks involved in a procedure using techniques evolved as well, with purpose-built Winston-Lutz phantoms
such as failure mode and effect analysis (FMEA) and fault-tree available that are more intuitive to setup than the original
analysis.80 spherical target.83
The high doses delivered with SRS/SRT combined with the A related, but critical quality assurance technique is the end-
sharp dose gradients create a requirement for extremely to-end test (aka the hidden target test) that attempts to evalu-
aggressive tolerances for targeting accuracy. Even small devia- ate the complete treatment uncertainty from simulation
tions from the intended target can cause significant changes in through delivery, albeit on a phantom. Although the details
tumor control probability. For example, Treuer et al studies a can take many forms, the test involves a phantom with an
cohort of 20 patients with an arteriovenous malformation and embedded target, as well as an embedded detector (usually
20 patients with brain metastasis, looking at the effect of target radiochromic film or radiosensitive gel). In some cases, the tar-
point deviations on predicted obliteration/tumor control and get and detector are designed as separate swappable inserts in
normal tissue complication probability. They found that a devi- the phantom. The phantom, with target embedded, is imaged
ation of 1.3 mm decreases rates of obliteration/local control by using regular clinical imaging protocols, and a treatment plan is
approximately 5% and could indicate a need for expanded treat- developed to treat the target to a specified dose. The phantom
ment margins.81 is then setup on the treatment machine and the treatment is
However, as discussed earlier, PTV margins are often not delivered. The resulting dose distribution as captured on film or
applied in the setting of SRS/SRT.56 Also, the assumptions of a gel is then compared with the original plan using either abso-
small number of beams and a large number of fractions that lute dose differences or a combined dose/distance metric such
underlie common margin formulas used in traditional radio- as gamma analysis.84,85
therapy are violated in the setting of SRS/SRT.82 Margins inher- Certain specialized SRS/SRT devices have associated special-
ently increase the volume of tissue irradiated in a treatment, ized quality assurance tests. For example, the most recent ver-
and this extra volume may by itself contribute to an increase in sion of the Gamma Knife uses a traceable set of specialized
complication rates. diode detectors to create calibration offsets for the treatment
The limited ability to compensate for beam delivery uncer- table. A set of master diode detectors is calibrated against a pre-
tainty by expanding treatment volumes reinforces the goal of cisely measured “reference” Gamma Knife. These master tools
minimizing beam delivery uncertainty as much as possible. The are then used to create calibration offsets for site-specific diode
AAPM TG-142 suggests some action levels for various parts of detectors that are used at clinical sites.50 Another example is
the radiotherapy treatment process based on subjective obser- the CyberKnife, which uses an “isocrystal” to define the
vation of the authors regarding what is likely to be achievable mechanical room coordinate system. Images of the isocrystal

38
The Physics of Radiosurgery

on the in-room X-ray system are used to determine the X-ray Training should include all personnel involved in the treatment
source/detector alignments.86 team so that the entire scope of the procedure is covered.
Ongoing training should include a review of clinical operational
procedures, a review of recent reports of misadministrations
5.17 Clinical Implementation of an and medical events, reviews of any vendor-reported safety
SRS/SRT Program notices, and simulations of clinical mishaps to rehearse emer-
gency response procedures.
Clinical implementation of an SRS/SRT program requires careful
up-front planning of scope of indications to be treated, staff
required to implement the clinical and technical aspects of the References
program, device selection, credentialing standards, and an [1] Hall EJ. Radiation biology. Cancer 1985; 55 (Suppl): 2051–2057
ongoing program of SRS/SRT-specific training. [2] Kirkpatrick JP, Meyer JJ, Marks LB. The linear-quadratic model is inappropri-
When developing the scope of the program, important con- ate to model high dose per fraction effects in radiosurgery. Semin Radiat On-
siderations include the clinical and technical expertise of the col 2008; 18: 240–243
[3] Timmerman RD. An overview of hypofractionation and introduction to this
treatment team, the expected patient load on the machine (and
issue of seminars in radiation oncology. Semin Radiat Oncol 2008; 18: 215–
whether or not this load is on top of a traditional radiotherapy 222
load), and the availability of published national protocols or [4] Wilcox EE, Daskalov GM, Lincoln H. Stereotactic radiosurgery-radiotherapy:
other clinical guidelines for treating each indication. Departures should Monte Carlo treatment planning be used for all sites? Pract Radiat On-
col 2011; 1: 251–260
from established clinical standards should only be attempted
[5] Adler JR Jr, Chang SD, Murphy MJ, Doty J, Geis P, Hancock SL. The CyberKnife:
with oversight and approval of the institutional review board. a frameless robotic system for radiosurgery. Stereotact Funct Neurosurg
The SRS/SRT requires workflows and time commitments that 1997; 69: 124–128
differ, and are in many instances greater, than what is required [6] Friedman WA, Bova FJ. The University of Florida radiosurgery system. Surg
for traditional radiotherapy treatments. Personnel and time Neurol 1989; 32: 334–342
[7] Bova F, Spiegelmann R, Friedman WA. A device for experimental radiosur-
estimates should bear this in mind. A variety of sources can be
gery. Stereotact Funct Neurosurg 1991; 56: 213–219
used to estimate required staffing levels. National professional [8] Wurm RE, Cosgrove VP, Schlenger L, et al. Commissioning of a micro-multi-
organizations such as the AAPM, American Association of Neu- leaf collimator for conformal stereotactic radiosurgery and radiotherapy.
rological Surgeons/Congress of Neurological Surgeons (AANS/ Front Radiat Ther Oncol 1999; 33: 64–77
[9] Benedict SH, Cardinale RM, Wu Q, Zwicker RD, Broaddus WC, Mohan R. In-
CNS), and American Society for Radiation Oncology (ASTRO)
tensity-modulated stereotactic radiosurgery using dynamic micro-multileaf
have published benchmarks that can aid in estimation.87 collimation. Int J Radiat Oncol Biol Phys 2001; 50: 751–758
Choice of treatment machine should take into consideration [10] Audet C, Poffenbarger BA, Chang P, et al. Evaluation of volumetric modulated
the overall scope of the radiation oncology and neurosurgery arc therapy for cranial radiosurgery using multiple noncoplanar arcs. Med
programs at the institution. In many cases, the existing treat- Phys 2011; 38: 5863–5872
[11] Kondziolka D, Flickinger JC, Hudak R. Results following gamma knife radio-
ment devices within the radiation oncology department will be
surgical anterior capsulotomies for obsessive compulsive disorder. Neurosur-
sufficient for SRS/SRT procedures with little or no modification. gery 2011; 68: 28–32, discussion 23–3
In other cases, a specialized radiosurgery treatment device such [12] Sheehan JP, Patterson G, Schlesinger D, Xu Z. γ knife surgery anterior capsu-
as a Gamma Knife or CyberKnife may make the best clinical and lotomy for severe and refractory obsessive-compulsive disorder. J Neurosurg
financial sense. The critical issue in equipment selection is to 2013; 119: 1112–1118
[13] Benedict SH, Bova FJ, Clark B, et al. Anniversary paper: the role of medical
ensure that the system as a whole (from imaging through deliv-
physicists in developing stereotactic radiosurgery. Med Phys 2008; 35: 4262–
ery) meets the strict requirements for accuracy and precision 4277
for SRS/SRT. [14] Wu A, Lindner G, Maitz AH, et al. Physics of gamma knife approach on con-
Strict credentialing standards and a regular program of SRS/ vergent beams in stereotactic radiosurgery. Int J Radiat Oncol Biol Phys 1990;
18: 941–949
SRT-specific training is another critical component of any radio-
[15] Kassaee A, Das IJ, Tochner Z, Rosenthal DI. Modification of Gill-Thomas-Cos-
surgery program. Several misadministrations in an SRS setting man frame for extracranial head-and-neck stereotactic radiotherapy. Int J Ra-
can be directly traced back to a lack of proper training one or diat Oncol Biol Phys 2003; 57: 1192–1195
more members of the treatment team. The American College of [16] Ekstrand KE, Hinson WH, Bourland JD, et al. The use of a Leksell-BRW adapter
Radiology (ACR) and ASTRO provide definitions and minimum for linac radiosurgery as an adjunct to Gamma Knife treatment. Phys Med Bi-
ol 2003; 48: 4105–4110
qualifications for neurosurgeons, radiation oncologists, medical
[17] Park JH, Han JH, Kim CY, et al. Effect of fiducial marker defects on stereotactic
physicists, dosimetrists, and radiation therapists who are par- target localization in the Leksell stereotactic system. Med Biol Eng Comput
ticipating in radiosurgery procedures.88 In addition, for devices 2011; 49: 775–782
that make use of radioactive materials, the U.S. Nuclear Regula- [18] Gilbeau L, Octave-Prignot M, Loncol T, Renard L, Scalliet P, Grégoire V. Com-
tory Commission and various agreement states have specific parison of setup accuracy of three different thermoplastic masks for the
treatment of brain and head and neck tumors. Radiother Oncol 2001; 58:
regulatory requirements for experience, credentials, and train-
155–162
ing required for authorized users (radiation oncologists) and [19] Fuss M, Salter BJ, Cheek D, Sadeghi A, Hevezi JM, Herman TS. Repositioning
authorized medical physicists performing radiosurgery. accuracy of a commercially available thermoplastic mask system. Radiother
Because of the different workflows, targeting strategies, pre- Oncol 2004; 71: 339–345
[20] Salter BJ, Fuss M, Vollmer DG, et al. The TALON removable head frame system
scription doses, and specialized treatment devices used in SRS/
for stereotactic radiosurgery/radiotherapy: measurement of the reposition-
SRT, specific training is recommended. Up-front training prior ing accuracy. Int J Radiat Oncol Biol Phys 2001; 51: 555–562
to clinical operation should involve vendor-specific training on [21] Sweeney R, Bale R, Vogele M, et al. Repositioning accuracy: comparison of a
the device(s) to be used for SRS/SRT, as well as observation at noninvasive head holder with thermoplastic mask for fractionated radiother-
centers with extensive experience in performing SRS/SRT. apy and a case report. Int J Radiat Oncol Biol Phys 1998; 41: 475–483

39
The Physics of Radiosurgery

[22] Schlesinger D, Xu Z, Taylor F, Yen CP, Sheehan J. Interfraction and intrafraction [47] Kuo JS, Yu C, Petrovich Z, Apuzzo ML. The CyberKnife stereotactic radiosur-
performance of the Gamma Knife Extend system for patient positioning and gery system: description, installation, and an initial evaluation of use and
immobilization. J Neurosurg 2012; 117 (Suppl): 217–224 functionality. Neurosurgery 2003; 53: 1235–1239, discussion 1239
[23] Baumert BG, Egli P, Studer S, Dehing C, Davis JB. Repositioning accuracy of [48] Mackie TR, Holmes T, Swerdloff S, et al. Tomotherapy: a new concept for the
fractionated stereotactic irradiation: assessment of isocentre alignment for delivery of dynamic conformal radiotherapy. Med Phys 1993; 20: 1709–1719
different dental fixations by using sequential CT scanning. Radiother Oncol [49] Andrews DW, Bednarz G, Evans JJ, Downes B. A review of 3 current radiosur-
2005; 74: 61–66 gery systems. Surg Neurol 2006; 66: 559–564
[24] Ryken TC, Meeks SL, Pennington EC, et al. Initial clinical experience with [50] Lindquist C, Paddick I. The Leksell Gamma Knife Perfexion and comparisons
frameless stereotactic radiosurgery: analysis of accuracy and feasibility. Int J with its predecessors. Neurosurgery 2007; 61 (Suppl): 130–140, discussion
Radiat Oncol Biol Phys 2001; 51: 1152–1158 140–141
[25] Ruschin M, Nayebi N, Carlsson P, et al. Performance of a novel repositioning [51] Lutz W, Winston KR, Maleki N. A system for stereotactic radiosurgery with a
head frame for gamma knife perfexion and image-guided linac-based intra- linear accelerator. Int J Radiat Oncol Biol Phys 1988; 14: 373–381
cranial stereotactic radiotherapy. Int J Radiat Oncol Biol Phys 2010; 78: 306– [52] Winston KR, Lutz W. Linear accelerator as a neurosurgical tool for stereotactic
313 radiosurgery. Neurosurgery 1988; 22: 454–464
[26] Das IJ, Ding GX, Ahnesjö A. Small fields: nonequilibrium radiation dosimetry. [53] Grebe G, Pfaender M, Roll M, Luedemann L, Wurm RE. Dynamic arc radiosur-
Med Phys 2008; 35: 206–215 gery and radiotherapy: commissioning and verification of dose distributions.
[27] Alfonso R, Andreo P, Capote R, et al. A new formalism for reference dosimetry Int J Radiat Oncol Biol Phys 2001; 49: 1451–1460
of small and nonstandard fields. Med Phys 2008; 35: 5179–5186 [54] Solberg TD, Boedeker KL, Fogg R, Selch MT, DeSalles AA. Dynamic arc radio-
[28] Charles PH, Cranmer-Sargison G, Thwaites DI, et al. A practical and theoretical surgery field shaping: a comparison with static field conformal and nonco-
definition of very small field size for radiotherapy output factor measure- planar circular arcs. Int J Radiat Oncol Biol Phys 2001; 49: 1481–1491
ments. Med Phys 2014; 41: 041707 [55] Clark GM, Popple RA, Young PE, Fiveash JB. Feasibility of single-isocenter
[29] Drzymala RE, Wood RC, Levy J. Calibration of the Gamma Knife using a new volumetric modulated arc radiosurgery for treatment of multiple brain meta-
phantom following the AAPM TG51 and TG21 protocols. Med Phys 2008; 35: stases. Int J Radiat Oncol Biol Phys 2010; 76: 296–302
514–521 [56] Nataf F, Schlienger M, Liu Z, et al. Radiosurgery with or without A 2-mm mar-
[30] Blogdanich W, Rebelo K. A pinpoint beam strays invisibly, harming instead of gin for 93 single brain metastases. Int J Radiat Oncol Biol Phys 2008; 70: 766–
healing. The New York Times. December 28, 2010. 772
[31] Wu A, Zwicker RD, Kalend AM, Zheng Z. Comments on dose measurements [57] Murphy MJ, Chang SD, Gibbs IC, et al. Patterns of patient movement during
for a narrow beam in radiosurgery. Med Phys 1993; 20: 777–779 frameless image-guided radiosurgery. Int J Radiat Oncol Biol Phys 2003; 55:
[32] Meltsner SG, DeWerd LA. Air kerma based dosimetry calibration for the Lek- 1400–1408
sell Gamma Knife. Med Phys 2009; 36: 339–350 [58] Solberg TD, Medin PM, Mullins J, Li S. Quality assurance of immobilization
[33] McDonald D, Yount C, Koch N, Ashenafi M, Peng J, Vanek K. Calibration of the and target localization systems for frameless stereotactic cranial and extrac-
Gamma Knife Perfexion using TG-21 and the solid water Leksell dosimetry ranial hypofractionated radiotherapy. Int J Radiat Oncol Biol Phys 2008; 71
phantom. Med Phys 2011; 38: 1685–1693 (Suppl): S131–S135
[34] McEwen M, DeWerd L, Ibbott G, et al. Addendum to the AAPM’s TG-51 proto- [59] Wurm RE, Erbel S, Schwenkert I, et al. Novalis frameless image-guided non-
col for clinical reference dosimetry of high-energy photon beams. Med Phys invasive radiosurgery: initial experience. Neurosurgery 2008; 62 (Suppl):
2014; 41: 041501 A11–A17, discussion A17–A18
[35] Sibata CH, Mota HC, Beddar AS, Higgins PD, Shin KH. Influence of detector [60] Noël G, Simon JM, Valery CA, et al. Radiosurgery for brain metastasis: impact
size in photon beam profile measurements. Phys Med Biol 1991; 36: 621– of CTV on local control. Radiother Oncol 2003; 68: 15–21
631 [61] Paddick I. A simple scoring ratio to index the conformity of radiosurgical
[36] Higgins PD, Sibata CH, Siskind L, Sohn JW. Deconvolution of detector size ef- treatment plans. Technical note. J Neurosurg 2000; 93 (Suppl 3): 219–222
fect for small field measurement. Med Phys 1995; 22: 1663–1666 [62] Hazard LJ, Wang B, Skidmore TB, et al. Conformity of linac-based stereotactic
[37] Bednarz G, Saiful Huq M, Rosenow UF. Deconvolution of detector size effect radiosurgery using dynamic conformal arcs and micro-multileaf collimator.
for output factor measurement for narrow Gamma Knife radiosurgery beams. Int J Radiat Oncol Biol Phys 2009; 73: 562–570
Phys Med Biol 2002; 47: 3643–3649 [63] Hill DL, Hawkes DJ, Crossman JE, et al. Registration of MR and CT images for
[38] Sauer OA, Wilbert J. Measurement of output factors for small photon beams. skull base surgery using point-like anatomical features. Br J Radiol 1991; 64:
Med Phys 2007; 34: 1983–1988 1030–1035
[39] D’Agostino J, Pelczynski L. An overview of cyclotron treatment, Bragg peak [64] Gross MW, Weber WA, Feldmann HJ, Bartenstein P, Schwaiger M, Molls M.
proton hypophysectomy and Bragg peak radiosurgery for arteriovenous mal- The value of F-18-fluorodeoxyglucose PET for the 3-D radiation treatment
formation of the brain. J Neurosurg Nurs 1979; 11: 208–214 planning of malignant gliomas. Int J Radiat Oncol Biol Phys 1998; 41: 989–
[40] Macdonald OK, Kruse JJ, Miller JM, et al. Proton beam radiotherapy versus 995
three-dimensional conformal stereotactic body radiotherapy in primary pe- [65] Pirzkall A, McKnight TR, Graves EE, et al. MR-spectroscopy guided target de-
ripheral, early-stage non-small-cell lung carcinoma: a comparative dosimet- lineation for high-grade gliomas. Int J Radiat Oncol Biol Phys 2001; 50: 915–
ric analysis. Int J Radiat Oncol Biol Phys 2009; 75: 950–958 928
[41] Chen CC, Chapman P, Petit J, Loeffler J. Proton radiosurgery in neurosurgery. [66] Nelson SJ, Graves E, Pirzkall A, et al. In vivo molecular imaging for planning
Neurosurg Focus 2007; 23: E5 radiation therapy of gliomas: an application of 1 H MRSI. J Magn Reson Imag-
[42] Deng J, Ma CM, Hai J, Nath R. Commissioning 6 MV photon beams of a stereo- ing 2002; 16: 464–476
tactic radiosurgery system for Monte Carlo treatment planning. Med Phys [67] Seo Y, Fukuoka S, Nakagawara J, et al. Effect of Gamma Knife radiosurgery on
2003; 30: 3124–3134 acoustic neurinomas. Assessment by 99mTc-DTPA-human serum albumin-
[43] Fragoso M, Wen N, Kumar S, et al. Dosimetric verification and clinical evalua- and 201TlCl-single photon emission computed tomography. Stereotact Funct
tion of a new commercially available Monte Carlo-based dose algorithm for Neurosurg 1996; 66 (Suppl 1): 93–102
application in stereotactic body radiation therapy (SBRT) treatment planning. [68] Constable RT, Henkelman RM. Contrast, resolution, and detectability in MR
Phys Med Biol 2010; 55: 4445–4464 imaging. J Comput Assist Tomogr 1991; 15: 297–303
[44] Xiao Y, Papiez L, Paulus R, et al. Dosimetric evaluation of heterogeneity cor- [69] Fransson A, Andreo P, Pötter R. Aspects of MR image distortions in radiother-
rections for RTOG 0236: stereotactic body radiotherapy of inoperable stage I– apy treatment planning. Strahlenther Onkol 2001; 177: 59–73
II non-small-cell lung cancer. Int J Radiat Oncol Biol Phys 2009; 73: 1235– [70] Jaffray DA, Siewerdsen JH. Cone-beam computed tomography with a flat-
1242 panel imager: initial performance characterization. Med Phys 2000; 27:
[45] Wilcox EE, Daskalov GM, Lincoln H, Shumway RC, Kaplan BM, Colasanto JM. 1311–1323
Comparison of planned dose distributions calculated by Monte Carlo and [71] Meeks SL, Bova FJ, Friedman WA, Buatti JM, Moore RD, Mendenhall WM.
Ray-Trace algorithms for the treatment of lung tumors with CyberKnife: a IRLED-based patient localization for linac radiosurgery. Int J Radiat Oncol Biol
preliminary study in 33 patients. Int J Radiat Oncol Biol Phys 2010; 77: 277– Phys 1998; 41: 433–439
284 [72] Cerviño LI, Pawlicki T, Lawson JD, Jiang SB. Frame-less and mask-less cranial
[46] Yu C, Shepard D. Treatment planning for stereotactic radiosurgery with pho- stereotactic radiosurgery: a feasibility study. Phys Med Biol 2010; 55: 1863–
ton beams. Technol Cancer Res Treat 2003; 2: 93–104 1873

40
The Physics of Radiosurgery

[73] Peng JL, Kahler D, Li JG, et al. Characterization of a real-time surface image- [81] Treuer H, Kocher M, Hoevels M, et al. Impact of target point deviations on
guided stereotactic positioning system. Med Phys 2010; 37: 5421–5433 control and complication probabilities in stereotactic radiosurgery of AVMs
[74] Fallone BG, Murray B, Rathee S, et al. First MR images obtained during mega- and metastases. Radiother Oncol 2006; 81: 25–32
voltage photon irradiation from a prototype integrated linac-MR system. [82] Gordon JJ, Siebers JV. Convolution method and CTV-to-PTV margins for finite
Med Phys 2009; 36: 2084–2088 fractions and small systematic errors. Phys Med Biol 2007; 52: 1967–1990
[75] Raaymakers BW, Lagendijk JJ, Overweg J, et al. Integrating a 1.5 T MRI scanner [83] Rowshanfarzad P, Sabet M, O’Connor DJ, Greer PB. Isocenter verification for
with a 6 MV accelerator: proof of concept. Phys Med Biol 2009; 54: N229– linac-based stereotactic radiation therapy: review of principles and techni-
N237 ques. J Appl Clin Med Phys 2011; 12: 3645
[76] Klein EE, Hanley J, Bayouth J, et al. Task Group 142, American Association of [84] Mack A, Czempiel H, Kreiner HJ, Dürr G, Wowra B. Quality assurance in ster-
Physicists in Medicine. Task Group 142 report: quality assurance of medical eotactic space. A system test for verifying the accuracy of aim in radiosurgery.
accelerators. Med Phys 2009; 36: 4197–4212 Med Phys 2002; 29: 561–568
[77] LoSasso T, Chui CS, Ling CC. Comprehensive quality assurance for the delivery [85] Ma L, Chuang C, Descovich M, Petti P, Smith V, Verhey L. Whole-procedure
of intensity modulated radiotherapy with a multileaf collimator used in the clinical accuracy of gamma knife treatments of large lesions. Med Phys 2008;
dynamic mode. Med Phys 2001; 28: 2209–2219 35: 5110–5114
[78] Fraass B, Doppke K, Hunt M, et al. American Association of Physicists in Medi- [86] Dieterich S, Cavedon C, Chuang CF, et al. Report of AAPM TG 135: quality as-
cine Radiation Therapy Committee Task Group 53: quality assurance for clin- surance for robotic radiosurgery. Med Phys 2011; 38: 2914–2936
ical radiotherapy treatment planning. Med Phys 1998; 25: 1773–1829 [87] Benedict SH, Yenice KM, Followill D, et al. Stereotactic body radiation ther-
[79] Stern RL, Heaton R, Fraser MW, et al. AAPM Task Group 114. Verification of apy: the report of AAPM Task Group 101. Med Phys 2010; 37: 4078–4101
monitor unit calculations for non-IMRT clinical radiotherapy: report of AAPM (Corrected in Med Phys 2012; 39:563 for dosage error in text)
Task Group 114. Med Phys 2011; 38: 504–530 [88] Seung SK, Larson DA, Galvin JM, et al. American College of Radiology (ACR)
[80] Huq MS, Fraass BA, Dunscombe PB, et al. A method for evaluating quality as- and American Society for Radiation Oncology (ASTRO) Practice Guideline for
surance needs in radiation therapy. Int J Radiat Oncol Biol Phys 2008; 71 the Performance of Stereotactic Radiosurgery (SRS). Am J Clin Oncol 2013;
(Suppl): S170–S173 36: 310–315

41
Radiosurgical Devices

6 Radiosurgical Devices
Daniel E. Hyer, Frank J. Bova, and John Buatti

response and normal tissue complications are based on such a


Key Points prescription technique.2,3 If radiosurgery is performed with a
new device that requires the use of a margin to account for
● Two of the most important aspects of radiosurgery are a uncertainties in patient positioning, the increase in volume of
sharp dose gradient and accurate target positioning that en- irradiated tissue is likely to add a significant risk of complica-
ables treatment of a radiographically defined target without tion to what may otherwise be a safe and effective treatment.
a margin. For example, the inclusion of a 2-mm margin would increase
● The addition of a margin to account for positioning uncer- the volume of irradiated tissue by nearly 60% for a 24-mm-
tainties directly impacts the target dose that may be safely diameter target. ▶ Table 6.1 shows the volumes of typical radio-
delivered. surgery targets, and the volume increases when a 2-mm margin
● When a new system is developed, a qualified physicist should is added.
perform an end-to-end test to understand its strengths and The addition of a margin to account for positioning uncer-
weaknesses. Pretreatment tests validating all system align- tainties directly impacts the target dose that may be safely
ments should also be conducted as part of any radiosurgery delivered. Relationships between radiosurgical volume and risk
program. of complications have long been established.4 For example, if it
● Frame-based radiosurgery has been the gold standard of is safe and effective to deliver 20 Gy to a 2.4-cm-diameter tar-
radiosurgery treatment since its inception. The frame-based get, then if a 2-mm margin is used, the largest lesion that could
approach provides reliable localization when paired with the be safely treated with 20 Gy would be reduced to 2.0 cm in
appropriate delivery equipment and its treatment efficacy is diameter (see ▶ Table 6.1). This not only reduces the effective-
well established. ness of the treatment for larger target volumes, but also need-
● To overcome frame-based limitations, noninvasive, often lessly exposes normal tissue to the target dose. This would be
referred to as frameless, positioning systems have been the surgical equivalent of removing the diseased tissue and
developed. With a frameless system, immobilization and then removing another 2-mm margin of normal brain.
localization are no longer inherently linked. Although positioning uncertainties certainly limit the ability
to effectively perform radiosurgery, the importance of a sharp
dose gradient must also be remembered. Values provided in
▶ Table 6.2 show the increase in irradiated tissue if one
6.1 Introduction assumes a system with a 6-mm dose gradient is used instead of
a system with a 3-mm gradient. In this context, gradient refers
Radiosurgery was first defined by Leksell as “a single high dose
to the distance between the prescription isodose line and a line
fraction of radiation, stereotactically directed to an intracranial
that is 50% of the prescription isodose line. If we again use the
region of interest.”1 By focusing multiple beams of radiation on
previously referenced dose-volume relationship, it can be seen
the target tissue, Leksell was able to produce a sharp dose gra-
that the target diameter that could be safely treated with 20 Gy
dient outside of the selected target tissue that allowed the sur-
rounding normal tissue to be spared. This differed from other
forms of fractionated radiotherapy at the time, as it did not rely
Table 6.1 Increase in target volume with the addition of a 2-mm margin
on the differential sublethal damage repair between the target
and normal tissue, but rather on dose concentration to the tar- Volume of target (cc)
get achieved by an enhanced technical delivery paradigm and a Diameter of target (cm) No margin 2-mm margin % increase
very rapid falloff of dose in the adjacent normal tissue. It was
1 0.52 1.44 174
evident early on that for this new therapy to be effective, the
target must be readily identified (generally by imaging) and 1.2 0.90 2.14 137
accurately positioned, leading to the stereotactic approach to
1.4 1.44 3.05 113
radiation delivery. If this was not possible, the sharp dose gra-
dient would actually work against the clinician as the target tis- 1.6 2.14 4.19 95
sue could easily be missed or undertreated and normal tissue
1.8 3.05 5.58 83
could receive a very high radiation dose.
This chapter will focus on stereotactic positioning systems 2 4.19 7.24 73
and collimators necessary to successfully deliver radiosurgery.
2.2 5.58 9.20 65
Positioning devices and collimators are both important topics
to explore, as the spatial accuracy of dose delivery and sharp- 2.4 7.24 11.49 59
ness of the beam profile dictate the treatment approach more 2.6 9.20 14.14 54
so than the type or design of the radiation source. Historically,
radiosurgery dose distributions have been prescribed to the 2.8 11.49 17.16 49
boundary of the enhancing volume (i.e., without margins added 3 14.14 20.58 46
to the clinical target volume). The clinical data regarding tumor

42
Radiosurgical Devices

would be decreased from 2.4 cm to approximately 1.8 cm when technologies can be broken down into two main categories:
moving from a 3-mm gradient to a 6-mm gradient. These two minimally invasive systems and noninvasive systems.5 Mini-
examples point out two of the most important aspects of radio- mally invasive systems include the popular frame-based sys-
surgery: sharp dose gradients and accurate target positioning tems that are attached to the patient’s skull through the use of
that enables treatment of a radiographically defined target pins, whereas noninvasive systems include devices such as
without a margin. maxillary fixation systems, thermoplastic masks, and in-room
imaging. Before discussing positioning systems in detail, a key
distinction should be made between localization, sometimes
6.2 Stereotactic Positioning referred to as alignment or positioning, and immobilization. In
Systems the simplest sense, localization refers to the ability to position
the target at a desired location, usually the isocenter of the
The term stereotactic refers to the use of a mathematically treatment device, and immobilization refers to the ability to
defined three-dimensional (3D) coordinate system to precisely hold the target in place during treatment delivery. It is impor-
locate targets inside of the brain. Various technologies tant to understand that these two aspects are not always linked,
have been developed for stereotactic localization, and these and both must be considered for radiosurgery. For example, a
thermoplastic mask may be used to immobilize a patient, but
provides little benefit for localization. Similarly, a cone-beam
Table 6.2 Increase in irradiated tissue due to gradient computed tomography (CBCT) scan at the treatment machine
may be used for localization, but provides no benefit in immo-
Volume of 50% of RX (cc)
bilization. The difference between a localization error and an
Diameter of immobilization error can be seen in ▶ Fig. 6.1.
target (cm) 3-mm gradient 6-mm gradient % increase The following sections detail devices commonly used in
1 2.14 5.58 160 radiosurgery and describe their proper use in each of these
categories.
1.2 3.05 7.24 137

1.4 4.19 9.20 120


6.3 Minimally Invasive Positioning
1.6 5.58 11.49 106
Systems
1.8 7.24 14.14 95
Minimally invasive systems, such as the Leksell,7 Brown-Rob-
2 9.20 17.16 86
erts-Wells,8 and Brainlab9 (Munich, Germany) frames, are rig-
2.2 11.49 20.58 79 idly affixed to the patient’s head during treatment. This is
accomplished through the use of pins that are tightened against
2.4 14.14 24.43 73
the patient’s skull, as shown in ▶ Fig. 6.2. By rigidly affixing a
2.6 17.16 28.73 67 frame to the patient’s skull, a geometric relationship is estab-
lished between the patient’s intracranial anatomy and the ster-
2.8 20.58 33.51 63
eotactic frame that has a known geometry. An external fiducial,
3 24.43 38.79 59 such as a localizer box, is then attached to the frame, and a
computed tomography (CT) scan is acquired. The positions of

Fig. 6.1 Images showing localization errors (left) and immobilization errors (right). With localization errors, the target moves away from the planned
position and the effect is a geometric miss. With immobilization errors, the target moves during treatment, and the effect is to blur the dose delivered
to the target.6 (Used with permission from JNSPG.)

43
Radiosurgical Devices

relies solely on mechanical positioning of the patient’s head


based on the stereotactic coordinate system that is derived
from the imaging acquired for treatment planning. The latest
Gamma Knife unit, called the Perfexion (PFX), was introduced
in 2006 and performs patient positioning with an automatic
positioning system (APS).12 The APS consists of a robotic treat-
ment table that is calibrated to the stereotactic coordinate
system and can automatically position the patient at each treat-
ment isocenter. The head frame is attached to the APS through
a couch mount. The mechanical treatment range in the X/Y/Z
orientations is (160/180/220) mm. In addition to these transla-
tions, the APS can also tilt the entire coordinate system to three
different gamma angles to allow additional clearance during
treatment: 70 degrees (chin up position), 90 degrees (horizon-
tal), or 110 degrees (chin down).
The manufacturer’s specification for agreement between the
radiation focal point and the APS mechanical isocenter is
0.4 mm—the alignment of all radiation beams to a single point
in space. However, one group found the agreement to be as
small as 0.26 mm.12 Additionally, reproducibility of stereotactic
coordinates—the accuracy that an individual coordinate can be
revisited—with the APS is stated to be better than 0.05 mm. The
mechanical accuracy afforded by this system is often considered
the gold standard to which other technologies are compared.
However, this mechanical accuracy of positioning is only one
component of a multifaceted treatment process. With such a
small mechanical accuracy, the primary source of localization
errors is the imaging acquired for treatment planning. Previous
studies have shown that determining the coordinates of a high
contrast object on a CT scan can be achieved with an accuracy
of approximately ± 1 voxel.13 The scan diameter required to
accommodate a stereotactic localizer is approximately 350 mm.
With a 512 × 512 imaging matrix, this results in an in-plane pix-
el dimension of 0.67 × 0.67 mm. Therefore, the positional error
Fig. 6.2 Minimally invasive head frame and localizer box. of the APS can be thought to be quantum limited, with the
quantum sink being the determination of the stereotactic coor-
dinates from the CT scan.
several fiducial rods embedded in the localizer box are then
located in each slice of the CT scan, and because the geometric
relationship between each fiducial rod and the frame is well 6.3.2 Linac Frame Systems
known, a coordinate system transformation can be determined
that will convert the native CT coordinate system to a new coor- Two main challenges present themselves when adapting a linac
dinate system based on the localizer’s fiducial system.10,11 to perform radiosurgery: achieving high mechanical isocentric
Applying this coordinate system transformation to the entire accuracy and accurately localizing the patient at the treatment
CT dataset will allow the position of any point in the image vol- machine. Several systems have been developed to overcome
ume to be uniquely referenced to the stereotactic ring’s coordi- these challenges; each are discussed in detail in the following
nate system, which is rigidly fixed to the patient’s head. The sections.
intracranial target can then be aligned to the treatment unit’s
isocenter using the frame and associated coordinate system— Floor Mounted
providing the localization necessary for treatment delivery.
With the frame-based approach, the frame also serves as an At the University of Florida, Gainesville, a floor-stand system
immobilization device, as it is rigidly mounted to the patient was developed to perform stereotactic radiosurgery with a
support system and holds the patient’s head motionless during standard linac.13,14 This system, shown in ▶ Fig. 6.3, is
treatment delivery. mounted to the treatment room floor and attaches to the
linac gantry via a sliding gimbal mount. The floor-stand sys-
tem links the beam isocenter to the patient rotation isocen-
ter, both of which are necessary for non-coplanar treatment
6.3.1 Gamma Knife Frame System delivery. This linkage relieves the burden of isocentric accu-
The Gamma Knife is a unique system for performing stereotac- racy from not only linac gantry rotation, but also the patient
tic radiosurgery (SRS), and it was developed around the frame- support system, with the latter being a subsystem that is not
based approach. Localization with the Gamma Knife system only subject to large changes in load due to varying patient

44
Radiosurgical Devices

Fig. 6.3 Floor-stand system for performing ra-


diosurgery. (a) Side view of the gantry and
gimbal in the 0 degree position; (b) top view of
the gantry and gimbal rotated through 90
degrees. The floor stand is attached to the linac
through a sliding gimbal bearing (A) and has
precision bearings for the gantry arm (B) as well
as couch rotation (C). The system produces a
mechanical accuracy of 0.2 ± 0.1 mm.13

size, but also one that historically has been difficult to main- the voxel size of the imaging modality used to determine the
tain due to component wear. Through the use of high-preci- desired isocenter locations, identical to the uncertainty of the
sion bearings and low-mass components, the floor stand’s Gamma Knife.
gantry arm and patient support system are capable of high-
precision rotations about the isocenter. The floor stand has
been shown to have a mechanical accuracy of 0.2 ± 0.1 mm as
Couch Mounted
determined through a Winston-Lutz test at various couch With a couch-mounted SRS system, the patient is placed on
and gantry positions. 13 This level of mechanical accuracy the standard linac treatment couch and their head is fastened
rivals that of Gamma Knife systems and cannot be achieved, to an adapter that overhangs the end of the treatment
even on modern linacs, without the use of the floor-stand couch.3 The adapter typically includes mechanical adjust-
system. ments to allow fine positioning of the patient’s head in 6
The floor-stand system also has provisions for accurately degrees of freedom. However, unlike the floor stand and
localizing the patient during radiosurgery. As seen in ▶ Fig. 6.3, Gamma Knife systems discussed so far, the positioning accu-
the patient’s head frame is attached to the patient support sys- racy and rigidity of a standard treatment couch is not suffi-
tem of the floor stand for treatment. The patient support sys- cient for absolute alignment of the linac’s isocenter to a
tem has manual adjustments in the longitudinal, lateral, and targeted stereotactic coordinate. Instead, couch-mount sys-
vertical directions along with a calibrated vernier scale. As part tems must rely on room lasers to align the target point to the
of the commissioning process, the vernier scale is calibrated linac’s isocenter. When room lasers are used, the stereotactic
such that (0,0,0) coincides with the center of the stereotactic coordinates are determined from the planning CT and then
volume of the Brown-Roberts-Wells frame, and the translation- transferred to the exterior of the localizer box through either
al axes are aligned with that of the head ring. Similar to the printouts or panels with a calibrated scale (see ▶ Fig. 6.4).
Gamma Knife system, the positioning of the patient is com- The room lasers, which are used as a link between the room
pletely mechanical and based on the stereotactic coordinate coordinates and stereotactic coordinates, are then aligned
system derived from the planning CT. The resolution of the ver- with the markings on the localizer box using the mechanical
nier scale used to set the stereotactic coordinates has a resolu- adjustments on the couch adapter. The use of such a system
tion of 0.1 mm. The main uncertainties in localization and brings the room laser systems into the quality assurance loop
treatment alignment when using the floor stand are based on for each radiosurgical procedure. For this reason, the room

45
Radiosurgical Devices

Imaging Dependent
When SRS treatments with linacs first started over 30 years
ago, megavoltage (MV) portal images were the only in-room
imaging modality available. Megavoltage portal images were
unsuitable due to their poor contrast, resolution, and inabil-
ity to visualize intracranial anatomy: all of which limited
their utility and adoption for SRS localization. However,
recent developments in image-guided radiotherapy, including
the advent of gantry-mounted kilovoltage (kV) CBCT, have
led some groups to localize frame-based SRS patients with
imaging rather than the mechanical or laser-based systems
discussed thus far.
With these systems, the patient’s head frame is fastened to
an adapter on the treatment couch as described in the pre-
vious section. At this point, rather than mechanical localiza-
tion based on the stereotactic coordinate system, a CBCT is
acquired of the patient’s head. An automatic registration
between the planning CT and (treatment) CBCT can then be
performed to calculate the shift that must be applied to local-
ize the patient to the treatment isocenter. One group eval-
uated the accuracy of such a workflow by acquiring the CBCT
with the localizer box still attached to the patient’s head and
mapping the CBCT dataset to the same stereotactic coordinate
system as the planning CT.16 Using this method, the mean
image registration accuracy between the CBCT and the plan-
ning CT dataset was found to be 0.28 mm. A potential advant-
age of CBCT-based SRS is the ability to use the 3D image to
detect frame slippage by comparing the relationship between
Fig. 6.4 Localizer box with computer-generated overlays for room laser the frame and patient anatomy in both the CBCT and planning
alignment. (Image courtesy of Joseph C. T. Chen, MD, PhD.) CT, which are taken at two different time points. Without a
CBCT, a depth helmet can be used to detect frame slippage,
but its interpretation is highly subjective and accuracy is 1 to
2 mm.5
lasers must be accurately aligned to the treatment machine
One of the biggest sources of uncertainty with an imaging-
isocenter before being used to position the patient. This can
dependent system such as CBCT is the potential misalignment
be performed by imaging a radiopaque marker at the inter-
between the treatment and imaging isocenter, which is gener-
section of the lasers to verify alignment before patient treat-
ally held to within a tolerance of < 1 mm.17 A misalignment of
ment. Demonstration of laser alignment prior to treatment
1 mm can severely limit the clinician’s ability to perform SRS
should be considered a necessary step with couch-mounted
when targeting small lesions and should be carefully evaluated
systems.
on an individual system basis. The isocentric wander of the
Using a couch-mounted system also puts significant iso-
machine should also be evaluated, as this will be another source
centric demands on the treatment machine. Isocenter wan-
of uncertainty, similar to the couch-mount systems described
der due to gantry, collimator, and couch rotations must be
in the previous section. As with laser-alignment systems, a
minimized to ensure accurate targeting. Wander can be mini-
Winston-Lutz test demonstrating coincidence between the
mized with a dedicated SRS linac that is tuned from the fac-
imaging and treatment isocenters should be part of the pre-
tory for this purpose. Values of 0.3 ± 0.1 and 0.6 ± 0.1 mm for
treatment quality assurance process. It should also be noted
gantry and couch isocentricity, respectively, are reported in
that a 6-degree-of-freedom adjustment of the head is necessary
the literature for a dedicated SRS linac. 15 The localization
for proper patient alignment when using a CBCT system for
accuracy of a couch-mount system, which relies on aligning
radiosurgery.
the room lasers to an isocenter that is not a point but rather
a distribution in space, will inherently be less accurate than
floor stand or Gamma Knife systems in which the isocenter
accuracy is on the order of 0.2 to 0.4 mm. Additionally, the 6.4 Noninvasive Positioning
verification of isocentric alignment at patient support angles
other than 0 degree can be difficult because the lasers do not
Systems
rotate with the patient for non-coplanar beam alignment. Minimally invasive frame-based radiosurgery has been the gold
Visual alignment of the room lasers to markings on the local- standard of radiosurgery treatment since its inception. The
izer box also have inherent inaccuracies that vary between frame-based approach provides reliable localization when
users and could easily account for an additional 0.5 mm of paired with the appropriate delivery equipment and its treat-
localization uncertainty. ment efficacy is well established. However, the use of a head

46
Radiosurgical Devices

frame is associated with substantial shortcomings relating to the teeth in the upper jaw provide the structure necessary to
patient comfort and clinical workflow. rigidly immobilize the patient’s head. A bite plate is made by
To overcome these shortcomings, several noninvasive, often filling an upper dentate impression tray with a rapidly harden-
referred to as frameless, positioning systems have been devel- ing dental material, placing the tray in the patient’s mouth, and
oped. With a frameless system, immobilization and localization waiting for the material to harden and form an impression
are no longer inherently linked. Due to this decoupling, immo- around the patient’s upper teeth. After hardening, the bite plate
bilization and localization will be discussed separately for can be removed from the patient’s mouth and connected to a
frameless systems in the following sections. couch-mounted frame to rigidly immobilize the patient’s head.
When fitted by experienced technicians, bite plates can offer
reproducible and rigid immobilization. The reproducibility of
6.4.1 Immobilization
reseating the bite plate in the patient’s mouth has been investi-
Thermoplastic Mask gated and reported to be as low as 0.24 mm.20 One group has
further developed the bite plate system by attaching a vacuum
One of the most common immobilization devices used in radio-
tube to the dental cast to provide added fixation by providing
therapy is a custom-made thermoplastic mask that conforms to
suction to the hard palate.19
the patient’s outer-body contour and is affixed to the patient
Elekta (Stockholm, Sweden) has commercialized a vacuum-
table (see ▶ Fig. 6.5). A thermoplastic mask is made of a low-
assisted bite plate called the Extend system, allowing the Gam-
melting-point plastic that is rigid at room temperature, but easy
ma Knife to be used for multifraction treatments. The system
to mold and stretch when heated. To create a mask, the patient
consists of a vacuum-monitored bite plate with associated frame,
is first positioned on the simulation table, generally in a supine
as shown in ▶ Fig. 6.6. The performance of the Extend system
orientation with a small head cushion to improve patient com-
was recently evaluated with a digital probe, and the mean intra-
fort, and the thermoplastic mask is warmed in a water bath to
fraction accuracy over 36 fractional treatments was found to be <
approximately 150°F so that it can be stretched and fitted to the
0.5 mm.6 It is expected that similar bite plate systems could
patient’s face. The mask should be molded to incorporate as
offer equivalent intrafraction accuracy. Because this system is
many hard body points as possible (bridge of nose, chin, etc.).
rigidly fixed to the APS, which is capable of reproducing coordi-
After fitting, the mask is attached to the treatment couch and
nates to within 0.05 mm, it can also be used for localization of
allowed to cool in place to provide a rigid mold of the patient in
the patient’s head, and the mean interfraction setup difference
the treatment position. Many variations of masks exist, mainly
was found to be 0.64 mm across the same patient cohort.
differing in extent of coverage (head or head and shoulder) and
material thickness. When properly used, intrafraction motion
as small as 0.7 mm has been reported with thermoplastic Ear Canal
masks, a testament to their rigidity and custom-fitted construc-
A unique noninvasive immobilization system that has not seen
tion.18 However, interfraction differences do occur and have
widespread use in the United States is the Laitinen stereotactic
been reported to be as large as 2 to 3 mm,18,19 clearly demon-
frame, which consists of two earplugs and a nasion held togeth-
strating the need for masks to be paired with a reliable localiza-
er with a reinforced plastic and aluminum frame.21 When prop-
tion device.
erly fitted, this frame provides a rigid system to immobilize the
patient’s head based on external anatomy. Reports of intrafrac-
Bite Plate tion motion using such a system are limited in the literature,
Bite plates, also called upper jaw fixation devices, offer a method but one may expect accuracy similar to the bite plate systems
for noninvasively immobilizing the patient’s head. Anatomically, described in the previous section.

Fig. 6.5 Thermoplastic mask used for immobili-


zation during frameless radiosurgery. (Image
courtesy of CIVCO Medical Solutions.)

47
Radiosurgical Devices

Fig. 6.6 Elekta extend vacuum-assisted bite plate


for immobilization. (Image courtesy of Elekta.)

6.4.2 Localization objects within the field-of-view of the two cameras.24 Objects
such as small, reflective aluminum-covered spheres are often
Multiplanar X-Ray Imaging used as they can be easily detected by light-emitting diode
(LED) cameras emitting infrared light. By establishing a rigid
X-ray imaging is becoming one of the most popular techniques
body relationship between the locations of the objects and a
for localization in radiosurgery. Several different systems exist,
reference point, such as the treatment isocenter, the optical
ranging from 2D stereoscopic imaging systems to 3D CBCT
tracking cameras can be used to effectively track the location of
systems. Cone-beam computed tomography systems can be
the target. This principle has been applied to intracranial ster-
applied to frameless radiosurgery in the same manner as
eotactic radiotherapy and SRS.25,26
frame-based radiosurgery, with the frame being replaced with
One of the most successful applications of such a system is
a frameless immobilization device. Two-dimensional stereo-
the Varian Optical Guidance Platform (Varian, Inc., Palo Alto,
scopic imaging systems are also used for radiosurgery and may
CA), which consists of a bite plate attached to an array of small
offer an advantage over CBCT systems for frameless radiosur-
reflective balls that can be tracked by ceiling-mounted cameras.
gery, as images can be quickly acquired throughout the treat-
An example of a bite plate with markers attached is shown in
ment duration at a wide range of couch and gantry angles to
▶ Fig. 6.7. Here the bite plate has been combined with a ther-
verify positioning. This is advantageous, as immobilization with
moplastic mask for immobilization. The average localization
a frameless approach may exhibit a wider range of intrafraction
error when using such a system has been reported to be
motion than with frame-based radiosurgery; therefore, imaging
0.82 mm.20 One of the biggest advantages of optical tracking is
during the treatment duration can provide desirable position
the ability to continuously monitor the position of the patient
verification.
rather than at discrete moments in time before or perhaps dur-
One 2D imaging system used for radiosurgery is the ExacTrac
ing treatment as is the case with X-ray images.
system by Brainlab. This system consists of two floor-mounted
kV X-ray units and two ceiling-mounted imaging detectors. As
with any 2D imaging system, a fixed relationship between Surface Monitoring
intracranial lesions and the cranial skeletal anatomy must be
The optical tracking systems described in the previous section
assumed due to the fact that soft-tissue anatomy cannot be
have recently been taken a step further with the introduction
readily visualized on a 2D image. However, even frame-based
of 3D surface imaging with a system of video cameras. Such
systems are subject to the same assumption of a fixed relation-
systems do not require the use of reflective spheres, but rather
ship between skeletal and intracranial anatomy. By acquiring
map and directly track the patient. One such system, marketed
two X-ray images in a stereoscopic geometry and registering
by Vision RT (London, UK) as the AlignRT system, yields sub-
these with digitally reconstructed radiographs (DRRs) calcu-
millimeter accuracy (0.75 mm) for the translational degrees of
lated from the treatment-planning system, 6D corrections can
freedom and less than 0.1 degree for each rotational axis in a
be calculated to position the patient at the treatment isocenter.
phantom study.27 The advantage of surface monitoring is that a
Such systems have a demonstrated submillimeter localization
surrogate for the patient, such as a bite plate, is not needed:
accuracy.22,23
This reduces the potential for error due to malaligned fitting or
incorrect seating. One disadvantage is that the surface motion
can be tracked only with respect to a reference image, which
Optical Tracking can be a surface contour extracted from the planning CT image
Optical tracking involves the use of stereoscopically mounted or a surface image acquired previously with AlignRT. In our
infrared cameras that effectively triangulate the location of experience, the latter provides a better reference surface, as the

48
Radiosurgical Devices

Fig. 6.7 Bite plate with optical array.

body contour determined from the planning CT is highly and hence the dose to normal tissue, outside of the target. The
dependent on a number of factors, including the threshold used dose gradient at the margin of the target is similar for Gamma
for automatic body contour detection. When capturing a refer- Knife and linac-based radiosurgery systems employing cones,
ence surface, an absolute localization device such as X-ray imag- with linac cones offering superior penumbra for large cone sizes
ing should be used first to accurately position the patient. The as a result of the source geometry.29,30 The lateral dose gradient
reference image should include hard structures such as the for a small cone can be visualized in (▶ Fig. 6.8), where the lat-
bridge of the nose in the region of interest; mobile structures eral falloff for a trigeminal neuralgia treatment is plotted for
such as the eyes should be excluded. To enable surface monitor- both Gamma Knife and linac-based plans with circular cones
ing, immobilization devices such as thermoplastic masks must and a varying numbers of treatment arcs.31 In general, cone-
be modified to have an opening around the patient’s face, based radiosurgery with a non-coplanar arc set should yield a
which may compromise their integrity; however, investigations 3D dose distribution with a penumbra (80–20%) in the range of
are underway to potentially perform radiosurgery with no 2 to 3 mm.
immobilization but by constant surface monitoring instead to Although a very sharp dose gradient can be achieved with
detect patient motion.28 a circular cone, some targets do not conform well to the
resultant spherical dose distribution and require multi-iso-
center plans to improve conformity. The use of multiple iso-
6.5 Collimators centers increases the degrees of freedom for developing a
treatment plan and allows the planner to customize not only
In recent years, SRS delivery techniques have expanded beyond the shape, but also the dose gradient in directions near crit-
the Gamma Knife, CyberKnife, and linac-based delivery using ical structures. However, with additional isocenters, the
circular collimators to include the adaptation of linacs with treatment time is also increased, amplifying concerns regard-
multileaf collimators. Although each of these systems offers ing immobilization for frameless patients and comfort for
unique features, they are all bound by the same principle that a frame-based patients. For these reasons, the use of a multi-
large number of beam paths are required to concentrate the leaf collimator (MLC) for irregularly shaped lesions has been
dose in the target area and create a steep dose gradient. It is gaining traction.
important to note that as new SRS delivery techniques emerge,
clinicians must examine the impact on dose conformity com-
pared with historical SRS delivery techniques using Gamma 6.7 Multileaf Collimator
Knife or linac-based delivery with circular collimators. A loss in
dose conformity may force the user to adopt a treatment sched- Compared with cones, a MLC offers the advantage of conform-
ule that deviates from historical radiosurgical data. Although ing to the shape of the target in the beam’s eye view rather than
the use of new equipment may be appropriate in certain clinical being limited to only circular shapes. Traditional MLCs are
situations, a cautious approach is warranted. designed with ~ 40 to 50 cm of distance between the isocenter
and the collimator to allow clearance when treating larger parts
of the body but at the cost of a larger geometric penumbra,
6.6 Cones which is a disadvantage for radiosurgery. To overcome this, sev-
eral manufacturers have created micro-MLCs, which are add-on
Circular collimators, called cones, are used to collimate the pho- devices that attach to the accessory tray of the linac.32,33 Micro-
ton beam near the patient and reduce the geometric penumbra, MLCs utilize leaves with widths of 2 to 3 mm projected to the

49
Radiosurgical Devices

Fig. 6.8 Lateral dose falloff from Gamma Knife


and linac radiosurgery systems for a left-sided
trigeminal neuralgia treatment. Linac plans were
developed with a number of different arc sets
and Gamma Knife plans were developed with and
without some sources plugged to improve
brainstem sparing.31 (Used with permission from
IOP Publishing Ltd.)

isocenter, compared with 5 mm at isocenter for standard MLCs. underlying principles of radiosurgery in mind—immobiliza-
The difference in penumbra (80–20%) for the Brainlab m3 tion, localization, and dose gradients—clinicians can effec-
micro-MLC compared with a standard Varian 120 leaf MLC was tively select equipment that meets the needs of their clinical
demonstrated to be about 1 mm (2.5 mm compared with practices. When a new system is brought into the clinic, a
3.5 mm).33 qualified physicist should perform an end-to-end test to
Although the MLC allows complex target shapes to be treated understand the strengths and weaknesses of the proposed
with a single isocenter, several other potential issues surface in technique. Pretreatment tests validating all system align-
their implementation. One issue is the ability to accurately ments should also be conducted as part of any radiosurgery
position the leaves. With a cone, the field size is fixed and can program.
be precisely machined to an extremely high tolerance. When
creating a shape with an MLC, one must rely on the accuracy of
leaf positioning, which in general is held to a tolerance of 1 mm References
projected to the isocenter.17 When treating a 10-mm-diameter
[1] Leksell L. The stereotaxic method and radiosurgery of the brain. Acta Chir
lesion, a 1-mm discrepancy in leaf position results in a substan- Scand 1951; 102: 316–319
tial portion of the target area that could potentially be under- [2] Ma L, Sahgal A, Larson DA, et al. Impact of millimeter-level margins on pe-
dosed or normal tissue that could potentially be overdosed. ripheral normal brain sparing for gamma knife radiosurgery. Int J Radiat On-
Additionally, the output factor for small fields is highly depend- col Biol Phys 2014; 89: 206–213
[3] Schell MC, Bova FJ, Larson DA, Leavitt DD, Lutz WR, Podgorsak EB, Wu A. TG-
ent on field size and errors in dose computation could quickly
42 Report on Stereotactic External Beam Irradiation. Report of the American
grow if the field size is 10 to 20% different than planned. When Association of Physicists in Medicine Radiation Therapy Committee Task
an MLC is used to treat multiple targets from a single isocenter, Group no. 54: Stereotactic Radiosurgery. College Park, MD: American Associ-
collimator rotational accuracy also becomes an important factor ation of Physicists in Medicine; 1995
[4] Chin LS, Ma L, DiBiase S. Radiation necrosis following gamma knife surgery: a
in accurately aligning the radiation beam with the target coordi-
case-controlled comparison of treatment parameters and long-term clinical
nate. Finally, due to the undulated shaping caused by the finite follow up. J Neurosurg 2001; 94: 899–904
width of each leaf, MLCs may not be suitable for target sizes less [5] Lightstone AW, Benedict SH, Bova FJ, Solberg TD, Stern RL, American Associa-
than ~ 1 cm. tion of Physicists in Medicine Radiation Therapy Committee. Intracranial ster-
Comparing cones with MLCs, one can draw the conclusion eotactic positioning systems: report of the American Association of Physicists
in Medicine Radiation Therapy Committee Task Group no. 68. Med Phys
that small and spherical targets are likely best treated with
2005; 32: 2380–2398
cone-based radiosurgery, whereas MLCs are best utilized when [6] Schlesinger D, Xu Z, Taylor F, Yen CP, Sheehan J. Interfraction and intrafraction
treating large, irregularly shaped targets. When properly com- performance of the Gamma Knife Extend system for patient positioning and
missioned for clinical use, both of these devices can effectively immobilization. J Neurosurg 2012; 117 (Suppl): 217–224
[7] Leksell L. Stereotaxis and Radiosurgery: An Operative System. Springfield, IL:
aid the neurosurgeon and radiation oncologist in developing
Thomas Publishing; 1971
quality treatment plans. [8] Heilbrun MP, Roberts TS, Apuzzo ML, Wells TH Jr, Sabshin JK. Preliminary ex-
perience with Brown-Roberts-Wells (BRW) computerized tomography ster-
eotaxic guidance system. J Neurosurg 1983; 59: 217–222
6.8 Conclusions [9] Verellen D, Linthout N, Bel A, et al. Assessment of the uncertainties in dose
delivery of a commercial system for linac-based stereotactic radiosurgery. Int
Advances in technology are responsible for an array of radio- J Radiat Oncol Biol Phys 1999; 44: 421–433
surgical devices commercially available for performing [10] Roberts TS, Brown R. Technical and clinical aspects of CT-directed stereotaxis.
Appl Neurophysiol 1980; 43: 170–171
radiosurgery. By evaluating this equipment with the major

50
Radiosurgical Devices

[11] Saw CB, Ayyangar K, Suntharalingam N. Coordinate transformations and cal- [22] Wurm RE, Erbel S, Schwenkert I, et al. Novalis frameless image-guided non-
culation of the angular and depth parameters for a stereotactic system. Med invasive radiosurgery: initial experience. Neurosurgery 2008; 62 (Suppl):
Phys 1987; 14: 1042–1044 A11–A17, discussion A17–A18
[12] Bhatnagar JP, Novotny J, Niranjan A, et al. First year experience with newly [23] Gevaert T, Verellen D, Tournel K, et al. Setup accuracy of the Novalis ExacTrac
developed Leksell Gamma Knife Perfexion. J Med Phys 2009; 34: 141–148 6DOF system for frameless radiosurgery. Int J Radiat Oncol Biol Phys 2012;
[13] Friedman WA, Bova FJ. The University of Florida radiosurgery system. Surg 82: 1627–1635
Neurol 1989; 32: 334–342 [24] Wagner TH, Meeks SL, Bova FJ, et al. Optical tracking technology in stereotac-
[14] Meeks SL, Bova FJ, Friedman WA, Buatti JM, Mendenhall WM. Linac scal- tic radiation therapy. Med Dosim 2007; 32: 111–120
pel radiosurgery at the University of Florida. Med Dosim 1998; 23: 177– [25] Meeks SL, Bova FJ, Wagner TH, Buatti JM, Friedman WA, Foote KD. Image lo-
185 calization for frameless stereotactic radiotherapy. Int J Radiat Oncol Biol Phys
[15] Rahimian J, Chen JC, Rao AA, Girvigian MR, Miller MJ, Greathouse HE. Geo- 2000; 46: 1291–1299
metrical accuracy of the Novalis stereotactic radiosurgery system for trigemi- [26] Meeks SL, Bova FJ, Friedman WA, Buatti JM, Moore RD, Mendenhall WM.
nal neuralgia. J Neurosurg 2004; 101 (Suppl 3): 351–355 IRLED-based patient localization for linac radiosurgery. Int J Radiat Oncol Biol
[16] Chang J, Yenice KM, Narayana A, Gutin PH. Accuracy and feasibility of cone- Phys 1998; 41: 433–439
beam computed tomography for stereotactic radiosurgery setup. Med Phys [27] Bert C, Metheany KG, Doppke K, Chen GT. A phantom evaluation of a stereo-
2007; 34: 2077–2084 vision surface imaging system for radiotherapy patient setup. Med Phys
[17] Klein EE, Hanley J, Bayouth J, et al. Task Group 142, American Association of 2005; 32: 2753–2762
Physicists in Medicine. Task Group 142 report: quality assurance of medical [28] Cerviño LI, Pawlicki T, Lawson JD, Jiang SB. Frame-less and mask-less cranial
accelerators. Med Phys 2009; 36: 4197–4212 stereotactic radiosurgery: a feasibility study. Phys Med Biol 2010; 55: 1863–
[18] Tryggestad E, Christian M, Ford E, et al. Inter- and intrafraction patient posi- 1873
tioning uncertainties for intracranial radiotherapy: a study of four frameless, [29] Plowman PN, Doughty D. Stereotactic radiosurgery, X: clinical isodosimetry
thermoplastic mask-based immobilization strategies using daily cone-beam of gamma knife versus linear accelerator X-knife for pituitary and acoustic tu-
CT. Int J Radiat Oncol Biol Phys 2011; 80: 281–290 mours. Clin Oncol (R Coll Radiol) 1999; 11: 321–329
[19] Sweeney R, Bale R, Vogele M, et al. Repositioning accuracy: comparison of a [30] Andrews DW, Bednarz G, Evans JJ, Downes B. A review of 3 current radiosur-
noninvasive head holder with thermoplastic mask for fractionated radiother- gery systems. Surg Neurol 2006; 66: 559–564
apy and a case report. Int J Radiat Oncol Biol Phys 1998; 41: 475–483 [31] Ma L, Kwok Y, Chin LS, Yu C, Regine WF. Comparative analyses of linac and
[20] Ryken TC, Meeks SL, Pennington EC, et al. Initial clinical experience with Gamma Knife radiosurgery for trigeminal neuralgia treatments. Phys Med Bi-
frameless stereotactic radiosurgery: analysis of accuracy and feasibility. Int J ol 2005; 50: 5217–5227
Radiat Oncol Biol Phys 2001; 51: 1152–1158 [32] Eaton D, Twyman N, Thomas S. Commissioning a miniature multileaf collima-
[21] Kalapurakal JA, Ilahi Z, Kepka AG, et al. Repositioning accuracy with the Laiti- tor for small field radiotherapy. Med Dosim 2010; 35: 1–6
nen frame for fractionated stereotactic radiation therapy in adult and pedia- [33] Monk JE, Perks JR, Doughty D, Plowman PN. Comparison of a micro-multileaf
tric brain tumors: preliminary report. Radiology 2001; 218: 157–161 collimator with a 5-mm-leaf-width collimator for intracranial stereotactic ra-
diotherapy. Int J Radiat Oncol Biol Phys 2003; 57: 1443–1449

51
Critical Structures and Tolerance of the Central Nervous System

7 Critical Structures and Tolerance of the Central Nervous


System
Siavash Jabbari, Lijun Ma, Young K. Lee, Simon S. Lo, Eric L. Chang, Jimm Grimm, Lance Altenau, Daniel White, Vikram Udani, Steven J. Goetsch,
David Larson, and Arjun Sahgal

estimates for both conventionally fractionated radiotherapy


Key Points and single-fraction stereotactic radiosurgery (SRS). New guide-
lines have recently emerged based on dose-volume histogram
● Accumulating clinical evidence and access to volumetrically analysis (DVH) data as outlined in the recent Quantitative Anal-
accurate dose-calculation tools have allowed evidence-based ysis of Normal Tissue Effects in the Clinic (QUANTEC) report. 2,3,
critical-structure dose-tolerance estimates for single-session 4,5 In this chapter, we summarize the current knowledge of dose

stereotactic radiosurgery (SRS). limits to selected critical organs-at-risk (OARs) in the central
● Evidence supports the current dose limits to selected critical nervous system (CNS) as they pertain to single-fraction SRS.
organs-at-risk in the central nervous system specific to sin- Dose limits for conventional fractionated radiation will only be
gle-fraction SRS and for normal brain tissue, brainstem, optic briefly discussed where applicable.
pathway tissue, auditory apparatus tissue, and spinal cord.
● Options for the treatment of radiation necrosis have im-
proved substantially in the past few years.
7.2 Dose Tolerances of CNS
Structures
7.1 Introduction 7.2.1 Brain
The concept and application of partial-volume dose tolerances Brain injury induced by SRS has traditionally been categorized
was systematically investigated by Emami et al in 1991, 1 and as “acute,” referring most commonly to reversible treatment-
recommendations emerged specific to conventionally fractio- induced edema occurring during or soon after completion of
nated external beam radiotherapy (1.8 to 2.0 Gy per fraction). therapy. “Early delayed” or “subacute” refers most commonly to
Given the limited evidence available, the dose tolerances rec- reversible treatment-induced edema occurring up to 12 weeks
ommended were based mainly on expert opinion and clinical after treatment. “Late” refers most commonly to radiation
experience. Furthermore, given that three-dimensional (3D) necrosis (▶ Fig. 7.1), which is considered a potentially reversible
treatment planning and volumetric dose-calculation tools were focal or diffuse process within the radiated target volume that
not available at the time, the Emami dose-volume estimates can develop several months to years postirradiation.6
were limited to crude estimates of the volume effect by stipu- The dose-limiting phase 1 study that guides mainstream SRS
lating dose limits to a 1/3 versus 2/3 versus full organ prescription practice was conducted by the Radiation Therapy
irradiation. Oncology Group (RTOG) and published in 2000. 7 These dose
In the following decades, accumulating clinical evidence and limits were based on the incidence of irreversible grade 3
access to volumetrically accurate dose-calculation tools have (severe) or any grade 4 (life-threatening) or grade 5 (fatal) RTOG
allowed evidence-based critical-structure dose-tolerance CNS toxicity. In total, acute and chronic grade 3 CNS toxicities

Fig. 7.1 a-e A 48-year-old man with a cerebellar metastases secondary to lung cancer was treated with stereotactic radiosurgery (SRS) to 18 Gy in a
single fraction. The series of axial T1 postgadolinium magnetic resonance images from left to right represent (a) the baseline image followed by
images taken every 2 months thereafter. After what looked like an excellent response, the lesion became more variegated, enhancing, and grossly
enlarged by 6 months. The last image (e) taken at 8 months post-SRS show signs and symptoms of ataxia. The patient underwent surgery, and the
surgical pathology confirmed radiation necrosis.

52
Critical Structures and Tolerance of the Central Nervous System

(irreversible edema requiring admission for intravenous [IV] on 2 of the 78 patients (2%) developing radiation necrosis
steroids) were observed in 10% of patients, grade 4 toxicity requiring surgical intervention.15 At this time, we do not have
(radionecrosis requiring reoperations) in 10%, and grade 5 CNS sufficient data to make any firm DVH-based recommendations
toxicity (fatality were reported) in 3%. Specific to the recom- for hypofractionated SRS, but data are emerging.
mended dose limits, the incidence of grade 3 to 5 acute and The DVH metrics to predict other well-known acute effects
chronic CNS toxicity was 10% with the recommended 24 Gy (nausea, vomiting, headaches, seizures, tumor-related edema,
dose for tumors ≤ 2 cm, 20% with the recommended 18 Gy dose and mass effect symptoms) have not been established. Simi-
for tumors 2.1 to 3 cm, and 14% for tumors > 3 cm in size treated larly, neurocognitive toxicity in the form of memory decline has
to the recommended 15 Gy dose. Multivariate analysis demon- been reported following SRS, whole-brain radiotherapy (WBRT)
strated that maximum tumor diameter and higher dose were plus SRS, and WBRT alone; however, the mechanism following
associated with a significantly increased risk of grade 3, 4, or 5 SRS alone is poorly understood, as the integral brain dose for a
neurotoxicity. For tumors up to 3 cm in diameter, these doses limited number of lesions is typically subclinical as compared
have been used for years with toxicity profiles that are much with WBRT.
lower than those in the Shaw et al study,11 and they remain the With respect to life-threatening acute toxicity, it is accepted
standard protocol for SRS. For tumors > 3 cm, treatment with that complications can occur if the whole brain is exposed to
single fraction SRS can at times yield serious complications, and 10 Gy in a single fraction, whereas more modest dose-fractiona-
data are more limited. A series from Han et al reported on sin- tion regiments (4 Gy × 5, 3 Gy × 10) have been found to be
gle fraction SRS for large tumors (> 14 cm3) only.8,9 The mini- acceptable and therapeutically equivalent per randomized tri-
mum and maximum diameters and volumes were 3 and 5.7 cm als.16,17 For conventionally fractionated partial brain radiother-
and 14.1 and 49.6 cm3, respectively. They based the prescription apy, dose limits of 60 to 72 Gy (if indicated) are acceptable per
dose according to volume, with 15 to 16 Gy for tumors 14 to the QUANTEC review.17
25 cm3, 12 to 14 Gy for tumors 26 to 35 cm3, and 10 to 11 Gy for
tumors > 36 cm3. Fifteen of 80 patients (18.8%) developed unac-
ceptable CNS toxicities, RTOG CNS grade 3 to 5 (died from cere-
7.2.2 Brainstem
bral edema).8 This underscores the care required when Brainstem radiation injury can be a life-threatening complica-
selecting patients with large brain metastases for SRS, and why tion following SRS. Often, necrosis of the brainstem tissue man-
fractionated SRS is emerging as a mainstream therapy for larger ifests clinically as focal motor, sensory, or cranial nerve deficits,
tumors. general neurological demise, and can cause death, especially if
Beyond the prescribed dose and tumor diameter, several ret- the necrosis occurs in the medulla oblongata. Specific to single-
rospective studies have been reported evaluating DVH-based fraction SRS, QUANTEC recommends a maximal point dose of
predictors of radiation necrosis.3,10,11,12 Importantly, one should 12.5 Gy as safe with respect to limiting the risk of toxicity to <
note that there is significant heterogeneity in the literature 5%.2,4,18,19 A recent review of the University of California San
with respect to the definition of radiation necrosis (radio- Francisco (UCSF) experience of brainstem metastases treated
graphic necrosis vs. symptomatic radionecrosis), the indications with SRS was reported by Kased et al. Forty-two patients with
treated (arteriovenous malformations, brain metastases, etc.), brainstem metastases were treated to median dose of 16 Gy,
and the SRS technique that confounds the generalizability of and the median target volume was 0.26 cc. Four of 42 patients
these studies. Nevertheless, one consistent parameter found to developed brainstem complications post-SRS.19 All four lesions
be predictive has been the volume of nontarget brain tissue associated with complications were over 1 cc in volume, yield-
exposed to 10 or 12 Gy in a single fraction (V10, V12). 2,3,13,14 ing a freedom from complication probability of 100% for tumor
The data support keeping the V12 no greater than 5 to 15 cc volumes < 1 cc vs. 40% for tumor volumes ≥ 1 cc at 6 months,
depending on location and eloquence of tissue irradiated and 100% vs. 0%, respectively, at 1 year (p < 0.001). There was no
(brainstem, thalamus, and basal ganglia are considered higher relationship between the risk of brainstem complications and
risk regions of the brain for necrosis). The QUANTEC guidelines the prescribed dose. In addition to larger volume, multivariate
also note a rapid increase in toxicity when the volume of nor- analysis suggested significantly greater complication risks with
mal brain tissue exposed to > 12 Gy exceeds 5 to 10 cc, and they melanoma or renal cell histology. Our current practice is 15 Gy
caution more stringent limits for eloquent regions.3 Treatment in a single fraction for brainstem metastases < 2 cm, with the
of radiation necrosis will be discussed at the end of the chapter. risk of complication weighed against tumor control.
Analogous dose-volume metrics predictive of radionecrosis There are clinical data that support higher doses to the edge
are as yet to be determined in the setting of hypofractionated of the brainstem from the trigeminal neuralgia literature. 20
SRS, which refers to delivery of SRS over two to five fractions. Dose exposure has ranged from 20 Gy to even as high as 45 Gy
This practice is emerging in particular for larger tumors, as in a single fraction to a very small volume of the brainstem.21
radiobiologically by fractionating the dose we can still dose The location of exposure being on the edge of the brainstem
escalate as opposed to dose de-escalate with single-fraction surface may be the factor that explains the rare incidence of
SRS, as the tumor size increases. This is due to the protective toxicity reported in SRS for trigeminal neuralgia.20 Ultimately,
effects afforded by fractionation on the normal brain tissue. typical practice is to limit the brainstem maximum point dose
One retrospective review of 5-fraction hypofractionated SRS to 12 to 15 Gy.10,11,14
identified the volume of normal brain tissue exposed to 28.8 Gy With respect to conventionally fractionated radiotherapy, the
(single-fraction equivalent of 14 Gy, assuming α/β = 2 Gy for the dose tolerance for < 5% risk of toxicity ranges between 54 to
normal brain) as predictive of radiation necrosis, and that vol- 59 Gy according to QUANTEC guidelines, depending on if a par-
ume should be restricted to no more than 7 cc. This was based tial or the complete brainstem volume is irradiated. 4

53
Critical Structures and Tolerance of the Central Nervous System

7.2.3 Optic Pathway concluded that there was no change in hearing grade in 51% of
patients treated, and 61% maintained useful hearing post-SRS. 27
Most studies and patterns of clinical practice suggest that the A small study of 15 patients with long-term assessments noted
maximum point dose should be 8 Gy in a single fraction to the improved sensorineural hearing preservation rates of 73% ver-
optic nerves and chiasm. The fear of causing blindness has sus 20% when the marginal tumor doses were ≤ 14 Gy.29 The
propagated this conservative threshold. QUANTEC recently rec- relationship between the single fractional mean dose tolerance
ommended a maximum dose of 10 to 12 Gy to the optic nerves and the marginal (maximum) dose tolerance of the cochlea,
and chiasm as yielding an acceptable risk of optic tissue injury, and the relative predictive value of each, is unknown. For exam-
especially in patients with limited therapeutic options and/or ple, Brown et al30 have determined a dose tolerance of 5 to 6 Gy
prognosis.4,22,23,24 for the cochlea (volumetric dose), whereas other investigators
The most extensive DVH-based toxicity analysis to the optic have found mean cochlear doses in the range of 3 to 4 Gy to be
chiasm was reported by the Mayo Clinic. Stafford et al reported predictive for hearing loss in single fraction radiosurgery. 31,32
on 215 patients (218 treatments) treated with single-fraction Interestingly, Paek et el reported that patients receiving mean
SRS, and the median optic nerve point maximum dose was brainstem cochlear nucleus doses of 6.9 Gy had improved hear-
10 Gy. Within this cohort, 66% had prior surgery or irradiation; ing outcomes as compared with those receiving 11.1 Gy. In this
radiation-induced optic neuropathy (RON) occurred in less than study, the cochlear nucleus dose was a more predictive factor
2% of cases treated with 12 Gy or less.23 for SHNL loss than the cochlear dose.33 At this time, there is
The QUANTEC review of the literature specific to convention- conflicting evidence as to the most predictive structure for
ally fractionated radiation supports a maximum dose limit of SNHL post–single-fraction SRS, and specific dose-tolerance lim-
54 to 55 Gy as associated with a low risk (< 2%) of RON, particu- its, but a marginal or maximum dose of < 12 to 14 Gy to the
larly when fraction size is kept below 2 Gy.4 This is also specific cochlea is a reasonable objective, and if possible, a mean dose
to conventional fractionation proton-based therapy. Notably, of < 4 to 6 Gy may be optimal.
fraction size appears to be critical in the risk of late radiation For hypofractionated treatments, one study reported a signif-
injury to the optic apparatus, and fraction sizes of > 1.9 Gy per icant risk of hearing loss when the cochlea receive a dose of
day may be associated with higher rates of toxicity in the 60 to 8 Gy in three sessions (a single fractional dose equivalence of
70 Gy dose range.4,25 Importantly, patient-specific variables 5.4 Gy assuming α/β = 3 Gy for the cochlea).34 Such a dose is in
have been reported to increase the risk of radiation-related agreement with the mean single fractional cochlea dose toler-
injury to the optic apparatus. For example, patients with pitui- ance of 5 to 6 Gy as reported by Brown et al.30 It is significantly
tary neoplasms may have an increased sensitivity to visual lower than the conventional mean dose tolerance of 35 to 45 Gy
pathway injury based on conventional dose-fractionation liter- (as discussed below).
ature,26 and a dose limit of 46 to 48 Gy at 1.8 Gy per day has The mean dose tolerance of the cochlea to conventional radi-
been recommended by some investigators. Generally, greater ation has been estimated as < 35 to 45 Gy with varying risk and
caution should be taken for patients with significant comorbid- severity of SNHL as the end point.28
ities such as advanced age and diabetes mellitus, as some
reports have suggested a factor contributing to an increased
risk of radiation-induced visual injury,4,25 although this rela- 7.2.5 Spinal Cord
tionship requires further study and validation. Similar factors Radiation myelopathy is a rare and feared complication follow-
have not been elucidated in the SRS literature. ing both conventionally fractionated radiation and SRS. Patients
typically develop focal motor and/or sensory deficits, bowel and
bladder deficits, and pain.35 With lateralized high-dose radia-
7.2.4 Auditory Apparatus tion, a Brown-Séquard syndrome may develop and is character-
The cochlea is generally regarded to be the relevant OAR with istic. Myokymia is one of the electromyographic (EMG) findings
respect to hearing toxicity (sensorineural hearing loss [SNHL]) that can prove radiation as the etiology. Characteristic mag-
following SRS. The majority of the literature arises from the SRS netic resonance imaging (MRI) findings include intramedullary
vestibular schwannoma literature. This is explained by the T2 signal change at the treated level and focal gadolinium en-
inherent proximity of the cochlea to the tumor, and routine hancement. The time course of symptom onset (typically
hearing assessments in these patients. However, due to the sig- occurring 6 months to 3 years posttherapy) is variable and
nificant heterogeneity in terms of OAR definition (cochlea vs. shorter times to complication have been observed with SRS
volume/length of acoustic nerve vs. both), tumor size, and versus conventional radiotherapy-induced radiation myelop-
tumor location (intracanalicular vs. extracanalicular), a wide athy. At this time, there are no human data to confirm varia-
range of dose tolerances for this end point have been reported. bility in the sensitivity of cervical versus thoracic versus
In a summary of the literature, we and the QUANTEC authors lumbar spinal levels to radiation injury. However, the cauda
conclude that lower doses to the cochlea are likely associated equina is generally more tolerant to radiation than the cord
with lower risk of SNHL, and marginal cochlear doses of 12 to itself. Prior irradiation clearly increases the risk of myelopathy,
14 Gy are associated with an acceptable risk for this end point and cumulative dose limits to the cord have been suggested.
and clinical setting.27,28 However, the modeling of time-dependent repair between
A large series of vestibular schwannoma patients treated courses is not known with enough certainty to make
with SRS was reported by Kondziolka et al. Their series con- recommendations.
sisted of 162 patients, and the mean target dose was 16 Gy. Sahgal et al meticulously documented known radiation myel-
Although cochlear dose was not reported, the authors opathy cases following stereotactic single-fraction SRS and

54
Critical Structures and Tolerance of the Central Nervous System

hypofractionated SRS. In addition, analyses were done in both limiting the spinal cord dose to ≤ 135.5 Gy cumulative biologi-
unirradiated36 and previously irradiated37 patients. The most cally effective dose, with each course limited to 98 Gy or less as
recent multi-institutional DVH-based analysis comprised nine the biological effective dose.38
known cases of radiation myelopathy in patients with no his-
tory of prior radiation, and the DVH data were compared with a
multi-institutional cohort of 66 previously unirradiated spine 7.3 Treatment of Cerebral
SRS patients. A logistical regression risk model36 was developed
and recommended maximal point doses to the thecal sac were Radiation Necrosis
reported—these are summarized in ▶ Table 7.1. Given a single
Steroid therapy, typically using dexamethasone, has been the
fraction, the authors recommended no more than 12.4 Gy as a
traditional mainstay treatment for symptomatic cerebral radia-
point maximum dose to the thecal sac or cord planning organ-
tion necrosis. Other agents include hyperbaric oxygen therapy,
at-risk volume (typically a 1.5-mm expansion on the true cord
pentoxifylline, antiplatelet medications, and anticoagulant
itself).
therapies. These treatments have shown limited efficacy in this
The separate reirradiation study consisted of 19 previously
setting. Surgical resection can be considered for severely
irradiated patients who were retreated with spine SRS, and
symptomatic patients refractory to steroid treatments. The
DVH data compared with five cases of reirradiation SRS-
humanized monoclonal anti-VEGF (vascular endothelial
induced radiation myelopathy.37 This study yielded dose limits
growth factor) antibody bevacizumab has demonstrated effi-
based on the amount of prior conventional radiation and
cacy in the treatment of symptomatic radiation necrosis. In a
recommended at least a 5- to 6-month interval between prior
retrospective review of 24 patients, 23 patients demonstrated
conventional radiation and retreatment spine SRS. The recom-
clinical and radiographic improvement with this therapy, and
mendations are summarized in ▶ Table 7.2.
an average dexamethasone dose reduction of 9.4 mg was
In the setting of no prior irradiation and conventional fractio-
achieved.39 Only one patient was reported to have an adverse
nated radiation to the full circumference of the spinal cord,
grade 3 toxicity secondary to the bevacizumab (pulmonary
doses below 50 Gy appears to be associated with a very low
embolism). A small randomized trial from the MD Anderson
(< 1%) risk of radiation-induced myelopathy. A total dose of
Cancer Center (Houston, TX) was reported. Fourteen patients
60 Gy in this setting seems to be associated with a ~ 5% risk of
were randomized to placebo versus bevacizumab and those
myelopathy; dose escalation to ~ 69 Gy precipitously increases
in the bevacizumab arm had significant clinical and radiolog-
this risk to ~ 50%.5 The risk of myelopathy with conventional
ical improvements, although with higher rates of adverse
reirradiation is not well established. Based on a literature
effects.40 The use of bevacizumab needs further study before
search that identified eight reports of reirradiation myelopathy
recommendations can be made, as its underlying effect on
and 40 individual patients as control, Nieder et al recom-
the process of radiation necrosis as opposed to the edema
mended “with due caution” a time interval of at least 6 months
alone is unknown.
(ideally more than 12–24 months) between courses, and
There is some interesting recent evidence utilizing diffusion
tensor imaging that has suggested variation in brainstem fiber
radiation sensitivity to injury with conventional irradiation. 41 It
Table 7.1 Spine SRS thecal sac maximum point volume doses for pa-
is likely that with advancing functional and fiber tract–based
tients with no prior radiation exposure36
imaging, further dose-tolerance optimization and refinement
1 Fraction 2 Fractions 3 Fractions 4 Fractions 5 Fractions can be achieved based on CNS structure subunits and individu-
12.4 Gy 17.0 Gy 20.3 Gy 23.0 Gy 25.3 Gy alized patient anatomy/physiology.41 The study and under-
standing of radiation injury and risk of radiation necrosis/
myelopathy is under rapid evolution and expansion, as at this
point little is known to guide clinical decision making and risk
Table 7.2 Spine SRS thecal sac maximum point volume doses for pa- prediction.
tients with prior radiation exposure (minimum time interval between
courses of 5 months)37
Prior conventional radiotherapy dose 1 Fraction 7.4 Summary
20 Gy in 5 fractions 9 Gy Based on the above literature review and citations, a reasonable
summary of OAR dose-tolerance parameters for intracranial
30 Gy in 10 fractions 9 Gy
single-fraction radiosurgery is provided in ▶ Table 7.3. We will
37.5 Gy in 15 fractions 9 Gy continue to gain a greater appreciation of the true dose toler-
ance to the critical organs-at-risk as the field progresses, and
40 Gy in 20 fractions *
what is needed are shared databases with dosimetric data and
45 Gy in 25 fractions * actuarial outcomes to understand the actuarial incidence of
50 Gy in 5 fractions * these toxicities. We suspect that risks are lower than expected
due to modern radiation treatment–planning systems allowing
Insufficient data with single-fraction SRS to make recommendations for accurate dose calculations and greater precision in planning
given these prior radiation doses. Refer to the original publication for
and delivery technology. This may translate to favorable tumor
detailed fractionated (2 to 5 fraction) SRS spinal cord dose limits and the
control to toxicity profiles, particularly for tumors adjacent to
rules that govern these limit.
critical organs at risk.

55
Critical Structures and Tolerance of the Central Nervous System

Table 7.3 Reasonable organ-at-risk (OAR) dose-tolerance parameters for intracranial single fraction SRS
OAR Volume parameter Dose Clinical end point Note

Brain 5–10 cc 12 Gy Radionecrosis Tolerance and clinical sensitivity dependent on


location and eloquence

Brainstem Dmax 12.5 Gy Brainstem radionecrosis For brainstem metastasis, 15 Gy to lesions < 2 cm in
volume is practiced

Optic pathway Dmax 10–12 Gy Radiation-induced visual injury Refers to optic nerve and chiasm

Cochlea Marginal (Dmax) dose 12–14 Gy Sensorineural hearing loss Cochlear nucleus may also be important but further
Mean dose data required
< 4–6 Gy

Abbreviation: Dmax = maximum point dose to structure.

[17] Tsao MN, Lloyd N, Wong RK, et al. Whole brain radiotherapy for the treat-
References ment of newly diagnosed multiple brain metastases. Cochrane Database Syst
Rev 2012; (4): CD003869
[1] Emami B, Lyman J, Brown A, et al. Tolerance of normal tissue to therapeutic
[18] Pollock BE, Gorman DA, Brown PD. Radiosurgery for arteriovenous malforma-
irradiation. Int J Radiat Oncol Biol Phys 1991; 21: 109–122
tions of the basal ganglia, thalamus, and brainstem. J Neurosurg 2004; 100:
[2] Levegrün S, Hof H, Essig M, Schlegel W, Debus J. Radiation-induced changes
210–214
of brain tissue after radiosurgery in patients with arteriovenous malforma-
[19] Kased N, Huang K, Nakamura JL, et al. Gamma knife radiosurgery for
tions: correlation with dose distribution parameters. Int J Radiat Oncol Biol
brainstem metastases: the UCSF experience. J Neurooncol 2008; 86: 195–
Phys 2004; 59: 796–808
205
[3] Lawrence YR, Li XA, el Naqa I, et al. Radiation dose-volume effects in the
[20] Smith ZA, Gorgulho AA, Bezrukiy N, et al. Dedicated linear accelerator radio-
brain. Int J Radiat Oncol Biol Phys 2010; 76 (Suppl): S20–S27
surgery for trigeminal neuralgia: a single-center experience in 179 patients
[4] Mayo C, Martel MK, Marks LB, Flickinger J, Nam J, Kirkpatrick J. Radiation
with varied dose prescriptions and treatment plans. Int J Radiat Oncol Biol
dose-volume effects of optic nerves and chiasm. Int J Radiat Oncol Biol Phys
Phys 2011; 81: 225–231
2010; 76 (Suppl): S28–S35
[21] Xue J, Goldman HW, Grimm J, et al. Dose-volume effects on brainstem dose
[5] Kirkpatrick JP, van der Kogel AJ, Schultheiss TE. Radiation dose-volume ef-
tolerance in radiosurgery. J Neurosurg 2012; 117 (Suppl): 189–196
fects in the spinal cord. Int J Radiat Oncol Biol Phys 2010; 76 (Suppl):
[22] Tishler RB, Loeffler JS, Lunsford LD, et al. Tolerance of cranial nerves of the
S42–S49
cavernous sinus to radiosurgery. Int J Radiat Oncol Biol Phys 1993; 27:
[6] Chao ST, Ahluwalia MS, Barnett GH, et al. Challenges with the diagnosis and
215–221
treatment of cerebral radiation necrosis. Int J Radiat Oncol Biol Phys 2013;
[23] Stafford SL, Pollock BE, Leavitt JA, et al. A study on the radiation tolerance of
87: 449–457
the optic nerves and chiasm after stereotactic radiosurgery. Int J Radiat Oncol
[7] Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treatment of
Biol Phys 2003; 55: 1177–1181
recurrent previously irradiated primary brain tumors and brain metasta-
[24] Leber KA, Berglöff J, Pendl G. Dose-response tolerance of the visual pathways
ses: final report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys
and cranial nerves of the cavernous sinus to stereotactic radiosurgery. J Neu-
2000; 47: 291–298
rosurg 1998; 88: 43–50
[8] Han JH, Kim DG, Chung HT, Paek SH, Park CK, Jung HW. Radiosurgery for large
[25] Parsons JT, Bova FJ, Fitzgerald CR, Mendenhall WM, Million RR. Radiation op-
brain metastases. Int J Radiat Oncol Biol Phys 2012; 83: 113–120
tic neuropathy after megavoltage external-beam irradiation: analysis of
[9] Han JH, Kim DG, Kim CY, Chung HT, Jung HW. Stereotactic radiosurgery for
time-dose factors. Int J Radiat Oncol Biol Phys 1994; 30: 755–763
large brain metastases. Prog Neurol Surg 2012; 25: 248–260
[26] Mackley HB, Reddy CA, Lee SY, et al. Intensity-modulated radiotherapy for pi-
[10] Flickinger JC, Kondziolka D, Pollock BE, Maitz AH, Lunsford LD. Complications
tuitary adenomas: the preliminary report of the Cleveland Clinic experience.
from arteriovenous malformation radiosurgery: multivariate analysis and
Int J Radiat Oncol Biol Phys 2007; 67: 232–239
risk modeling. Int J Radiat Oncol Biol Phys 1997; 38: 485–490
[27] Kondziolka D, Lunsford LD, McLaughlin MR, Flickinger JC. Long-term out-
[11] Flickinger JC, Kondziolka D, Maitz AH, Lunsford LD. Analysis of neurological
comes after radiosurgery for acoustic neuromas. N Engl J Med 1998; 339:
sequelae from radiosurgery of arteriovenous malformations: how location af-
1426–1433
fects outcome. Int J Radiat Oncol Biol Phys 1998; 40: 273–278
[28] Bhandare N, Jackson A, Eisbruch A, et al. Radiation therapy and hearing loss.
[12] Chin LS, Ma L, DiBiase S. Radiation necrosis following gamma knife surgery: a
Int J Radiat Oncol Biol Phys 2010; 76 (Suppl): S50–S57
case-controlled comparison of treatment parameters and long-term clinical
[29] Niranjan A, Lunsford LD, Flickinger JC, Maitz A, Kondziolka D. Dose reduction
follow up. J Neurosurg 2001; 94: 899–904
improves hearing preservation rates after intracanalicular acoustic tumor ra-
[13] Blanchard N, Bernier V, Anxionnat R, et al. [Radiosurgery of cerebral arte-
diosurgery. Neurosurgery 1999; 45: 753–762, discussion 762–765
riovenous malformations: a prescription algorithm]. Cancer Radiother
[30] Brown M, Ruckenstein M, Bigelow D, et al. Predictors of hearing loss after
2009; 13: 1–10
gamma knife radiosurgery for vestibular schwannomas: age, cochlear
[14] Flickinger JC, Kondziolka D, Lunsford LD, et al. Arteriovenous Malformation
dose, and tumor coverage. Neurosurgery 2011; 69: 605–613, discussion
Radiosurgery Study Group. Development of a model to predict permanent
613–614
symptomatic postradiosurgery injury for arteriovenous malformation pa-
[31] Baschnagel AM, Chen PY, Bojrab D, et al. Hearing preservation in patients
tients. Int J Radiat Oncol Biol Phys 2000; 46: 1143–1148
with vestibular schwannoma treated with Gamma Knife surgery. J Neurosurg
[15] Inoue HK, Sato H, Seto K, et al. Five-fraction CyberKnife radiotherapy for large
2013; 118: 571–578
brain metastases in critical areas: impact on the surrounding brain volumes
[32] Yomo S, Carron R, Thomassin JM, Roche PH, Régis J. Longitudinal analysis of
circumscribed with a single dose equivalent of 14 Gy (V14) to avoid radiation
hearing before and after radiosurgery for vestibular schwannoma. J Neuro-
necrosis. J Radiat Res (Tokyo) 2014; 55: 334–342
surg 2012; 117: 877–885
[16] Borgelt B, Gelber R, Larson M, Hendrickson F, Griffin T, Roth R. Ultra-rapid
[33] Paek SH, Chung HT, Jeong SS, et al. Hearing preservation after gamma
high dose irradiation schedules for the palliation of brain metastases: final
knife stereotactic radiosurgery of vestibular schwannoma. Cancer 2005;
results of the first two studies by the Radiation Therapy Oncology Group. Int J
104: 580–590
Radiat Oncol Biol Phys 1981; 7: 1633–1638

56
Critical Structures and Tolerance of the Central Nervous System

[34] Hayden Gephart MG, Hansasuta A, Balise RR, et al. Cochlea radiation dose [38] Nieder C, Grosu AL, Andratschke NH, Molls M. Proposal of human spinal cord
correlates with hearing loss after stereotactic radiosurgery of vestibular reirradiation dose based on collection of data from 40 patients. Int J Radiat
schwannoma. World Neurosurg 2013; 80: 359–363 Oncol Biol Phys 2005; 61: 851–855
[35] Schultheiss TE, Kun LE, Ang KK, Stephens LC. Radiation response of the cen- [39] Sadraei NH, Dahiya S, Chao ST, et al. Treatment of cerebral radiation necrosis
tral nervous system. Int J Radiat Oncol Biol Phys 1995; 31: 1093–1112 with bevacizumab: The Cleveland Clinic experience. Am J Clin Oncol 2013: 24
[36] Sahgal A, Weinberg V, Ma L, et al. Probabilities of radiation myelopathy spe- [40] Levin VA, Bidaut L, Hou P, et al. Randomized double-blind placebo-controlled
cific to stereotactic body radiation therapy to guide safe practice. Int J Radiat trial of bevacizumab therapy for radiation necrosis of the central nervous sys-
Oncol Biol Phys 2013; 85: 341–347 tem. Int J Radiat Oncol Biol Phys 2011; 79: 1487–1495
[37] Sahgal A, Ma L, Weinberg V, et al. Reirradiation human spinal cord tolerance [41] Uh J, Merchant TE, Li Y, et al. Differences in brainstem fiber tract response to
for stereotactic body radiotherapy. Int J Radiat Oncol Biol Phys 2012; 82: radiation: a longitudinal diffusion tensor imaging study. Int J Radiat Oncol Bi-
107–116 ol Phys 2013; 86: 292–297

57
The Neuropathology of Radiosurgery

8 The Neuropathology of Radiosurgery


György Szeifert, Douglas Kondziolka, Marc Levivier, José Lorenzoni, and L. Dade Lunsford

sharply delimited lesion can be made at any desired site in the


Key Points central nervous system.”1 The basic histopathological lesion
(▶ Fig. 8.1) created by high-energy ionizing radiation in neural
● Three main histological types of gamma radiolesions that are tissue is a coagulation necrosis within the target volume, sur-
acute, subacute, and chronic variants of tissue reactions have rounded by a distinct boundary between the necrosis and adja-
been recognized in different neoplasms irrespective of their cent normal structures, according to the sharp radiation falloff.2
ontogenetic nature. Whereas target necrosis was the goal with maximum doses
● No significant relation has been found between the morpho- above 100 Gy, almost all human tumors are irradiated at lower,
logical characteristics of the evoked histological reaction and more cytostatic doses. Early lesions appeared in the spinal cord
the time interval elapsed after stereotactic radiosurgery. This following irradiation with doses of 400 and 200 Gy on the third
relative time and environment autonomy of the developed and ninth day, respectively. They were sharply defined and had
pathological lesions with a similar histological picture in about the same width as the beam. In the cerebral hemispheres,
different neoplasms suggests a vascular mechanism and/or a the earliest alterations were observed 14 days after irradiation
genetically directed origin presumably induced by the with 200 Gy, and the changes between 2 and 8 weeks were sim-
ionizing energy of high-dose photon irradiation. ilar. A striking feature of these lesions was the paucity of cellu-
● Endothelial damage of the vessel wall appears to be among lar reaction at the periphery of necrosis: The tissue further
the earliest morphological changes following irradiation away from the lesion appeared undamaged. However, irradia-
accompanied by an increased apoptotic activity in the tion with ~ 400 Gy produced a very different lesion. The irradi-
surrounding brain tissue. ated hemisphere of the animal was markedly swollen and the
● These pathophysiological processes might contribute to midline structures were displaced. The affected region sur-
stromal vessel occlusion followed by necrosis of tumor rounding the necrosis was at least the size of the necrotic area
parenchyma, and could boost cellular death evoked by the itself. Other parts of the brain showed no pathological changes.
radiation effect of stereotactic radiosurgery. These were the early experimental observations after single
high-dose irradiation. This phase of postirradiation reaction
was called the necrotic stage. The next period of postradiosur-
8.1 Introduction gery changes was the resorption stage. This stage was character-
ized by the resorption of cellular debris, macrophage reaction,
In our study to explore short- and long-term tissue reactions
and the beginning of glial scar formation around the lesion.
evoked by single high-dose irradiation in different human brain
These changes were observed in goats between 18 and 28
tumors and experimental animals following Gamma Knife
months following high-dose single-session proton irradiation.
radiosurgery, histopathological investigations demonstrated
The late stage was characterized histologically by prominent
that Gamma Knife radiosurgery generates degenerative and
glial scar formation surrounding a cavity.1–5
proliferative tissue reactions in the parenchyma, stroma, and
Radiosurgery, a term first coined by Professor Lars Leksell, has
vessels of the irradiated target volumes. Three main histological
become a successful treatment modality in the neurosurgical
types of gamma radiolesions—acute, subacute, and chronic var-
arsenal over the past four decades.6,7 Since 1967, when the first
iants of tissue reactions—were recognized in different neo-
patient with a craniopharyngioma was treated in Stockholm with
plasms irrespective of their oncogenetic nature. There was no
the prototype Gamma Knife, more than one million patients
significant relation between morphological characteristics of
worldwide have undergone Gamma Knife radiosurgery using sev-
the evoked histological reaction and the time interval elapsed
eral different models. Patients have also been treated with other
after Gamma Knife surgery. The relatively equivalent temporal
radiosurgical technologies such as modified linear accelerators or
and histological development of the pathological lesions
charged particle devices. Because the treatment indications and
regardless of the brain location or the tumor cell of origin sug-
the number of treated patients have been increasing exponen-
gests a vascular mechanism and/or a genetically mediated res-
tially, it is important to evaluate the pathological processes that
ponse resulting from high-dose photon-ionizing energy.
result from radiosurgery. Such an analysis provides a better
Experimental studies indicate that endothelial damage of blood
understanding of the radiobiology and the pathophysiological
vessel walls appears to be among the earliest morphological
mechanisms that lead to beneficial therapeutic effects as well as
changes; this is accompanied by an increased apoptotic activity
the undesired side effects of radiosurgery. In addition to further
in the surrounding brain tissue. These pathophysiological proc-
technical advances in the method of dose delivery, the future of
esses might contribute to stromal vessel occlusion followed by
radiosurgery rests on a better understanding of the biological
cell death of tumor parenchyma. The vascular effect then boosts
basis of radiation, which will enable treatment of new disorders.8
the cellular death evoked by stereotactic radiosurgery (SRS).
Now that we have amassed a great deal of clinical experience in
radiosurgery, it is time to review the pathological fundamentals
of the effect of single high-dose irradiation.2,9,10
8.2 Background The term radiosurgery refers to the application of ionizing
In 1958, Larsson et al published in Nature the results of animal radiation energy in experimental biology or clinical medicine
experiments that indicated “with high-energy protons a for the precise and complete destruction of chosen target

58
The Neuropathology of Radiosurgery

Fig. 8.1 Sharply demarcated experimental gamma radiolesion (i.e., coagulation necrosis) towards surrounding brain tissue in rat brain 6 months after
160 Gy irradiation. (a) Hematoxylin and eosin (H&E), × 100. (b) Masson’s trichrome for fibrin stain, × 100. (c) A complete cavity developed 1 year after
single high-dose irradiation: H&E, × 100.

structures containing either healthy or pathological tissue, undergone previous Leksell Gamma Knife (LGK) radiosurgery.
without significant concomitant or late radiation damage to These patients had radiological and clinical progression that
adjacent tissues.11 Therefore, the goal of radiopathology should warranted removal of the lesion via craniotomy. The tumors
be to study the short- and long-term effects of high-dose studied included metastases, astrocytomas, meningiomas, ves-
focused irradiation on neural tissue and to examine its patho- tibular schwannomas, and hemangioblastomas. Radiosurgery
logical effect using histological, tissue culture, and biological– was performed using the LGK model C (Elekta Instruments AB,
biochemical methods. Radiopathology includes an analysis Stockholm, Sweden). Dose planning was based on magnetic res-
using ultrastructural studies of the tissue, cellular, genetic, and onance imaging (MRI) and computed tomography (CT). Treated
molecular changes in both the human body and in experimen- volumes ranged between 266 and 25.600 mm3 (median:
tal animals. In addition, studies using cell lines and other in 4.700 mm3). Tumors received 12 to 20 Gy as marginal dose
vitro experiments aid our understanding of the effects of ioniz- (median: 16 Gy) at 30 to 60% isodose line (median: 50%), with
ing radiation delivered from radiosurgical devices. A better 24 to 40 Gy maximal dose (median: 32 Gy).
understanding of radiobiological processes will enhance the Histopathological investigations were performed on surgical
quality of radiosurgery and stimulate the development of new pathology materials. Resected specimens were fixed in 10%
applications for the benefit of patients in need. neutral buffered formaldehyde, processed routinely, and
embedded in paraffin. Besides routine hematoxylin and eosin
and Masson’s trichrome staining, immunohistochemical reac-
8.3 Materials and Methods tions were carried out for glial fibrillary acidic protein (GFAP),
Histopathological investigations were performed on surgically vimentin, S100, neurofilament, synaptophysin, EMA, pankera-
resected brain tumor specimens from patients who had tin, CK7, CK20, CAM5.2, CD3, CD20, CD31, and CD68 (PGM1)

59
The Neuropathology of Radiosurgery

Fig. 8.2 Steps of experimental Gamma Knife radiosurgery. (a) The rat stereotactic frame; (b) placement of the Leksell Model G base ring around the
rat; (c) the rat in the focus of the C unit gamma knife using a 4 mm isocenter. (d) A laser is used to mark the target for beam intersection.

antigens to characterize the phenotypic nature of the tumor develop in an early or delayed manner, but chronic-type tissue
cells and the reactive cell population around/or infiltrating neo- response evolves only in a delayed fashion.12
plastic tissues. Ki67 and p53 reactions were used to assess pro- Histopathological changes attributable to radiosurgery were
liferative activity of tumor cells. Biotin–streptavidin–peroxidase observed within the tumor parenchyma, connective tissue stro-
complex methods were performed according to standard proto- ma, and vessels. These changes were either degenerative or
cols on 5-µm paraffin sections. proliferative. Degenerative alterations occurred mostly in the
Rat experimental radiosurgery was carried out according to parenchyma of different tumors, whereas proliferative proc-
standard protocols using a small animal stereotactic radiosur- esses were realized first in the connective tissue stroma and the
gery frame specific for Gamma Knife (▶ Fig. 8.2). Sacrificed ani- blood vessels within the neoplasm. The basic histopathological
mals were fixed via a perfusion procedure with 10% neutral alteration attributable to the ionizing energy of high-dose radi-
buffered formaldehyde. Removed whole-brain basal surfaces ation was recognized as a well-circumscribed lesion with sharp
were correlated to isodose curves (▶ Fig. 8.3a,b), then cut for demarcation from adjacent tissues that corresponded to the
5-mm slices marking the contralateral, nonirradiated side steep radiation falloff of the delivered dose (▶ Fig. 8.4a–c).
(▶ Fig. 8.3c,d), and processed for histology as described above. Regarding the histological and cellular composition of these
very high-dose gamma radiosurgical lesions, three main types
were realized. In the acute-type reaction, coagulation necrosis
8.4 Results constituted the center with a network of acidophilic fibrinoid
The morphological appearance of various lesions suggestive of material and amorphous homogeneous tissue debris
radiation effect in different tumors was not found to correlate (▶ Fig. 8.4d). No cellular reaction or scanty basophilic hyperchro-
with the histopathological type of the irradiated neoplasm nor matic apoptotic cells—characterized by nuclear fragmentation
with the time interval between radiosurgery and craniotomy. and pyknosis intermingled with scattered polymorphonuclear
The temporal development of radiation responses included leukocytes and some dilated postcapillary venules—surrounded
immediate (milliseconds to hours), early (days to weeks), and this necrotic core. There was no prominent macrophage or lym-
delayed (months to years after exposure) effects. However, the phocytic infiltration, reactive gliosis, or scar tissue formation. A
morphological and clinical appearance can be described as similar histological picture of the acute-type lesion could be
acute, subacute, or chronic types. The acute reaction may observed either at an early or a delayed time interval after

60
The Neuropathology of Radiosurgery

Fig. 8.3 Pathological processing of irradiated rat brains. (a) The rat brain prior to fixation; (b) marking the lesion site for histology; (c) preparation of
the specimen. (d) Sections are made of the brain to process for histopathology.

radiosurgery. These parenchymal changes were accompanied periphery of the central necrotic region. A reactive gliotic scar
with alterations of stromal vessels around the necrotic core mantle formed by astrocytic elements and abundant glial fila-
characterized by endothelial destruction, fibrinoid necrosis, ments constituted the outer border of the subacute radiosurgi-
undulation of the internal elastic membrane with vacuolation, cal response, as confirmed by GFAP immune positivity.
and accumulation of eosinophilic material (transudation) in the The third, chronic-type or “end-stage” pathological reaction,
vessel wall. was detected several years after radiosurgery. The central part
The second type of gamma radiosurgical lesion was termed of the radiolesion was replaced by hypocellular scar tissue with
as subacute. These pathological responses were observed sev- degenerative changes that included hyaline deposition and/or
eral months to years after radiosurgery. The main histological focal calcification that was demarcated sharply from the
feature of these changes was an inflammatory tissue response. surrounding tumor tissue (▶ Fig. 8.6a). Scattered fibrocytes,
The central coagulation necrotic core was surrounded by a mac- fibroblasts, and sporadic focal lymphocytic infiltration were
rophage rim (▶ Fig. 8.5a). These macrophages revealed phago- identified surrounding dense collagen bundles (▶ Fig. 8.6b).
cytotic activity and mainly CD68 (PGM1), but sometimes CD31 Immunohistochemical markers for tumor antigens such as
immunohistochemical reactivity as well. The necrosis in this S100 or neurofilament reactions demonstrated considerable
targeted volume was usually circumscribed, homogeneous, and decrease in these areas. Advanced postirradiation vasculopathy
sharply demarcated from surrounding remaining tumor tissue with subendothelial spindle-shaped cell proliferation and hya-
as compared with other adjacent tissue changes outside of line degeneration resulted in subtotal or even complete luminal
the target volume irradiation. These changes included an irreg- obliteration of blood vessels within the high-dose volume.
ular multifocal appearance with entrapped tumor islands Acute-type histological changes were found at 2 to 17
(▶ Fig. 8.5b). A granulation tissue zone extended beyond the months (median: 11 months) after radiosurgery. The subacute-
macrophage layer and contained abundant small vessels, capil- type histological response developed at 4 to 59 months
laries, arterioles, and venules accompanied by inflammatory (median: 16 months) after radiosurgery. The chronic-type tis-
cells, fibrocytes, and fibroblasts expressing vimentin positivity. sue response was observed 18 to 82 months (median: 32
Postirradiation vasculopathy was also observed at the months) following radiosurgery.

61
The Neuropathology of Radiosurgery

Fig. 8.4 Acute-type tissue responses following


vestibular schwannoma radiosurgery. (a) Gam-
maPlan treatment planning of the tumor at the
time of radiosurgery. (b) Characteristic magnetic
resonance signal changes 2.5 months following
irradiation. (c) Sharply demarcated tissue necrosis
at the boundary of the irradiation (hematoxylin
and eosin [H&E], × 200). (d) Necrotic tissue debris
and scattered pyknotic, apoptotic cells in the
center of the irradiated volume, 2.5 months
following radiosurgery (H&E, × 300).

Experimental radiosurgical responses in animals demon- The experimental basis for brain tumor radiosurgery has
strated endothelial destruction in the irradiated vessel wall been well described and provides evidence for the effectiveness
(▶ Fig. 8.7) and enhanced apoptotic activity in the surrounding of high-dose irradiation.17–23 Human pathological case reports
brain tissue at an early stage (▶ Fig. 8.8). Late postirradiation and reviews support the experimental results.24–31 Our purpose
vasculopathy with narrowed channels lumina and even ische- here was to evaluate the effect of radiosurgery on experimental
mic vessel occlusion occurred ~ 1 year after radiosurgery animals and treated human brain tumors that required delayed
(▶ Fig. 8.9). resection. These pathological data include an evaluation of vari-
ous different tumor types and a review of the histology at vari-
ous intervals after irradiation.
8.5 Discussion In human brain tumors, the morphology of the radiosurgical
lesion appeared to be similar to that found in experimental ani-
The continuous development of stereotactic radiosurgery tech- mals. Our studies confirmed that ionizing energy from high-
niques and treatment of new disorders warrant further radio- dose irradiation evoked either degenerative or proliferative his-
biological and pathological research to improve therapeutic topathological changes in the investigated brain tumors. Degen-
interventions and to reduce undesired side effects. The original erative changes including necrosis and apoptosis occurred in
intention of radiosurgery was to treat functional disorders,6,7,13 the parenchyma of the tumor and resulted in cell destruction.
but soon after its introduction arteriovenous malformations In contrast, fibrinoid and calcium deposition generally
(AVMs)14 and brain tumors15,16 in high-risk surgical regions appeared in the connective tissue stroma and blood vessel wall.
became major targets for the method. Proliferative processes such as granulation tissue formation,

62
The Neuropathology of Radiosurgery

Fig. 8.5 (a) Subacute-type tissue reaction. Central necrosis surrounded


by an inflammatory cell reaction consisting of mostly macrophages in a
renal cell carcinoma metastasis 7 months after Gamma Knife radio- Fig. 8.6 (a) Chronic-type infiltration with prominent lymphocytic
surgery (hematoxylin and eosin [H&E], × 300). (b) Irregular tumor component in a metastatic tumor 12 months following radiosurgery
necrosis outside of the irradiated target volume (H&E, × 200). (hematoxylin and eosin [H&E], × 200). (b) End-stage lesion consisting
of hypocellular scar tissue with hyaline degeneration and dystrophic
calcification sharply demarcated towards brain tissue in a metastatic cc
15 months after radiosurgery (H&E, × 100).

inflammation, macrophage invasion, fibrocyte–fibroblast pro-


liferation, and scar tissue production were found in the stroma
and vasculature of tumors. Such changes appear to provide a noted in a delayed fashion and indicate that these changes are
scavenger function that facilitates the replacement of the end stage.
destroyed neoplastic tissue (necrotic debris) with scar tissue. Radiosurgery destroys or inactivates the targeted neoplastic
Scar tissue itself consists of hyaline degenerated collagen bun- cell proliferation either by a dose-related direct, early cytotoxic
dles and glial fibrils that have a certain propensity for contrac- effect (coagulation necrosis, apoptosis) or by late vascular ische-
tion. This contraction of scar tissue might play a role in the mic changes. It has been suggested that the radiobiological
volumetric regression of the tumor as noted frequently on fol- mechanism of radiosurgery on benign tumors is a combination
low-up imaging studies. of both cytotoxic and vascular effects.32 The direct cellular influ-
Three main histopathological responses of radiosurgery were ence may be the consequence of lysosomal membrane injury
delineated. Acute-type gamma responses were characterized by releasing various hydrolytic enzymes with the cytoplasm,
tissue necrotic changes that developed either early or at a which begins a cascade of events resulting in cellular necrosis.
delayed time interval. Subacute-type responses demonstrated In addition, indirect DNA damage leading to strand breaks
resorptive activity either early or in a delayed manner. Chronic caused by the high dose of radiation delivered to the target vol-
responses showed evidence of attempted tissue repair, but only ume by the intersection of the converging photon beams. This
at the delayed stage. These changes seem to progress consecu- high dose to a small target volume leads to cell death at the
tively over time without any significant relationship between start of the next cell cycle (apoptosis, i.e., programmed cell
tumor histological type or time interval after irradiation. The death). It is for this reason that rapidly proliferating tumors
only consistent observation was that chronic-type lesions were with high cellular mitotic rates, such as metastases or

63
The Neuropathology of Radiosurgery

Fig. 8.7 Destruction of endothelial cell layer 2 months after 15 Gy irradiation (Factor-VIII immune reaction, × 200).

Fig. 8.8 Enhanced apoptotic activity in the brain tissue 2 months after 15 Gy irradiation (Caspase-3 immune reaction, × 200).

malignant gliomas, react earlier to radiosurgery than do slowly response rate of radiosurgery. Previous publications described
growing benign tumors with lower proliferation rates. Nonneo- effects such as decreased tumor cell population, increased
plastic lesions that are not proliferating, such as AVMs, react fibrous stroma, central necrosis, macrophage rim development,
with an even more prolonged latency period. Late vascular lymphocytic infiltration, vascular changes, and reactive astrocy-
effects of radiosurgery likely are mediated by the progressive tosis.25,26,36,37 Jagannathan et al concluded that radiation
vascular reaction. Radiosurgical doses used in clinical practice necrosis and tumor radiation resistance were the most common
usually do not affect normal high-flow brain vessels.33 However, causes precipitating a need for surgical resection after radiosur-
the abnormal pathological vessels of tumors or vascular malfor- gery in patients with brain metastasis.38 They have found that
mations seems to have a higher sensitivity to the energy of survival of patients with viable tumors was significantly lower
high-dose photon radiation delivered by radiosurgery. The dis- than survivals in patients in whom only necrosis was seen.
crepancy in this vascular response provides a pathophysiologi- Kano et al reported that a shorter interval from stereotactic
cal basis for selective radiosurgical irradiation in both tumors radiosurgery to resection was associated with a higher rate of
and nonneoplastic brain targets such as AVMs.34,35 tumor recurrence.39 If a resection was necessary within two
To date, most histopathological studies evaluated morpholog- months after radiosurgery, the pathology always showed viable
ical changes in cerebral tumors that required delayed resection tumor.40
after radiosurgery. However, both postoperative and or delayed Histopathological and immunohistochemical investigations
autopsy studies are relatively rare because of the high clinical suggest that radiosurgery evokes necrotic changes in brain

64
The Neuropathology of Radiosurgery

Fig. 8.9 Narrowed vessel lumina and ischemic stroke in the frontal lobe 1 year after 70 Gy irradiation (hematoxylin and eosin, × 100).

metastases and their vascular stroma accompanied by a ster-


ile inflammatory reaction.41 Three histological types of tissue
8.6 Conclusions
responses were observed in metastases as well. The tissue From the results of the study described here, we propose a
responses disclosed a certain temporal development, but did histopathological classification system for tissue responses
not have a significant correlation with the histological origin observed in patients who had brain tumors that progressed
of the tumors. The inflammatory reaction had a prominent after radiosurgery and required subsequent craniotomy. We
role in the local tumor control process. Tumors that required observed that single high-dose irradiation evoked three forms
craniotomy for recurrent disease in < 5 months after radiosur- of histological reactions: acute, subacute, and chronic types.
gery were found to have minimal-to-no inflammatory reac- The specimens were composed initially of coagulation necrosis,
tions. Neoplasms that required resection > 5 months after usually accompanied by an inflammatory response and vascular
irradiation demonstrated a moderate-to-intense inflamma- changes followed by scar tissue formation in a variety of cere-
tory response. Immunohistochemistry revealed a predomi- bral neoplasms with different histological subtypes. The time
nance of CD68-positive macrophages in the subacute-type course of these histopathological reactions suggests that the
lesions and CD3-positive T lymphocytes in the chronic-type response gradually evolves from the acute-type, to the suba-
tissue reactions. The sparse inflammatory reactivity in less cute-type, to the chronic-type morphologies. We also wit-
responsive metastatic neoplasms (those requiring cranioto- nessed postirradiation vasculopathy to varying extents in the
my < 5 months) raises the question of either an inherent tumors analyzed. A more favorable tumor response to radiosur-
delay in the response of these tumors to radiosurgery or pos- gery was connected with a brisker CD68-positive and CD3-pos-
sibly an impairment of the immune system in these patients. itive cellular reactions; however, given the paucity of data
The pathophysiological mechanism by which radiosurgery further studies are required.
evokes an inflammatory response in brain metastases may The similar morphological appearance of radiosurgery effects
originate from the treated tumor cells or the vascular endo- in different tumors and different patients suggests that the
thelium. For example, ionizing radiation may damage the evoked histopathological reaction is likely related to the biolog-
blood–tumor barrier, permitting cellular elements in the ical effect of radiosurgery. We believe that these tissue
blood, especially leukocytes, to traverse the vessel wall and responses represent both direct cellular effects and vascular
infiltrate the surrounding tumor parenchyma. This theory is mechanisms induced by single high-dose irradiation. The acute
supported by the histopathological observation that many of effect indicates direct cellular damage influenced by cytokines
these inflammatory cell infiltrates appeared to originate and released at the time of injury. Subacute and chronic effects
propagate in perivascular spaces of the tumor and surround- might be mediated by the inflammatory cascade triggered by
ing brain tissue. It is also possible that focused irradiation the ionizing energy of radiosurgery. Our human pathological
stimulates the immune system of the brain, similar to what observations are in accordance with the results of genetic and
has been observed in other pathological conditions such as pharmacological studies that revealed the critical role of both
AVMs after radiosurgery.32,34,35,42 The original concept of radiation-induced microvasculature and tumor cell damage,
immune privilege of the central nervous system already has constituting the fundamental elements of the tumor response
been modified, and a variety of studies have demonstrated to radiation.44 One goal of radiobiological research in the future
that both afferent and efferent immune pathways are present could be to focus on stimulation of these pathways to enhance
in the normal central nervous system. 43 efficacy and selectivity of radiosurgery. 45

65
The Neuropathology of Radiosurgery

[25] Thompson BG, Coffey RJ, Flickinger JC, Lunsford LD. Stereotactic radiosurgery
References of small intracranial tumors: neuropathological correlation in three patients.
Surg Neurol 1990; 33: 96–104
[1] Larsson B, Leksell L, Rexed B, Sourander P, Mair W, Andersson B. The high-en-
[26] Hirato M, Hirato J, Zama A, et al. Radiobiological effects of gamma knife radio-
ergy proton beam as a neurosurgical tool. Nature 1958; 182: 1222–1223
surgery on brain tumors studied in autopsy and surgical specimens. Stereo-
[2] Szeifert GT, Kondziolka D, Lunsford LD, et al. Introduction: the contribution
tact Funct Neurosurg 1996; 66 (Suppl 1): 4–16
of pathology to radiosurgery. Prog Neurol Surg 2007; 20: 1–15
[27] Szeifert GT, Massager N, Brotchi J, Levivier M. Morphological redifferentiation
[3] Leksell L, Larsson B, Andersson B, Rexed B, Sourander P, Mair W. Lesions in
in a malignant astrocytic tumor after gamma knife radiosurgery. J Neurosurg
the depth of the brain produced by a beam of high energy protons. Acta Ra-
2002; 97 (Suppl): 627–630
diol 1960; 54: 251–264
[28] Szeifert GT, Salmon I, David P, et al. Tumor control and growth in a patient
[4] Andersson B, Larsson B, Leksell L, et al. Histopathology of late local radiole-
with two cerebral metastases treated with the Leksell Gamma Knife. In:
sions in the goat brain. Acta Radiol Ther Phys Biol 1970; 9: 385–394
Kondziolka D, ed. Radiosurgery. Vol. 4. Basel, Switzerland: Karger;
[5] Wennerstrand J, Ungerstedt U. Cerebral radiosurgery. II. An anatomical study
2002:152–161
of gamma radiolesions. Acta Chir Scand 1970; 136: 133–137
[29] Kwon Y, Khang SK, Kim CJ, Lee DJ, Lee JK, Kwun BD. Radiologic and histopa-
[6] Leksell L. The stereotaxic method and radiosurgery of the brain. Acta Chir
thologic changes after Gamma Knife radiosurgery for acoustic schwannoma.
Scand 1951; 102: 316–319
Stereotact Funct Neurosurg 1999; 72 (Suppl 1): 2–10
[7] Leksell L. Cerebral radiosurgery. I. Gammathalanotomy in two cases of in-
[30] Nielsen SL, Kjellberg RN, Asbury AK, Koehler AM. Neuropathologic effects of
tractable pain. Acta Chir Scand 1968; 134: 585–595
proton-beam irradiation in man. I. Dose-response relationships after treat-
[8] Kondziolka D, Lunsford LD, Witt TC, Flickinger JC. The future of radiosurgery:
ment of intracranial neoplasms. Acta Neuropathol 1972; 20: 348–356
radiobiology, technology, and applications. Surg Neurol 2000; 54: 406–414
[31] Szeifert GT, Figarella-Branger D, Roche PH, Régis J. Histopathological observa-
[9] Szeifert GT, Atteberry DS, Kondziolka D, Levivier M, Lunsford LD. Cerebral
tions on vestibular schwannomas after Gamma Knife radiosurgery: the Mar-
metastases pathology after radiosurgery: a multicenter study. Cancer 2006;
seille experience. Neurochirurgie 2004; 50: 327–337
106: 2672–2681
[32] Kondziolka D, Lunsford LD, Flickinger JC. The radiobiology of radiosurgery.
[10] Szeifert GT, Levivier M, Lorenzoni J, Nyáry I, Major O, Kemeny AA. Morpho-
Neurosurg Clin N Am 1999; 10: 157–166
logical observations in brain arteriovenous malformations after gamma knife
[33] Major O, Szeifert GT, Radatz MW, Walton L, Kemeny AA. Experimental
radiosurgery. Prog Neurol Surg 2013; 27: 119–129
stereotactic gamma knife radiosurgery. Vascular contractility studies of
[11] Larsson B. Radiobiological fundamentals in radiosurgery. In: Steiner L, Lind-
the rat middle cerebral artery after chronic survival. Neurol Res 2002; 24:
quist C, Forster D, Backlund EO, eds. Radiosurgery: Baseline and Trends. New
191–198
York, NY: Raven Press; 1992
[34] Schneider BF, Eberhard DA, Steiner LE. Histopathology of arteriovenous
[12] Fajardo L-GLF, Berthrong M, Anderson RE, eds. Radiation pathology. New
malformations after gamma knife radiosurgery. J Neurosurg 1997; 87:
York, NY: Oxford University Press; 2001
352–357
[13] Leksell L. Sterotaxic radiosurgery in trigeminal neuralgia. Acta Chir Scand
[35] Szeifert GT, Kemeny AA, Timperley WR, Forster DM. The potential role of my-
1971; 137: 311–314
ofibroblasts in the obliteration of arteriovenous malformations after radio-
[14] Steiner L, Leksell L, Greitz T, Forster DM, Backlund EO. Stereotaxic radiosur-
surgery. Neurosurgery 1997; 40: 61–65, discussion 65–66
gery for cerebral arteriovenous malformations. Report of a case. Acta Chir
[36] Julow J, Slowik F, Kelemen J, Gorácz I. Late post-irradiation necrosis of the
Scand 1972; 138: 459–464
brain. Acta Neurochir (Wien) 1979; 46: 135–150
[15] Leksell L. A note on the treatment of acoustic tumours. Acta Chir Scand 1971;
[37] Yamamoto M. Radiosurgery for metastatic brain tumors. Prog Neurol Surg
137: 763–765
2007; 20: 106–128
[16] Backlund EO, Johansson L, Sarby B. Studies on craniopharyngiomas. II. Treat-
[38] Jagannathan J, Bourne TD, Schlesinger D, et al. Clinical and pathological char-
ment by stereotaxis and radiosurgery. Acta Chir Scand 1972; 138: 749–759
acteristics of brain metastasis resected after failed radiosurgery. Neurosur-
[17] Lunsford LD, Altschuler EM, Flickinger JC, Wu A, Martinez AJ. In vivo biologi-
gery 2010; 66: 208–217
cal effects of stereotactic radiosurgery: a primate model. Neurosurgery 1990;
[39] Kano H, Kondziolka D, Lobato-Polo J, Zorro O, Flickinger JC, Lunsford LD. T1/
27: 373–382
T2 matching to differentiate tumor growth from radiation effects after ster-
[18] Kondziolka D, Lunsford LD, Claassen D, Pandalai S, Maitz AH, Flickinger JC. Ra-
eotactic radiosurgery. Neurosurgery 2010; 66: 486–491, discussion 491–492
diobiology of radiosurgery: Part II. The rat C6 glioma model. Neurosurgery
[40] Kano H, Kondziolka D, Zorro O, Lobato-Polo J, Flickinger JC, Lunsford LD. The
1992; 31: 280–287, discussion 287–288
results of resection after stereotactic radiosurgery for brain metastases. J
[19] Blatt DR, Friedman WA, Bova FJ, Theele DP, Mickle JP. Temporal character-
Neurosurg 2009; 111: 825–831
istics of radiosurgical lesions in an animal model. J Neurosurg 1994; 80:
[41] Szeifert GT, Salmon I, Rorive S, et al. Does gamma knife surgery stimulate cel-
1046–1055
lular immune response to metastatic brain tumors? A histopathological and
[20] Inoue HK, Kohga H, Hirato M, Nakamura M, Ohye C. Neurobiologic effects of
immunohistochemical study. J Neurosurg 2005; 102 (Suppl): 180–184
radiosurgery: histologic, immunohistochemical and electron-microscopic
[42] Szeifert GT, Salmon I, Balèriaux D, Brotchi J, Levivier M. Immunohistochemi-
studies of a rat model. Stereotact Funct Neurosurg 1994; 63: 280–285
cal analysis of a cerebral arteriovenous malformation obliterated by radiosur-
[21] Linskey ME, Martinez AJ, Kondziolka D, et al. The radiobiology of human
gery and presenting with re-bleeding. Case report. Neurol Res 2003; 25:
acoustic schwannoma xenografts after stereotactic radiosurgery evaluated in
718–721
the subrenal capsule of athymic mice. J Neurosurg 1993; 78: 645–653
[43] Wikstrand C, Ashley DM, Bigner DD, Hale LP. Cellular immunology. Lympho-
[22] Niranjan A, Wolfe D, Tamura M, et al. Treatment of rat gliosarcoma brain tu-
cyte populations, cytokines and target-effector systems. In: Bigner D, McLen-
mors by HSV-based multigene therapy combined with radiosurgery. Mol
don R, Bruner JM, eds. Russell and Rubinstein’s Pathology of Tumors of the
Ther 2003; 8: 530–542
Nervous System. 6th ed. Vol. 1. London: Arnold; 1998:231–249
[23] Niranjan A, Moriuchi S, Lunsford LD, et al. Effective treatment of experimental
[44] Fuks Z, Kolesnick R. Engaging the vascular component of the tumor response.
glioblastoma by HSV vector-mediated TNF alpha and HSV-tk gene transfer in
Cancer Cell 2005; 8: 89–91
combination with radiosurgery and ganciclovir administration. Mol Ther
[45] Storer K, Tu J, Karunanayaka A, et al. Coadministration of low-dose lipopoly-
2000; 2: 114–120
saccharide and soluble tissue factor induces thrombosis after radiosurgery in
[24] Szeifert GT, Massager N, DeVriendt D, et al. Observations of intracranial neo-
an animal arteriovenous malformation model. Neurosurgery 2007; 61: 604–
plasms treated with gamma knife radiosurgery. J Neurosurg 2002; 97
610, discussion 610–611
(Suppl): 623–626

66
9 Stereotactic Radiosurgery for
Part III Arteriovenous Malformations 68

Vascular Indications 10 Stereotactic Radiosurgery for


Cavernomas 81

11 Stereotactic Radiosurgery for Dural


Arteriovenous Fistulas 91

III
Stereotactic Radiosurgery for Arteriovenous Malformations

9 Stereotactic Radiosurgery for Arteriovenous


Malformations
L. Dade Lunsford, Ajay Niranjan, Hideyuki Kano, Edward A. Monaco III, and John C. Flickinger

bleeding event, smaller AVM size, deep venous drainage, and


Key Points high flow rates have been suggested by some as increasing the
potential for subsequent bleeding. For-larger volume AVM
● Radiosurgery is associated with arteriovenous malformation (average diameter 4–5 cm), observation may be the only rea-
obliteration rates that average 78% of patients at 3 to 5 sonable strategy in view of the risks of even multimodality ma-
years. nagement.2 This may be especially true for patients who have
● Hemorrhage risks related to a history of prior arteriovenous never bled previously. Endovascular embolization using a varie-
malformation hemorrhage and are significantly higher in ty of particulate, glue, or coil methods may be used as an
patients with associated aneurysms. adjunct prior to craniotomy and surgical removal.3,4 Endovascu-
● Embolization prior to radiosurgery for intracranial arteriove- lar management has also been used in preparation for SRS,
nous malformations worsens eventual obliteration rates, but although its role prior to radiosurgery has declined with the
is an important part the management of dural arteriovenous realization that embolization rarely leads to significant volu-
fistulas. metric reduction. Although the flow within the AVM may
● Adverse radiation effects are related to dose, volume, and change after embolization, SRS must include the original vol-
location of the arteriovenous malformation. ume. In contrast, before surgical removal, embolization may
provide major benefit, either by reducing flow, or eliminating
deep-seated feeders that would otherwise be a significant pro-
blem during AVM resection. Recanalization of embolized AVM
9.1 Introduction components over time may require repeat SRS.
Surgical removal is an important option for patients with
Arteriovenous malformations (AVMs) are congenital vascular
lobar vascular malformations of suitable size, especially at cen-
anomalies comprised of an abnormal number of blood vessels
ters of excellence with extensive AVM experience. Incomplete
that are abnormally constructed. The blood vessels directly
removal requires adjuvant management, perhaps including
shunt blood from arterial input to the venous system without
radiosurgery. Spetzler and Martin, among others, defined the
an intervening capillary network to dampen pressure. Both
relationship of AVM volume, pattern of venous drainage, and
abnormal blood vessel construction and abnormal blood flow
location within critical areas of the brain as important consider-
lead to a risk of rupture and intracranial hemorrhage. In addi-
ations that help to facilitate outcome prediction at the time of
tion, patients with lobar vascular malformations may suffer
surgical resection at centers of excellence. Outcomes after AVM
from intractable vascular headaches or develop seizure disor-
radiosurgery do not correlate with the same predictions of the
ders. The annual incidence of AVM recognition is thought to be
Spetzler-Martin (SM) scale when microsurgery is used.5 Out-
approximately 10,000 patients per year in the United States.
comes after radiosurgery may be predicted based on volume,
However, the increasing reliance on magnetic resonance imag-
location, age, angioarchitecture, and dose delivered.6 Stereotac-
ing (MRI) has led to an increasing recognition of these vascular
tic radiosurgery is an excellent management strategy for
anomalies even in patients with minimal symptoms. The deci-
patients with AVMs < 30 mm in average diameter (for a single
sion making relative to management of an AVM must be care-
procedure). Staged procedures are used for larger vascular mal-
fully evaluated based on a number of risk factors. The options
formations or for those that were incompletely obliterated 3
for management include observation, endovascular emboliza-
years or more after an initial procedure.
tion alone or in preparation for other adjuvant management,
The chief benefit of radiosurgery management is risk reduc-
craniotomy and surgical removal, or stereotactic radiosurgery
tion; the chief deficit of radiosurgery is the latency interval that
(SRS).1 All treatments may be done in one or more stages.
is required to achieve complete obliteration of the AVM.7,8 The
In general, the following factors are evaluated when a patient
latency interval is generally 2 to 3 years, but in selected patients
is seen with an AVM: The patient’s age, associated medical con-
it may be longer. Arteriovenous malformation radiosurgery has
dition, history of a prior hemorrhagic event, prior management
been used for children not suitable for other management strat-
if any, overall volume and morphology, location of the AVM, ini-
egies as well as for older patients who have significant medical
tial presenting symptoms (headache, seizures, local neurologic
risk factors for surgical removal.
deficits), the AVM angio architecture (e.g., compact vs. diffuse
nidus), estimation of its surgical risks, presence of a proximal or
intranidal aneurysm and prior surgical experience in training.
In making a decision for management strategies, we often
9.2 The History of Radiosurgery
employ a decision-tree algorithm as shown in ▶ Fig. 9.1. Radiation to obliterate abnormal blood vessels in the brain is a
Optimal management depends on careful consideration of procedure that was first considered in the late 1960s. Raymond
the above factors and the estimated risk of subsequent hemor- Kjellberg, using the Harvard-affiliated proton facility in Cam-
rhage. The patient’s clinical presentation and location are bridge, Massachusetts, advocated proton Bragg peak stereotac-
important issues as well as symptoms in each patient. Age, prior tic radiation during the 1970s and early 1980s.9,10 More than

68
Stereotactic Radiosurgery for Arteriovenous Malformations

Fig. 9.1 Clinical algorithm for choosing management option for patients with intracranial arteriovenous malformations.

1,000 AVM patients were treated, but the dose-planning techni- Linear accelerator (linac) technologies have been adapted for
que was quite rudimentary. The technology of the Bragg peak SRS. Betti, working in France and Argentina,13 Barcia-Salorio in
proton facility was designed to provide a low exit dose based on Spain,14,15 and Columbo in Italy,16,17,18 were pioneers in the
the radiophysiological characteristics of this technology. The application of photon radiation using newer-generation linacs.
doses that were actually used in this series of patients were In addition, surgeons and radiation oncologists working at the
quite low, and do not correspond to doses that we now know Joint Center in Boston, Massachusetts,19 and in Gainesville, Flor-
may be effective in the obliteration of an AVM. Although Kjell- ida,20 used modified linacs to treat a large number of vascular
berg maintained that Bragg peak radiation stabilized AVM malformations. Most centers continue to evaluate SRS as part of
blood vessel walls and reduced the subsequent risk of hemor- an overall management plan, which may include embolization,
rhage (in comparison with age-related survivals from a life microsurgical removal, or radiosurgery alone or in combination.
insurance table), only 20% of patients had complete obliteration We will discuss our current viewpoint relative to the role of
of their AVMs over time. Fabrikant and Steinberg, working at embolization subsequently.
the Lawrence Livermore Laboratory in Berkley, California,
began to use a helium ion beam to perform multisession AVM
irradiation in the 1980s.11 9.3 The University of Pittsburgh
In Stockholm, Lars Leksell and Ladislau Steiner initiated work
with the first-generation Leksell Gamma Knife unit in Stock-
Experience
holm.12 The first patient was treated in March 1970, using the Our first AVM patient was treated in August 1987,21 and we
original prototype 179-cobalt-60 source photon beam unit reported our initial experience with 227 patients in 1991. 22
designed by Larsson and Leksell. The target definition was Confirming the work of others, we noted that the 2-year success
based on biplane angiography done during the procedure itself. rate in terms of complete obliteration was related to nidus vol-
Patients were observed for a period in preparation for a larger ume and dose. In high-dose cases (AVM < l cc), obliteration rates
experience that began to emerge in Stockholm using the sec- of 100% were noted, which declined to 85% for volumes of 1 to
ond-generation unit built in 1975. 4 cc and 58% for AVMs larger than 4 cc in volume.

69
Stereotactic Radiosurgery for Arteriovenous Malformations

Table 9.1 Patient demographics of twenty-year radiosurgery experience Table 9.2 Brain locations and grades of 1,050 arteriovenous malforma-
at the University of Pittsburgh 1987–2010 tions (AVM)

N 1,050 AVM location (N = 906)

Patient age Temporal 17.8%

Median 36 years Frontal 18.4%

Range 3–80 years Parietal 17.6%

Gender Thalamus/basal ganglia 15.3%

Male 546 (52%) Occipital 11.5%

Female 504 (48%) Cerebellar 6.4%

Presenting symptoms Brainstem 6.3%

Hemorrhage 472 (45%) Dural 2.8%

Seizures 233 (22%) Corpus callosal 1.8%

Headache 195 (19%) Intraventricular 1%

Sensory motor deficit 84 (8%) Pineal 1%

Incidental 66 (6%) Spetzler-Martin grade

Prior management Grade I 2.2%

Embolization 210 (20%) Grade II 24.0%

Surgery 73 (7%) Grade III 43.8%

Grade IV 15%

Grade V 3%
Our subsequent 25-year experience in Pittsburgh, Pennsyl-
vania, has now increased to more than 1,400 patients who Grade VI 12%
have undergone Gamma Knife radiosurgery for their AVM
Coexistence of aneurysm 98 (9.3%)
using one or more radiosurgical procedures. We have more
recently analyzed the outcome data of 906 patients who
underwent radiosurgery between 1987 and 2004 (▶ Table 9.1,
▶ Table 9.2, ▶ Table 9.3). Our median patient age was 36 Table 9.3 Radiosurgical parameters of arteriovenous malformations
years (with a range of 3–80). Typical symptoms at presenta- treated at the University of Pittsburgh
tion included hemorrhage (46%), seizures (24%), and head- AVM radiosurgical parameters
ache (18%). Eight percent had neurologic deficits (8%). An
AVM volume
incidental AVM was detected in 4% of patients. Prior manage-
ment strategies included surgical removal or clot evaluation Median 3.4 mL
in 7%. Twenty-one percent underwent one or more interven-
Range 0.065–57.7 mL
tional procedures (embolization). The median target volume
was 3.4 cc (range .065–57.7 cc). The median margin dose was Radiosurgery dose
20 Gy (range 13–32 Gy). A single procedure was performed in Median 20 Gy
865 (95.5%) of patients, and repeat radiosurgery for incom-
Range 13–32 Gy
plete nidus obliteration after 3 years was needed in 113
(12.5%) patients. Prospective volume-staged radiosurgery Radiosurgery
was performed in 41 (4.5%) patients. Single session 1,000/1,050 (95.2%)
At a median follow-up of 3 years, complete nidus obliteration
was achieved in 78% (confirmed by angiography or MRI). In Prospective volume staged 50/1,050 (5.0%)
addition, 21% of patients achieved subtotal obliteration of the Repeat radiosurgery 130/1,050 (12.4%)
nidus. During the follow-up interval, 38 bleeds (4.1%) occurred
after the procedure. Seizure control was improved in 51% of
patients who presented with seizures. Adverse radiation effects
(AREs) resulting in neurological deficits developed in 24 A subset of (n = 217) patients with AVMs who underwent
patients (2.6%), and the detection of new T2 signal increase sur- Gamma Knife surgery at our institution was classified as SM
rounding the AVM target in 108 patients (12%). We noted long- grade I or II.23 The median target volumes was 2.3 cc (range
term complications such as delayed cyst formation or encepha- 0.1–14.1 cc). The median margin dose was 22 Gy (range 15–
lomalacia in 16 patients (1.7%). No patient in this AVM series 27 Gy). Arteriovenous malformation obliteration was confirmed
has developed a radiation-related tumor at the time of this by MRI in 148 patients and by angiography in 100 patients with
writing. a median follow-up of 64 months (range 6–247 months). The

70
Stereotactic Radiosurgery for Arteriovenous Malformations

actuarial rates of total obliteration determined by angiography hemorrhage during the latency period and seven patients died.
or MRI after one SRS procedure were 58, 87, 90, and 93% at 3, 4, The rate of post-SRS AVM hemorrhage was 4.5, 6.2, 9.0, 11.2,
5, and 10 years, respectively. The median time to complete and 15.4% at 1, 2, 3, 5, and 10 years, respectively. The overall
MRI-determined obliteration was 30 months. Cumulative rates annual hemorrhage rate was 4.7%. When five patients with sev-
of AVM hemorrhage 1, 2, 3, 5, and 10 years after SRS were 3.7, en hemorrhages occurring earlier than 6 months after SRS were
4.2, 4.2, 5.0, and 6.1%, respectively. This corresponded to rates removed from this analysis, the annual hemorrhage rate
of annual bleeding risk of 3.7, 0.3, and 0.2% for years 0–1, 1–5, decreased to 2.7%. Larger-volume AVMs had a higher risk of
and 5–10, respectively, after SRS. The presence of a coexisting hemorrhage after SRS. Permanent neurologic deficits due to
aneurysm proximal to the AVM correlated with a significantly AREs developed in six patients (4.5%), and in one patient a
higher hemorrhage risk. Temporary symptomatic adverse radi- delayed cyst developed 56 months after SRS.
ation effects developed in five patients (2.3%) after SRS, and Sixty-seven patients had AVMs in the brainstem.27 In this ser-
two patients (1%) developed delayed cysts. ies, 51 patients (76%) had a prior hemorrhage. The median tar-
We performed SRS in 474 patients with SM grade III AVMs. 24 get volume was 1.4 cc (range 0.1–13.4 cc). The median margin
The AVMs were categorized by scoring the size (S), drainage dose was 20 Gy (range 14–25.6 Gy). Obliteration of the AVMs
(D), and location (L): IIIa was a small AVM (S1D1L1, n = 282); IIIb was eventually documented in 35 patients at a median follow-
was a medium/deep AVM (S2D1L0, n = 44); and IIIc was a me- up of 73 months (range 6–269 months). The actuarial rates of
dium/eloquent AVM (S2D0L1, n = 148). The median target vol- documentation of total obliteration were 41, 70, 70, and 76% at
ume was 3.8 mL (range 0.1–26.3 mL) and the margin dose was 3, 4, 5, and 10 years, respectively. Higher rates of AVM oblitera-
20 Gy (range 13–25 Gy). Eighty-one patients (17%) underwent tion were associated only with a higher margin dose. Four
prior embolization, and 58 (12%) underwent prior resection. At patients (6%) suffered a hemorrhage during the latency period,
a mean follow-up of 89 months, the total obliteration rates and two patients died. The rate of AVM hemorrhage after SRS
documented by angiography or MRI for all SM grade III AVMs was 3.0, 3.0, and 5.8% at 1, 5, and 10 years, respectively. The
increased from 48% at 3 years to 69% at 4 years, 72% at 5 years, overall annual hemorrhage rate was 1.9%. Permanent neuro-
and 77% at 10 years. The SM grade IIIa AVMs were more likely logic deficits due to AREs developed in seven patients (10%)
to obliterate than other subgroups. The cumulative rate of hem- after SRS, and a delayed cyst developed in two patients (3%).
orrhage was 2.3% at 1 year, 4.4% at 2 years, 5.5% at 3 years, 6.4% Higher 12-Gy volumes and higher SM grades were associated
at 5 years, and 9% at 10 years. The SM grade IIIb AVMs had a sig- with a higher risk of symptomatic AREs.
nificantly higher cumulative rate of hemorrhage. Symptomatic Sixty-four patients (median age 47 years, range 8–75 years)
adverse radiation effects were detected in 6%. underwent SRS for a cerebellar AVM.28 Forty-seven patients
At the University of Pittsburgh, 135 children underwent (73%) presented with an intracranial hemorrhage. The median
SRS.25 The median target volume was 2.5 cc (range 0.1–17.5 cc). target volume was 3.85 cc (range 0.2–12.5 cc), and the median
The median margin dose was 20 Gy (range 15–25 Gy). The marginal dose was 21 Gy (range 15–25 Gy). AVM obliteration
actuarial rates of total obliteration documented by angiography was confirmed by MRI or angiography in 40 patients at a
or MRI at 71.3 months (range 6–264 months) were 45, 64, 67, median follow-up of 73 months (range 4–255 months). The
and 72% at 3, 4, 5, and 10 years, respectively. The median time actuarial rates of total obliteration were 53% at 3 years, 69% at 4
to complete angiographically documented obliteration was 48.9 years, and 76% at 5 and 10 years. Elevated obliteration rates
months. Of 81 patients with 4 or more years of follow-up, 57 were statistically higher in patients who underwent AVM SRS
patients (70%) had total obliteration documented by angiogra- without prior embolization. A smaller AVM volume was also
phy. Factors associated with a higher rate of documented AVM associated with a higher rate of obliteration. Four patients (6%)
obliteration were smaller AVM target volume, smaller maxi- sustained a hemorrhage during the latency period and three
mum diameter, and larger margin dose. The rates of AVM hem- died. The cumulative rates of AVM hemorrhage after SRS were
orrhage after SRS were 0, 1.6, 2.4, 5.5, and 10.0% at 1, 2, 3, 5, 6% at 1, 5, and 10 years. This correlated with an overall annual
and 10 years, respectively. The overall annual hemorrhage rate hemorrhage rate of 2.0% during the latency interval. One pa-
was 1.8%. Larger-volume AVMs were associated with a signifi- tient experienced a hemorrhage 9 years after confirmed MRI
cantly higher risk of hemorrhage after SRS. Permanent neuro- and angiographic obliteration. A permanent neurologic deficit
logic deficits due to adverse radiation effects developed in two due to adverse radiation effects developed in one patient (1.6%)
patients (1.5%) after SRS; in one patient (0.7%), delayed cyst for- and temporary complications were seen in two additional
mation occurred. patients (3.1%).
Fifty-six patients with basal ganglia AVMs and 77 with thala- Eighty-seven patients underwent SRS for AVMs in the region
mic underwent SRS.26 Of these 113 (85% of 133 patients) had a of the sylvian fissure.29 Before undergoing SRS, 40 (46%) of
prior hemorrhage. The median target volume was 2.7 cc (range these patients had experienced hemorrhage and 36 (41%) had
0.1–20.7 cc), and the median margin dose was 20 Gy (range 15– had seizures. The median target volume of the AVM was 3.85 cc
25 Gy). Obliteration of the AVM eventually was documented on (range 0.1–17.7 cc), and the median marginal dose of radiation
MRI in 78 patients and on angiography in 63 patients in a was 20 Gy (range 13–25 Gy). Over a median follow-up period of
median follow-up period of 61 months (range 2–265 months). 64 months (range 3–275 months), AVM obliteration was con-
The actuarial rates documenting total obliteration after radio- firmed by MRI or angiography for 43 patients. The actuarial
surgery were 57, 70, 72, and 72% at 3, 4, 5, and 10 years, respec- rates of confirmation of total obliteration were 35% at 3 years,
tively. Factors associated with a higher rate of AVM obliteration 60% at 4 and 5 years, and 76% at 10 years. Of the 36 patients
included basal ganglia location, a smaller size, and a higher who had experienced seizures before SRS, 19 (53%) achieved
margin dose. Fifteen (11%) of 133 patients suffered a outcomes of Engel class I after treatment. The rate of seizure

71
Stereotactic Radiosurgery for Arteriovenous Malformations

improvement was 29% at 3 years, 36% at 5 years, 50% at 10 MRI scans are not possible (those with undetermined implants,
years, and 60% at 15 years. No seizures developed after SRS in pacemakers, old aneurysm clips, etc.), we use contrast-
patients who had been seizure-free before treatment. The enhanced stereotactic computed tomography angiography
actuarial rate of AVM hemorrhage after SRS was 5% at 1, 5, and (CTA). The development of axial imaging as part of the imaging
10 years. This rate equated to an annual hemorrhage rate dur- paradigm is critical for AVM management. This has allowed us
ing the latency interval of 1%; no hemorrhages occurred after to make superior 3D conformal plans much more carefully in
confirmed obliteration. No permanent neurologic deficits comparison with using two-plane angiography alone.
developed as an adverse effect of radiation; however, delayed Imaging studies are placed in a dose-planning computer sys-
cyst formation occurred in three patients. tem (Elekta Instruments, Inc., Atlanta, GA, USA), which has both
One-hundred eighty-eight patients had ventricular-region high speed and high resolution. We begin the planning using
AVMs.30 The median patient age was 32 years (range 3–80 MRI while the patient is undergoing angiography. Angiography
years), the median target volume was 4.6 cc (range 0.1–22 cc), is used to fine tune or idealize the angiographic plan. In both
and the median marginal dose was 20 Gy (range 13–27 Gy). the 4-C and Perfexion models currently used, optimization of
Arteriovenous malformation obliteration was confirmed by the dose planning is achieved by confining the sharpest falloff
MRI or angiography in 89 patients during a median follow-up isodose (usually the 50%) to the edge of the 3D-defined volume.
of 65 months (range 2–265 months). The actuarial rates of total Each slice is looked at serially to confirm excellent conformality
obliteration were 32% at 3 years, 55% at 4 years, 60% at 5 years, (conforming the edge dose to the 3D target volume). High selec-
and 64% at 10 years. Higher rates of AVM obliteration were tivity (rapid falloff of the radiation dose outside of the target) is
obtained in the 26 patients with intraventricular AVMs. equally important (▶ Fig. 9.2). We also assess the patient for the
Twenty-five patients (13%) sustained a hemorrhage during the presence of proximal and intranidal aneurysms. We strive to
initial latency interval after Gamma Knife surgery, indicating an reduce dose to draining veins when feasible. Typical doses at
annual hemorrhage rate of 3.4% prior to AVM obliteration. No the margin of the AVM are 18 to 25 Gy and never below 16.
patient experienced a hemorrhage after AVM obliteration was Maximal doses are usually 36 to 50 Gy at the margin of the
confirmed by imaging. Permanent neurologic deficits due to AVM. Final dose selection depends on the volume and location,
adverse radiation effects developed in seven patients (4%). which also helps to estimate ARE risks. We also consider the
presence of preexisting neurologic conditions, the patient’s age,
and prior bleeding history. Because the dose to the surrounding
9.4 The Stereotactic Radiosurgical brain is a critical predictor of ARE, we must maximize confor-
Procedure mality and selectivity.
At the conclusion of the procedure, patients receive 20 to
We perform intracranial radiosurgery using the Leksell Gamma 40 mg of methylprednisolone. This coupled with anticonvul-
Knife (beginning with the model unit U, and proceeding on to sants in lobar AVM patients tends to reduce the risk of periope-
the B, C, 4-C, and in recent experience the Perfexion model) rative seizures. For those patients who have previously
(Elekta AB, Stockholm, Sweden). The patient’s clinical studies undergone embolization in an attempt to reduce flow or to
and imaging are reviewed for suitability for SRS. As noted previ- achieve volumetric reduction of the AVM, or for those who have
ously, we evaluate the bleeding history, the age of the patient, had intracranial surgery or hematoma evacuation, Gamma
existing comorbidities, location, and clinical symptomatology. Knife radiosurgery may be used as an adjuvant strategy rather
Patients with lobar AVMs were placed prophylactically on anti- than a primary management. We often perform radiosurgery
convulsants for a period of 2 to 4 weeks around the time of the once the patient has achieved a stable neurologic improvement,
procedure. This has reduced the risk of a perioperative seizure but almost never within the first month after an ictal event
event from as high as 5% in year 1 of our 20-year experience to such as a bleed or embolization. For patients who have had
a risk of less than 1% at the current time. We require that all intracranial hemorrhages, we prefer to wait between 1 and 3
women within the child-bearing age have a recent negative months to see if there will be a regional clot reabsorption. The
pregnancy test or we perform it on the day of the procedure. AVM nidus should not be compressed by clot at the time of the
Patients are evaluated preoperatively by the neurosurgeon, radiosurgery. Failure of radiosurgery can be traced in some
the radiation oncologist, and the nursing team. On the morning ways to inadequate planning, inadequate recognition of the 3D
of the procedure, the patients arrive at 6:00 am and begin con- geometry of the AVM, reappearance of a component of the AVM
scious sedation using oral lorazepam followed by intravenous previously embolized, or reappearance of a component of the
conscious sedation (fentanyl and midazolam) as needed. Scalp AVM that was previously compressed by intracerebral
anesthetic injection using a combination of Marcaine and Xylo- hematoma.
caine is injected at the sites of pin application. At the current Follow-up imaging is recommended at regular intervals.
time, we prefer to use titanium pins with plastic inserts to min- Whenever possible, imaging is done at our center, but if the pa-
imize the risk of MRI artifacts. General anesthesia may be tient lives at a distance, we recommended the images be done
required for frame application in imaging in patients younger at an imaging site closer to home. Communication with the
than 12 years of age. patient’s referring physician team is critical. We normally rec-
Neurodiagnostic imaging follows using both intravenous par- ommend MRI scans as 6 months and then annually to assess
amagnetic contrast-enhanced three-dimensional (3D) volumet- the effect of radiosurgery. If at the end of 3 years MRI suggests
ric MRI scan, and a whole-head T2 fast spin-echo imaging complete obliteration, then we request that a repeat DSA be
sequence. Patients subsequently undergo biplane digital sub- performed. If MRI clearly defines residual nidus, angiography is
traction angiography (DSA) as the next step. In patients where delayed, and the patient is contacted to suggest the possible

72
Stereotactic Radiosurgery for Arteriovenous Malformations

Fig. 9.2 (a) Anteroposterior and lateral carotid


artery angiograms documenting the left frontal
arteriovenous malformation with a volume of 4.7
cc at the time of the initial stereotactic radio-
surgery (SRS). The margin dose was 22 Gy. (b)
Anteroposterior and lateral carotid artery angio-
grams, 4 years after initial SRS, showing a residual
nidus with a volume of 2 cc in the SRS field. This
residual nidus was treated by the second SRS with
a margin dose of 20 Gy. (c) Anteroposterior and
lateral carotid artery angiograms, 4 years after
the second SRS, showing the absence of nidus.

need for repeat radiosurgery to achieve the final obliteration re- begin until the radiosurgical procedure has been completed for
sponse. In such cases, repeat stereotactic imaging may include all volumetric components of the AVM.
both repeat MRI as well as angiography.
For those patients who have large AVMs (determined by mul-
tiplying the MRI X, Y, Z dimensions of the AVM and dividing by 9.5 Current Outcomes of AVM
2, a rough approximation of an ellipsoid volume), we recom-
mend consideration of prospective staged SRS. Staged radio-
Stereotactic Radiosurgery
surgery, with two stages separated usually by 3 months, is In the absence of treatment, the overall risk of a spontaneous
recommended for AVMs larger than 15 cm3. We also consider it bleed from a brain AVM appears to range from 1 to 5% per year
for those AVMs between 10 and 15 cm3. For those AVM volumes depending upon various risk factors.31 In the Finnish popula-
10 cm3 or smaller, we normally perform single-stage radiosur- tion-based 24-year study, hemorrhage was a relatively constant
gery. The interval between stages varies from 3 to 6 months lifetime risk with an annual death risk of approximately 1% and
based on the goal of some radiobiological repair of surrounding approximately a 4% risk of bleed per year.31 Recent work
brain during the observation interval. However, we do not want reported from the cooperative trial in Canada suggested that an
to wait too long, as the goal of protection from bleeding cannot annual bleed rate may be as high as 5% (C. Wallace, personal

73
Stereotactic Radiosurgery for Arteriovenous Malformations

communication). We performed an individualized analysis of Clearly, for a child the risk is very large, whereas for a senior
the hemorrhage risk of AVM patients before radiosurgery. 32 Our citizen the remaining lifetime risk of a bleed for a patient who
findings demonstrated an overall crude annual hemorrhage has never bled before may be reasonably low.
rate of 2.4% per year.
There were several factors associated with hemorrhage risk,
the primary one being a prior hemorrhage (an identification of 9.6 Bleeding Risks after AVM
a single draining vein on angiography), and the detection of a
diffuse AVM nidus on angiography. Pollock et al constructed an
Stereotactic Radiosurgery
estimation of bleed risks related on these significant factors We have also analyzed the bleeding rate during the observation
(▶ Table 9.4 and ▶ Table 9.5).32 For low-risk AVMs (no prior interval (latency interval) after radiosurgery and before com-
hemorrhage and no other risk factors, diffuse nidus or single plete AVM obliteration.35 For this study, we evaluated 312
draining vein), the annual risk rate was approximately 1% per patients with clinical angiographic outcomes followed for an
year. In contrast, the risk of a second hemorrhage for patients average of 4 years. Twenty-one patients suffered AVM bleeds at
with additional risk factors ranged from 2.0 to 3.7% for AVMs a median of 8 months after radiosurgery. The overall total risk
with a compact nidus up to 8.94% for those with high-risk fea- of postradiosurgery hemorrhage per patient was 7.4%, after
tures of the angioarchitecture (one draining vein or diffuse exclusion of additional bleeding risk factors such as untreated
morphology). aneurysms. The actual hemorrhage rate from a patent AVM
In talking with families, we often use a rule of thumb to esti- before complete obliteration was 4.8% per year during the first
mate the lifetime bleeding risk related to a patient’s age. The 2 years after radiosurgery, and 5% per year for the third to fifth
age at which the risk of a bleed is greater than the risk of mor- years after radiosurgery, if the AVM continued to be unobliter-
bidity from radiosurgery may well depend upon the location ated. Our data have not provided strong evidence to date that
and size of the AVM. A simple lifetime analysis risk rate sug- there is a protective benefit of radiosurgery that gradually
gests that a patient’s age subtracted from 105 will give the total emerges during the observation years, even before complete
cumulative risk of that patient having a bleeding event.33,34 obliteration has occurred. In contrast, studies from the Univer-
sity of Tokyo experience reported by Maruyama and col-
leagues36 as well as reinterpretation of the outcome data from
Table 9.4 Estimate annual risk of initial and second hemorrhages in arte- the Karolinska experience37of Steiner have suggested that there
riovenous malformation (AVM) patients with an arteriovenous malforma-
may be some protective benefit to AVMs even before complete
tion (AVM)32
obliteration of their nidus occurs.
AVM characteristics Estimated risk of intracranial hemorrhage Our recent analysis showed that at our institution, 33
1st bleed 2nd bleed patients (8%) sustained an additional hemorrhage after SRS. 38
The overall annual hemorrhage rate until obliteration after SRS
Low-risk AVM (well-
was 1.3%. The presence of a patent aneurysm was significantly
defined nidus and > 1 1.00% 3.70%
draining vein) associated with an increased rehemorrhage risk after SRS
(annual hemorrhage rate, 6.4%) compared with patients with a
High-risk AVM (diffuse clipped or embolized aneurysm (annual hemorrhage rate, 0.8%;
nidus or only one 2.20% 8.90%
p = 0.033). Patients who have a proximal unsecured aneurysm
draining vein)
have an increased risk of postradiosurgical hemorrhage. If the

Table 9.5 Estimated lifetime risk of hemorrhage according to history of prior hemorrhage and whether any high-risk morphological risk features
(increased risk diffuse morphology or one draining vein) are absent or present 32,34
Patient age at Lifetime risk of
Expected survival (y)
diagnosis (y) intracranial bleed

Low-hemorrhage-risk AVMs High-hemorrhage-risk AVMs

No prior bleed Prior bleed No prior bleed Prior bleed

15 77 46 90.5 75.1 99.7

25 67 40.4 86.1 68.9 99.2

35 78 34.8 80.4 61.9 98.2

45 79 28.7 72.4 53.4 95.9

55 80 22 61.2 43 90.4

65 83 16.4 49.5 33.2 81.5

75 86 10.4 34.1 21.9 64.3

85 91 5.8 20.3 12.6 43

Abbreviation: AVM, arteriovenous malformation.

74
Stereotactic Radiosurgery for Arteriovenous Malformations

aneurysm is immediately proximal to the AVM, it will likely was seen in 18% of previously embolized patients, but only 5%
close as the AVM obliterates. We have not found that intranidal of nonembolized patients. We have also noted that the success-
aneurysms increase the risk of bleeding during the latency ful obliteration for the same-volume AVMs may be slightly low-
interval. For those patients with aneurysms in more than one er in women and is higher in children; however, it occurs more
arterial branch proximal to their AVM, we believe that the completely and at a faster rate in children than in adults.
aneurysm requires a different management algorithm that
should be determined based on those characteristics that guide
whether surgery or endovascular is best management. Such 9.8 Adverse Effects of
aneurysms generally do not go away at the time that the AVM is
obliterated.
Radiosurgery
To date, in our experience no patient has suffered a hemor- Early adverse effects are relatively rare and include headache
rhage after definitive high-resolution angiography has con- from the frame application, nausea from conscious sedation
firmed complete obliteration of their AVM. We have a single medications, and the relatively small risk of developing seizures
patient with more than 50 aneurysm clips who has undergone in patients with subcortical lobar AVMs.39,40,41,42 It is for this
multiple surgical procedures. The AVM was thought to be obli- reason that prophylactic anticonvulsants are used in these
terated after radiosurgery, but the patient suffered a subse- patients, but not in patients with deep-seated AVMs. Late AREs
quent hemorrhage. A new AVM was identified on follow-up of radiosurgery are relatively rare. We evaluated data from 85
angiography (neither CT nor MRI scan was feasible in this pa- AVM patients who developed symptomatic complications after
tient). The possibility of recanalization of previously treated Gamma Knife radiosurgery and compared this with 337
AVM requiring additional treatment and monitoring is impor- patients who had no complications and were evaluated as part
tant. For this reason, even in those patients who have angio- of another multi-institutional study.43 Thirty-five of 85 patients
graphic complete closure of their AVM, we recommend MRI were classified as having permanent symptomatic sequelae. We
scans at 2-year intervals to assess the overall brain response to constructed various models to study the effects of AVM location
detect late AREs or delayed cyst development. and the volume of tissue receiving 12 Gy or more (the 12-Gy
Although the studies of both Karlsson et al and Maruyama et volume) with the risk of developing permanent postradiosur-
al36,37provide some evidence of an overall reduction in bleeding gery AREs. The locations of AVMs in increasing order of risk
rates during the latency interval, this hypothesis remains were frontal, temporal, interventricular, parietal, cerebellar, cor-
largely unproven at the present time. In addition, both ours as pus callosum, occipital, medulla, thalamus, basal ganglia, and
well as other outcome studies have shown that even with com- pons and midbrain. We were able to construct statistical mod-
plete obliteration, the hemorrhage rate is not zero. For patients els predicting the risk of permanent radiation sequelae with a
who have defined angiographic obliteration, we may safely esti- 12-Gy volume. Such data are very important for subsequent
mate that the lifetime risk of a bleed in such patients is now less planning of additional patients relative to a risk:benefit analy-
than 1%. sis. This database, though useful, was constructed with a rela-
tively small number of complications considering the large
number of patients and the relatively large number of variables.
9.7 Obliteration of an It is likely that the risk prediction for some brainstem locations
is significantly overestimated. Certainly, the risk of complica-
Arteriovenous Malformation after tions is expected to be high for large-volume AVMs in critical
locations of the brain. It is for this reason that for larger-volume
Stereotactic Radiosurgery AVMs, 15 cc or more, we clearly recommend staging with the
Of the 996 patients with AVMs in our series, 407 patients had goal to increase the obliteration rate while maintaining safety.
sustained an hemorrhage.38 The overall rate of total obliteration We generally divide the AVM into two volumes of approxi-
defined by angiography or MRI was 56, 77, 80, and 82% at 3, 4, mately equal proportion. At the time of the first procedure
5, and 10 years, respectively. Furthermore, we believe that there (which is done with angiography and MRI), we devise a plan for
is approximately a 95% accuracy that MRI-detected obliteration the entire AVM and then gradually subtract isocenters from the
will be confirmed by follow-up angiography.39 There are some plan until we develop a 50% volume that corresponds to the ini-
patients who are unwilling to undergo repeat angiography even tial stage. The second-stage volume is then done to reproduce
though this remains the gold standard for detection of res- the original plan at an interval of 3 to 6 months. This strategy
ponse. Even the presence of an early draining vein, without dis- promotes the brain’s ability to repair normal tissue and to pre-
cernible nidus, is a sign of satisfactory response. To our vent adverse radiation effects.
knowledge, no patient has subsequently bled when only an Other complications of AVM radiosurgery are relatively rare.
early draining vein is seen. Follow-up angiography afterwards Such complications have taken many years to fully understand
at 6 months to 1 year in such cases invariably shows loss of the or even develop. These risks include the risk of hemorrhage
early draining vein as well. despite obliteration (1% lifetime), the risk of temporary or per-
Failure of obliteration is multifactorial, and as noted earlier manent radiation injury, the risk of late cyst formation at the
may be related to dose, volume, inadequate recognition of the site of the obliterated nidus, and the long-term risk for radia-
3D geometry, recanalization of the previously embolized com- tion-induced tumor. Cyst formation after AVM radiosurgery
ponent, or a clot-compressed AVM that was subtotally treated. was first reported by Japanese investigators who had sent
In our studies designed to detect the reasons for marginal fail- patients to receive Gamma Knife radiosurgery in Sweden in the
ure, we noted that such persistence of out-of-field residual AVM early years of radiosurgery.44 Cyst formation has also been

75
Stereotactic Radiosurgery for Arteriovenous Malformations

reported in other long-term follow-up studies.45,47 In our 20- repeat SRS were a greater number of prior hemorrhages, larger
year experience, we have detected 16 patients (1.7%) with AVM target volume at initial SRS, larger target volume at repeat
delayed cyst formation. We also observed that patients who SRS, initial AVM volume reduction less than 50%, and a higher
developed delayed cyst formation were more likely to have had Pollock-Flickinger score. Symptomatic adverse radiation effects
prior bleeds. This raises the intriguing possibility that residual developed in 5 patients (4.8%) after initial SRS and in 10
iron deposition in the brain tissue may serve as a radiation sen- patients (9.5%) after repeat SRS. Prior embolization and a higher
sitizer that could potentiate the effects of radiosurgery on a SM grade were significantly associated with higher rates of
long-term basis. Such cysts have been managed with observa- adverse radiation effects after repeat SRS. Delayed cyst forma-
tion, simple drainage, cyst shunting, or surgical fenestration. tion occurred in five patients (4.8%) at a median of 108 months
Patients with perioperative T2 signal change without additional after repeat SRS (range 47–184 months). In this group, the per-
neurologic problems do not require additional treatment. manent neurologic injury was slightly higher than would be
In our experience of more than 14,000 Gamma Knife proce- expected with those patients who had no prior radiation
dures, we have not identified a patient who fits the Cahan injury.53 However, it should be recalled that most patients had
requirements for a radiation-related tumor.46 Recently, Sheehan relatively larger AVMs to begin with, and received relatively
and Steiner reported two patients who have had radiosurgery lower radiation margin doses. If we are unable to give in a sin-
for their AVM and developed delayed meningiomas.48 As a rela- gle procedure more than 16 Gy or more, we believe that staged
tively common tumor in older patients, it is not clear from a radiosurgery must be considered to try to increase the chances
population basis that this represents a significant increased risk of the obliteration rate. We know that the successful oblitera-
compared with the natural history of meningioma develop- tion response declines rapidly below a 15-Gy edge dose.
ment. There are, however, two additional cases of a glioblasto-
ma reported after AVM radiosurgery.49,50 With more than half a
million patients having undergone Gamma Knife radiosurgery 9.10 The Management of Large
over the last 40 years, the denominator (number treated) is rel- Arteriovenous Malformations
atively known, but the numerator (the incidence of radiation-
related tumors) remains largely an estimate. We provide an Large AVMs pose a major challenge both for surgical resection
estimate to patients that the gross risk of developing a radia- embolization and radiosurgery. Multimodality management is
tion-related tumor is approximately 4 in 50,000, a 1 in 12,500 probably going to be critical for such AVMs that have any hope
risk. We do, nonetheless, warn our patients that the risk of a of treatment. Embolization of an AVM has been performed
radiation-related tumor may be as high as 1 in 1,000, although using a variety of agents over the course of time, including glue,
neither our personal experience nor data published from Shef- coils, silk threads, polyvinyl alcohol, and more recently Onyx
field, England, can confirm this incidence.51 (ev3, Plymouth, MN). Prior studies have shown the recanaliza-
tion rates of 14 to 15% of patients who had prior AVM emboliza-
tion. Because we cannot use a single-staged radiosurgical
9.9 Repeat Radiosurgery procedure for such large-volume AVMs, we have recommended
a staged prospective approach. Although fractionated radio-
For those patients who have a residual nidus identified by therapy (2–4 Gy per fraction to a total dose of 50 Gy) has virtu-
imaging 3 or more years after radiosurgery, we recommend ally no significant benefit54 in terms of AVM obliteration while
repeat radiosurgery. For such patients, we are occasionally able having significant long-term side-effect risks, radiosurgery div-
to increase the dose slightly to the residual smaller-volume ided into two sessions seems to have an acceptable risk predic-
AVMs. We evaluated the outcomes and risks of repeat SRS for tion. Kjellberg using stereotactic Bragg peak radiotherapy
incompletely obliterated cerebral AVMs.52 During a 20-year achieved obliteration rates of < 20%, although he postulated that
period, repeat SRS was performed in 105 patients who had such radiation stabilized blood vessel walls reduced the risk of a
incompletely obliterated AVMs at a median of 40.9 months after subsequent bleed.10 Most of the AVMs treated by this technique
initial SRS (range 27.5–139 months). The median AVM target had large-volume AVMs. In a group of 48 patients treated by
volume was 2.3 cm3 (range 0.1–18.2 cm3) at the time of the sec- Pan et al,55 an obliteration rate of only 25% was noted after 14
ond procedure. The median margin dose at repeat SRS was months in a single radiosurgical strategy when relatively larger
18 Gy. The actuarial rate of total obliteration by angiography or doses were given to the edge (16.5–17.7 Gy). Approximately
MRI after repeat SRS was 35, 68, 77, and 80% at 3, 4, 5, and 10 37% had moderate and 12% had severe adverse radiation effects
years, respectively. The median time to complete angiographic when the AVM volume was larger than 10 cc. We noted the
or MRI obliteration after repeat SRS was 39 months. Factors rather narrow window between dose response and complica-
associated with a higher rate of AVM obliteration were smaller tion, and the low chance of receiving a total obliteration with
residual AVM target volume and a volume reduction of 50% or low dose. We switched to prospective volume staging
more after the initial procedure. Seventeen patients (16%) had (▶ Fig. 9.3) as an option to manage larger AVMs unsuitable for
hemorrhage after repeat SRS and six patients died. The cumula- primary single-session radiosurgery. At our institution, 47
tive actuarial rates of new AVM hemorrhage after repeat SRS patients with large AVMs underwent SRS using two or more
were 1.9, 8.1, 10.1, 10.1, and 22.4% at 1, 2, 3, 5, and 10 years, stages. Eighteen patients (38%) had a prior hemorrhage and 21
respectively, which translate to annual hemorrhage rates of patients (45%) underwent prior embolization. 56 The median
4.05 and 1.79% of patients developing new postrepeat-SRS interval between the first-stage SRS and the second-stage SRS
hemorrhages per year for years 0–2 and 2–10 following repeat was 4.9 months (range 2.8–13.8 months). The median target
SRS. Factors associated with a higher risk of hemorrhage after volume was 11.5 cc (range 4.0–26 cc) in the first-stage SRS and

76
Stereotactic Radiosurgery for Arteriovenous Malformations

Fig. 9.3 The graph showing higher percentage of


arteriovenous malformation obliteration rates
with higher margin doses. MR, magnetic reso-
nance.

9.5 cc in the second-stage SRS. The median margin dose was more effective for radiosurgery. However, embolization can
16 Gy (range 13–18 Gy) for both stages. In 17 patients, AVM only be effective if it permanently reduces the nidus volume.
obliteration was confirmed after two to four SRS procedures at Reduction in flow from an AVM does not provide improvement
a median follow-up of 87 months (range 0.4–209 months). Five in radiosurgical outcome data. Our analysis suggests that radio-
patients had near-total obliteration (volume reduction > 75%, surgical embolization had a negative effect on AVM obliteration
but residual AVM). The actuarial rates of total obliteration after rates.59 We analyzed the long-term benefits and risks of SRS for
two-stage SRS were 7, 20, 28, and 36% at 3, 4, 5, and 10 years, patients with AVMs who underwent prior embolization. 60 At
respectively. The 5-year total obliteration rate after the initial our institution, 120 patients underwent embolization followed
staged volumetric SRS with a margin dose of 17 Gy or more was by SRS. In this series, 64 patients (53%) had at least one prior
62% (p = 0.001). Sixteen patients underwent additional SRS at a hemorrhage. The median number of embolizations varied from
median interval of 61 months (range 33–113 months) after the one to five. The median target volume was 6.6 cc (range 0.2–
initial two-stage SRS. The overall rates of total obliteration after 26.3). The median margin dose was 18 Gy (range 13.5–25.0 Gy).
staged and repeat SRS were 18, 45, and 56% at 5, 7, and 10 After embolization, 25 patients (21%) developed symptomatic
years, respectively. Ten patients sustained hemorrhage after neurologic deficits. The overall rates of total obliteration docu-
staged SRS, and five of these patients died. Three of 16 patients mented by either angiography or MRI were 35, 53, 55, and 59%
who underwent repeat SRS sustained hemorrhage after the at 3, 4, 5, and 10 years, respectively. Factors associated with a
procedure and died. Based on Kaplan-Meier analysis (excluding higher rate of AVM obliteration were smaller target volume,
the second hemorrhage in the patient who had two hemor- smaller maximum diameter, higher margin dose, timing of
rhages), the cumulative rates of AVM hemorrhage after SRS embolization during the most recent 10-year period (1997–
were 4.3, 8.6, 13.5, and 36.0% at 1, 2, 5, and 10 years, respec- 2006), and lower Pollock-Flickinger score. Nine patients (8%)
tively. This corresponded to annual hemorrhage risks of 4.3, 2.3, had a hemorrhage during the latency period, and seven patients
and 5.6% for years 0–1, 1–5, and 5–10 after SRS. Multiple hem- died of hemorrhage. The actuarial rates of AVM hemorrhage
orrhages before SRS correlated with a significantly higher risk after SRS were 0.8, 3.5, 5.4, 7.7, and 7.7% at 1, 2, 3, 5, and 10
of hemorrhage after SRS. Symptomatic adverse radiation effects years, respectively. The overall annual hemorrhage rate was
were detected in 13% of patients, but no patient died as a result 2.7%. Factors associated with a higher risk of hemorrhage after
of an adverse radiation effect. Delayed cyst formation did not SRS were a larger target volume and a larger number of prior
occur in any patient after SRS. hemorrhages. Permanent neurologic deficits due to AREs devel-
oped in three patients (2.5%) after SRS, and one patient had
delayed cyst formation 210 months after SRS. No patient died
9.11 The Role of Preradiosurgical of AREs. A larger 12-Gy volume was associated with higher risk
Embolization of symptomatic AREs. Using a case-control–matched approach,
the authors found that patients who underwent embolization
Embolization has a major benefit in preparation for craniotomy prior to SRS had a lower rate of total obliteration (p = 0.028)
and microsurgical removal.57 Its primary benefit is reducing the than patients who had not undergone embolization (▶ Fig. 9.4).
flow through the AVM, or removal of deep-seated feeders that Others have reported that up to 30% of patients who had AVM
may be problematic for subsequent surgical removal. We used embolization subsequently had an increase in the nidus volume
embolization early in our experience for larger-volume AVMs. 58 when an angiogram was performed at the time of radiosurgical
The goal was to decrease the volume of the AVM to make it targeting.61 Twelve percent of embolized AVMs showed

77
Stereotactic Radiosurgery for Arteriovenous Malformations

Fig. 9.4 Radiosurgery dose plans of staged radiosurgery for large-volume arteriovenous malformation (AVM). The medial part of nidus was selected
for stage I treatment. A margin dose of 16 Gy was prescribed to this target (yellow line). The nidus volume for stage II radiosurgery (outlined in blue)
was also defined at the time of first radiosurgery. Dose plan of second-stage radiosurgery showing complete coverage of the remaining AVM nidus.

recanalization within a year.62 Unlike surgery that removes an and radiosurgery.65 The timing of the individual procedures
AVM nidus within a few weeks of embolization, radiosurgery is critical. In general, we prefer to do radiosurgery first in a
takes several years to be fully effective. This latency interval, newly diagnosed patient, at a time when the entire connec-
unfortunately, allows sufficient time for embolized AVM com- tion of the fistula can be defined. Most of these lesions
ponents to recanalize, remodel, or even recruit new feeding occur in the region of the transverse or sigmoid sinus, and
blood vessels. In a report using both embolization and radiosur- may be associated with pulsatile tinnitus. 66 Others may
gery, permanent neurologic deficits ranged from 5 to 12% of occur in the cavernous sinus and are associated with diplo-
patients with a mortality rate of 1.5 to 2.7% of patients. 61,62,63 pia, impaired vision, or exophthalmos. Lesions of the superi-
Complete obliteration rates varied from 47 to 55%.61,62,63 In a or sagittal sinus may cause papilledema, vision loss, and
study of 47 patients who had radiosurgery and embolization in hydrocephalus. Those lesions that have cortical venous
comparison with 47 matched patients who were treated with drainage are prone to have intracranial hemorrhages, pro-
radiosurgery alone, nidus obliteration was achieved in 47% of gressive deficits, or seizures. Because such patients have an
the embolization group, but in 70% of the radiosurgery group. 64 overall intracranial hemorrhage rate of approximately 2%
Experience using adhesive agents such as Onyx is still in its per year, management of these lesions may be very impor-
early stages; therefore, whether radiosurgery will have a signif- tant. 66 We generally recommend early radiosurgery fol-
icant improvement in patients who have previously undergone lowed the same day by embolization. Patients have their
liquid adhesive embolization is yet to be defined. frame placed and undergo MRI, followed by initial angiogra-
phy, and are transferred for Gamma Knife radiosurgery with
a femoral sheath catheter left in position. After completion
9.12 Treatment of Dural of the radiosurgical component of the procedure, they are
Arteriovenous Malformations transferred back to the interventional radiology suite where
they undergo embolization. The combination of radiosur-
In contrast to the experience with intracranial AVMs, dural gery and embolization provides both early symptom relief
vascular malformations or dural arteriovenous fistulas of the DAVF and long-term relief of the condition as fistula
(DAVFs) (▶ Fig. 9.5) may indeed benefit from embolization closure by radiosurgery.

78
Stereotactic Radiosurgery for Arteriovenous Malformations

Fig. 9.5 (a) Axial T1-weighted contrast-enhanced magnetic resonance images showing the transverse sinus dural arteriovenous fistula (AVF). (b)
Digital subtraction lateral carotid artery angiogram showing the radiosurgery dose plan covering the transverse sinus dural AVF. (c) Digital subtraction
anteroposterior carotid artery angiogram showing the radiosurgery dose plan covering the transverse sinus dural AVF.

9.13 Future Directions References


Radiosurgery of an AVM is a well-established management [1] Deruty R, Pelissou-Guyotat I, Morel C, Bascoulergue Y, Turjman F. Reflections
strategy for AVMs at the present time. With more than 25 on the management of cerebral arteriovenous malformations. Surg Neurol
1998; 50: 245–255, discussion 255–256
years of experience using the Gamma Knife technique, and
[2] Han PP, Ponce FA, Spetzler RF. Intention-to-treat analysis of Spetzler-Martin
with thousands of patients worldwide who have undergone grades IV and V arteriovenous malformations: natural history and treatment
this procedure, we understand the overall success rate, the paradigm. J Neurosurg 2003; 98: 3–7
complication rate, and the various alternative strategies. We [3] Ledezma CJ, Hoh BL, Carter BS, Pryor JC, Putman CM, Ogilvy CS. Complications
of cerebral arteriovenous malformation embolization: multivariate analysis
believe that at the present time, embolization has an impor-
of predictive factors. Neurosurgery 2006; 58: 602–611, discussion 602–611
tant role prior to microsurgery, an important role after [4] Raymond J, Iancu D, Weill A, et al. Embolization as one modality in a com-
radiosurgery for the management of DAVFs, and a relatively bined strategy for the management of cerebral arteriovenous malformations.
limited role in the prospective management of AVMs within Interv Neuroradiol 2005; 11 (Suppl 1): 57–62
the brain. Although outcome studies continue to be looked [5] Spetzler RF, Martin NA. A proposed grading system for arteriovenous malfor-
mations. J Neurosurg 1986; 65: 476–483
at both in Canada and in the United States, we suspect that
[6] Pollock BE, Flickinger JC. A proposed radiosurgery-based grading system for
most patients with an intracranial AVM require strong con- arteriovenous malformations. J Neurosurg 2002; 96: 79–85
sideration for intervention, providing that the management [7] Liscák R, Vladyka V, Simonová G, et al. Arteriovenous malformations after
strategy has an acceptable risk to the patient. It seems Leksell gamma knife radiosurgery: rate of obliteration and complications.
Neurosurgery 2007; 60: 1005–1014, discussion 1015–1016
unlikely that higher doses will be feasible. Today the ability
[8] Pollock BE, Gorman DA, Coffey RJ. Patient outcomes after arteriovenous mal-
to conform dose and to restrict dose (selectivity) seem to be formation radiosurgical management: results based on a 5- to 14-year fol-
maximized with the current imaging techniques as well as low-up study. Neurosurgery 2003; 52: 1291–1296, discussion 1296–1297
radiation delivery systems, including the new Perfexion [9] Kjellberg RN. Stereotactic Bragg peak proton beam radiosurgery for cerebral
Gamma Knife (Elekta AB). We may be able to increase the arteriovenous malformations. Ann Clin Res 1986; 18 (Suppl 47): 17–19
[10] Kjellberg RN, Hanamura T, Davis KR, Lyons SL, Adams RD. Bragg-peak proton-
chance of obliteration by developing specific radiation sen-
beam therapy for arteriovenous malformations of the brain. N Engl J Med
sitizers delivered by microcatheters immediately before 1983; 309: 269–274
radiosurgery. To date, brain-protection technologies or [11] Fabrikant JI, Levy RP, Steinberg GK, et al. Heavy-charged-particle radiosurgery
drugs have been disappointing. The greatest remaining for intracranial arteriovenous malformations. Stereotact Funct Neurosurg
issue for AVM radiosurgery is the risk of a latency interval 1991; 57: 50–63
[12] Steiner L, Leksell L, Greitz T, Forster DM, Backlund EO. Stereotaxic radiosur-
hemorrhage, so efforts to reduce the latency interval would
gery for cerebral arteriovenous malformations. Report of a case. Acta Chir
be important. The recognition of late long-term side effects Scand 1972; 138: 459–464
including the possible risk of radiation-related tumors or [13] Betti OO, Munari C, Rosler R. Stereotactic radiosurgery with the linear accel-
late cyst development continues to be evaluated. Within the erator: treatment of arteriovenous malformations. Neurosurgery 1989; 24:
311–321
context of a disease that has a 1% annual mortality left
[14] Barcia-Salorio JL, Barcia JA, Soler F, Hernández G, Genovés JM. Stereotactic ra-
untreated, the current data have shown that SRS is a safe diotherapy plus radiosurgical boost in the treatment of large cerebral arterio-
and effective management strategy that can be applied to venous malformations. Acta Neurochir Suppl (Wien) 1993; 58: 98–100
both lobar as well as deep-seated AVMs. Many AVM patients [15] Barcia-Salorio JL, Soler F, Hernandez G, Barcia JA. Radiosurgical treatment of
were never treatable by any technique prior to the develop- low flow carotid-cavernous fistulae. Acta Neurochir Suppl (Wien) 1991; 52:
93–95
ment of radiosurgery.
[16] Colombo F, Benedetti A, Pozza F, Marchetti C, Chierego G. Linear accelerator
Disclaimer: Dr. Lunsford is a stockholder and consultant with radiosurgery of cerebral arteriovenous malformations. Neurosurgery 1989;
Elekta AB, Stockholm, Sweden. 24: 833–840

79
Stereotactic Radiosurgery for Arteriovenous Malformations

[17] Colombo F, Cavedon C, Francescon P, et al. Three-dimensional angiography [41] Flickinger JC, Kondziolka D, Maitz AH, Lunsford LD. Analysis of neurological
for radiosurgical treatment planning for arteriovenous malformations. J Neu- sequelae from radiosurgery of arteriovenous malformations: how location af-
rosurg 2003; 98: 536–543 fects outcome. Int J Radiat Oncol Biol Phys 1998; 40: 273–278
[18] Colombo F, Pozza F, Chierego G, Casentini L, De Luca G, Francescon P. Linear [42] Flickinger JC, Kondziolka D, Pollock BE, Maitz AH, Lunsford LD. Complications
accelerator radiosurgery of cerebral arteriovenous malformations: an update. from arteriovenous malformation radiosurgery: multivariate analysis and
Neurosurgery 1994; 34: 14–20, discussion 20–21 risk modeling. Int J Radiat Oncol Biol Phys 1997; 38: 485–490
[19] Loeffler JS, Alexander E III, Siddon RL, Saunders WM, Coleman CN, Winston [43] Flickinger JC, Kondziolka D, Lunsford LD, et al. A multi-institutional analysis
KR. Stereotactic radiosurgery for intracranial arteriovenous malformations of complication outcomes after arteriovenous malformation radiosurgery. Int
using a standard linear accelerator. Int J Radiat Oncol Biol Phys 1989; 17: J Radiat Oncol Biol Phys 1999; 44: 67–74
673–677 [44] Hara M, Nakamura M, Shiokawa Y, et al. Delayed cyst formation after radio-
[20] Friedman WA, Bova FJ, Mendenhall WM. Linear accelerator radiosurgery for surgery for cerebral arteriovenous malformation: two case reports. Minim In-
arteriovenous malformations: the relationship of size to outcome. J Neuro- vasive Neurosurg 1998; 41: 40–45
surg 1995; 82: 180–189 [45] Izawa M, Hayashi M, Chernov M, et al. Long-term complications after gamma
[21] Altschuler EM, Lunsford LD, Coffey RJ, Bissonette DJ, Flickinger JC. Gamma knife surgery for arteriovenous malformations. J Neurosurg 2005; 102
knife radiosurgery for intracranial arteriovenous malformations in childhood (Suppl): 34–37
and adolescence. Pediatr Neurosci 1989; 15: 53–61 [46] Cahan WG, Woodard HQ, Higinbotham NL, Stewart FW, Coley BL. Sarcoma
[22] Lunsford LD, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery arising in irradiated bone: Report of 11 cases. Cancer 1948; 1: 3–29
for arteriovenous malformations of the brain. J Neurosurg 1991; 75: 512– [47] Pan HC, Sheehan J, Stroila M, Steiner M, Steiner L. Late cyst formation follow-
524 ing gamma knife surgery of arteriovenous malformations. J Neurosurg 2005;
[23] Kano H, Lunsford LD, Flickinger JC, et al. Stereotactic radiosurgery for arterio- 102 (Suppl): 124–127
venous malformations, Part 1: management of Spetzler-Martin Grade I and II [48] Sheehan J, Yen CP, Steiner L. Gamma knife surgery-induced meningioma. Report
arteriovenous malformations. J Neurosurg 2012; 116: 11–20 of two cases and review of the literature. J Neurosurg 2006; 105: 325–329
[24] Kano H, Flickinger JC, Yang HC, et al. Stereotactic radiosurgery for Spet- [49] Berman EL, Eade TN, Brown D, et al. Radiation-induced tumor after stereotac-
zler-Martin Grade III arteriovenous malformations. J Neurosurg 2014; tic radiosurgery for an arteriovenous malformation: case report. Neurosur-
120: 973–981 gery 2007; 61: E1099–, discussion E1099discussion E
[25] Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arte- [50] Kaido T, Hoshida T, Uranishi R, et al. Radiosurgery-induced brain tumor. Case
riovenous malformations, part 2: management of pediatric patients. J Neuro- report. J Neurosurg 2001; 95: 710–713
surg Pediatr 2012; 9: 1–10 [51] Rowe J, Grainger A, Walton L, Silcocks P, Radatz M, Kemeny A. Risk of malig-
[26] Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arte- nancy after gamma knife stereotactic radiosurgery. Neurosurgery 2007; 60:
riovenous malformations, Part 4: management of basal ganglia and thalamus 60–65, discussion 65–66
arteriovenous malformations. J Neurosurg 2012; 116: 33–43 [52] Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arte-
[27] Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arte- riovenous malformations, Part 3: outcome predictors and risks after repeat
riovenous malformations, Part 5: management of brainstem arteriovenous radiosurgery. J Neurosurg 2012; 116: 21–32
malformations. J Neurosurg 2012; 116: 44–53 [53] Maesawa S, Flickinger JC, Kondziolka D, Lunsford LD. Repeated radiosurgery
[28] Bowden G, Kano H, Tonetti D, Niranjan A, Flickinger J, Lunsford LD. Stereotac- for incompletely obliterated arteriovenous malformations. J Neurosurg 2000;
tic radiosurgery for arteriovenous malformations of the cerebellum. J Neuro- 92: 961–970
surg 2014; 120: 583–590 [54] Karlsson B, Lindqvist M, Blomgren H, et al. Long-term results after fractio-
[29] Bowden G, Kano H, Tonetti D, et al. Stereotactic radiosurgery for sylvian fis- nated radiation therapy for large brain arteriovenous malformations. Neuro-
sure arteriovenous malformations with emphasis on hemorrhage risks and surgery 2005; 57: 42–49, discussion 42–49
seizure outcomes. J Neurosurg 2014; 121: 637–644 [55] Pan DH, Guo WY, Chung WY, Shiau CY, Chang YC, Wang LW. Gamma knife
[30] Bowden G, Kano H, Yang HC, Niranjan A, Flickinger J, Lunsford LD. Gamma radiosurgery as a single treatment modality for large cerebral arteriovenous
Knife surgery for arteriovenous malformations within or adjacent to the ven- malformations. J Neurosurg 2000; 93 (Suppl 3): 113–119
tricles. J Neurosurg 2014; 121: 1416–1423. [56] Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arte-
[31] Ondra SL, Troupp H, George ED, Schwab K. The natural history of sympto- riovenous malformations, Part 6: multistaged volumetric management of
matic arteriovenous malformations of the brain: a 24-year follow-up assess- large arteriovenous malformations. J Neurosurg 2012; 116: 54–65
ment. J Neurosurg 1990; 73: 387–391 [57] Mathis JA, Barr JD, Horton JA, et al. The efficacy of particulate embolization
[32] Pollock BE, Flickinger JC, Lunsford LD, Bissonette DJ, Kondziolka D. Factors combined with stereotactic radiosurgery for treatment of large arteriovenous
that predict the bleeding risk of cerebral arteriovenous malformations. Stroke malformations of the brain. AJNR Am J Neuroradiol 1995; 16: 299–306
1996; 27: 1–6 [58] Dawson RC III, Tarr RW, Hecht ST, et al. Treatment of arteriovenous malfor-
[33] Brown RD Jr. Simple risk predictions for arteriovenous malformation hemor- mations of the brain with combined embolization and stereotactic radiosur-
rhage. Neurosurgery 2000; 46: 1024 gery: results after 1 and 2 years. AJNR Am J Neuroradiol 1990; 11: 857–864
[34] Kondziolka D, McLaughlin MR, Kestle JR. Simple risk predictions for arterio- [59] Pollock BE, Flickinger JC, Lunsford LD, Maitz A, Kondziolka D. Factors associ-
venous malformation hemorrhage. Neurosurgery 1995; 37: 851–855 ated with successful arteriovenous malformation radiosurgery. Neurosurgery
[35] Pollock BE, Flickinger JC, Lunsford LD, Bissonette DJ, Kondziolka D. Hemor- 1998; 42: 1239–1244, discussion 1244–1247
rhage risk after stereotactic radiosurgery of cerebral arteriovenous malforma- [60] Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arte-
tions. Neurosurgery 1996; 38: 652–659, discussion 659–661 riovenous malformations after embolization: a case-control study. J Neuro-
[36] Maruyama K, Shin M, Tago M, Kishimoto J, Morita A, Kawahara N. Radiosur- surg 2012; 117: 265–275
gery to reduce the risk of first hemorrhage from brain arteriovenous malfor- [61] Miyachi S, Negoro M, Okamoto T, et al. Embolisation of cerebral arteriovenous
mations. Neurosurgery 2007; 60: 453–458, discussion 458–459 malformations to assure successful subsequent radiosurgery. J Clin Neurosci
[37] Karlsson B, Lax I, Söderman M. Risk for hemorrhage during the 2-year latency 2000; 7 (Suppl 1): 82–85
period following gamma knife radiosurgery for arteriovenous malformations. [62] Gobin YP, Laurent A, Merienne L, et al. Treatment of brain arteriovenous mal-
Int J Radiat Oncol Biol Phys 2001; 49: 1045–1051 formations by embolization and radiosurgery. J Neurosurg 1996; 85: 19–28
[38] Kano H, Kondziolka D, Flickinger JC, et al. Aneurysms increase the risk of re- [63] Henkes H, Nahser HC, Berg-Dammer E, Weber W, Lange S, Kühne D. Endovascu-
bleeding after stereotactic radiosurgery for hemorrhagic arteriovenous mal- lar therapy of brain AVMs prior to radiosurgery. Neurol Res 1998; 20: 479–492
formations. Stroke 2012; 43: 2586–2591 [64] Andrade-Souza YM, Ramani M, Scora D, Tsao MN, terBrugge K, Schwartz ML.
[39] Pollock BE, Kondziolka D, Flickinger JC, Patel AK, Bissonette DJ, Lunsford Embolization before radiosurgery reduces the obliteration rate of arteriove-
LD. Magnetic resonance imaging: an accurate method to evaluate arterio- nous malformations. Neurosurgery 2007; 60: 443–451, discussion 451–452
venous malformations after stereotactic radiosurgery. J Neurosurg 1996; [65] Houser OW, Campbell JK, Campbell RJ, Sundt TM Jr. Arteriovenous malforma-
85: 1044–1049 tion affecting the transverse dural venous sinus—an acquired lesion. Mayo
[40] Flickinger JC, Kondziolka D, Lunsford LD, et al. Arteriovenous Malformation Clin Proc 1979; 54: 651–661
Radiosurgery Study Group. Development of a model to predict permanent [66] Awad IA, Little JR, Akarawi WP, Ahl J. Intracranial dural arteriovenous malfor-
symptomatic postradiosurgery injury for arteriovenous malformation pa- mations: factors predisposing to an aggressive neurological course. J Neuro-
tients. Int J Radiat Oncol Biol Phys 2000; 46: 1143–1148 surg 1990; 72: 839–850

80
Stereotactic Radiosurgery for Cavernomas

10 Stereotactic Radiosurgery for Cavernomas


Gabor Nagy and Andras A. Kemeny

obliteration after SRS.4 Since then, increasing worldwide clinical


Key Points experience together with documented histopathological cases
seem to support the initial intuition: SRS is recommended as a
● The natural history of cerebral cavernomas is varied accord- treatment option for surgically inaccessible CCM with repeated
ing to their anatomical location. Many are silent and the risk hemorrhages.5 Moreover, a more proactive attitude, recom-
of persisting disability after one bleed is low for hemispheric mending SRS early soon after the first presentation, has
lesions, whereas those found in the thalamus, basal ganglia, recently been proposed by some large centers to avoid the step-
and brainstem typically behave more aggressively with in- wise neurologic deterioration caused by repeated hemorrhages
creased rebleed risk resulting in higher cumulative morbidity of deep-seated lesions in light of the safety of modern radiosur-
following subsequent hemorrhages. However, despite our gical techniques.6,7
increasing knowledge on natural history, currently we are However, the skepticism about the effectiveness and safety of
unable to predict future behavior of an individual lesion at SRS that dominated views among neurovascular experts even
presentation. 10 years ago8 still exists.9,10 Admittedly, there is no reliable
● At present, there is no high-quality evidence to define the radiological measure (“end point”) currently available to dem-
relative roles of microsurgery, stereotactic radiosurgery, and onstrate cure; thus, the proponents’ argument is based only on
wait-and-watch policy in the management of detected observational statistics, whereas as in all interventions, even
cerebral cavernomas. Although their indications are partly SRS carries the risk of morbidity. On the other hand, one is also
overlapping, their role in the management of cerebral caver- aware of the disadvantages of microsurgery, from surgical com-
nomas is rather complementary. plications to persisting risk of bleed after surgery. 11,12 Therefore,
● Stereotactic radiosurgery using current protocols appears to should SRS be considered a viable alternative for microsurgery
be an effective treatment alternative for deep-seated cere- for symptomatic cases in terms of efficacy and for wait-and-
bral cavernomas with multiple hemorrhages—reducing pre- watch policy in a neurologically intact patient in terms of
treatment annual rebleed rates from about 30% to near zero safety? By reviewing SRS literature recently, we concluded that
within 2 years after treatment. It is also effective for cerebral the quality of published articles was disappointingly heteroge-
cavernomas causing therapy-resistant epilepsy, especially if nous and recommended standard data collection and treatment
applied early—within 3 years after presentation. In modern criteria based on our current knowledge on natural history and
stereotactic radiosurgical series radiation only induces a low modern SRS techniques.7 In this chapter, after a brief summary
rate of mild morbidity. of natural history, we critically review the literature of CCM SRS
● We recommend early stereotactic radiosurgery soon after based on our standard criteria addressing the issues frequently
presentation in neurologically intact or minimally disabled raised by critics of SRS and arguing for the safety and effective-
patients harboring deep-seated cerebral cavernomas. In our ness of modern CCM SRS.
opinion, waiting for the cumulative morbidity of the natural
history to justify an otherwise low-risk intervention does not
serve the patient well. 10.2 Natural History of
Cavernomas
10.1 Introduction Cerebral cavernomas with distinct pathological and magnetic
resonance imaging (MRI) characteristics (▶ Table 10.1) com-
Owing to earlier and more reliable radiological detection, as well pose a large proportion of the previously described angiograph-
as to our better understanding of their natural history, manage- ically occult vascular malformations (AOVMs) (▶ Fig. 10.1).13,14,
ment strategy of cerebral cavernomas (CCMs; also known as 15 Their estimated prevalence is 0.15 to 0.9%16,17,18; 76% of them

cavernous malformations, cavernous angiomas, or cavernous are located supratentorially, 8% in the basal ganglia/thalamus,
hemangiomas) has significantly changed during the last two and 18% in the brainstem. Multiple CCMs are found in 19% of
decades.1,2 However, due to the lack of high-quality evidence, the patients,19 more frequently in familiar forms constituting at
their optimal management is still debated. Consensus seems to least 6% of all cases.20 A contemporary population-based pro-
exist in the treatment of hemispheric (superficial) CCMs: micro- spective study detected approximately 6 cases/million/year in
surgery for symptomatic and observation for incidental lesions. Scotland.21 Forty-seven to 60% of the cases are asymptomatic at
Deep and eloquent lesions in the brainstem, thalamus, and basal detection, and only 9.3% of the lesions initially found inciden-
ganglia, especially those after one or no hemorrhage, pose a tally or presented with seizures go on to cause hemorrhage or
dilemma because they seem to behave more aggressively if focal neurologic deficit within 5 years, but the risk of a second
untreated, whereas any intervention would carry a higher risk.3 event increases to 42.4%.22 When patients become sympto-
Stereotactic radiosurgery (SRS) was introduced for the treat- matic, typically in their 30 s, 37% present with seizures, 36%
ment of CCMs based on the assumption that their pathological with hemorrhage, 23% with headaches, and 22% with focal neu-
vessels would respond similarly to arteriovenous malforma- rologic deficits.19 Twenty-one to 33% of CCMs is associated with
tions (AVMs), which are known to undergo thrombo- developmental venous anomalies (DVAs) in adults20,23 that are

81
Stereotactic Radiosurgery for Cavernomas

Table 10.1 MRI appearance of CCMs according to time of bleed and classification
MRI appearance Pathological characteristics

Hyperacute (< 24 h) T1: Iso- or hyperintense


T2: Iso- or hyperintense center, hypointense periphery,
hyperintense rim

Acute (1–3 d) T1: Iso- or hypointense


T2: Hypointense

Subacute—Type I Subacute bleed, surrounded by a rim of hemosiderin-


Early (3–7 d) T1: Hyperintense stained macrophages and gliosis
T2: Hypointense
Late (7–14 d) T1: Hyperintense
T2: Hyperintense

Chronic (> 14 d) or unbled


Type II T1: Reticulated mixed signal Loculated areas of bleed and thrombosis of different age,
T2: Reticulated mixed signal, surrounded by hypointense surrounded by gliotic and hemosiderin-stained brain, may
rim be areas of calcification in larger lesions
GE: Reticular core, with hypointense magnifying rim
Type III T1: Iso- or hypointense Resolved hemorrhage, surrounded by hemosiderin-stained
T2: Hypointense with hypointense rim magnifying lesion size gliotic brain
GE: Hypointense, magnifying greater than T2
T1: Not or poorly visible May be telangiectasia
Type IV T2: Not or poorly visible
GE: Punctate hypointense

Abbreviations: CCMs, cerebral cavernomas; MRI, magnetic resonance imaging.

thought to promote CCM formation20 and to increase the risk of observational studies, with only few exceptions.31,33 Prospective
hemorrhage (▶ Fig. 10.1).24 studies estimated rebleed rate between 4.5 and 22.9%/y.32,34 The
cumulative incidence of rebleed was found 56% after 5 years and
72% after 10 years.35 Moreover, even lesions that presented with
10.2.1 Definition of Clinical nonhemorrhagic symptoms have been found to have higher risk
Hemorrhage of bleeding than incidental lesions (2.18 and 0.33%/y, respec-
tively).23 Several studies suggest that increased rebleed risk is
The key factor influencing CCM management is the definition of
time limited and decreases a few years after the first hemor-
clinical hemorrhage, as the first clinical bleed seems to have
rhage (“temporal clustering”).23,35,36 However, even if this were
major impact on later behavior of the lesion.25 This definition is
the case, risk of rebleeding seems to be increased for at least 5
far from obvious. Not all clinical events (acute neurologic deteri-
years after the initial bleed.37,38 Brainstem and thalamic/basal
oration) are associated with evidence of concurrent hemor-
ganglia CCM are generally observed to have higher initial and
rhage,26 whereas hemosiderin ring is almost always present
rebleed rates (2.3 to 6.8 and 21 to 60%, respectively).11,39 It is not
even in clinically silent cases. The latter is explained by ultra-
clear, however, whether deep-seated CCM are more prone to
structural studies that suggest a compromised blood–brain bar-
bleeding, or whether any bleed is more likely to be symptomatic
rier at the site of a CCM that may lead to a chronic erythrocyte
due to higher functional density of deep eloquent structures.
leak into the surrounding brain and consequently to deposition
For therapeutic decision making, it is fundamental to note
of hemosiderin even in the absence of clinically significant hem-
the difference between first and repeated hemorrhages and
orrhage.27 In our opinion, the most comprehensive and clinically
also the distinct behavior of superficial and deep-seated lesions.
useful definition is described by Al-Shahi Salman et al: A hem-
Moreover, it is also possible that there are even more distinct
orrhage is a clinical event with acute or subacute onset symp-
subpopulations, some lesions behaving aggressively with a high
toms with radiological, pathological, surgical, or cerebrospinal
risk of rebleeding temporarily or for a much longer period after
fluid evidence of recent extra- or intralesional hemorrhage,
a first hemorrhage, whereas others are more quiescent. If this is
whereas the mere existence of a hemosiderin ring or the sole
the case, the proportion of more unstable lesions is unknown
increase in diameter are not considered as hemorrhage.15
and currently we are unable to predict from a first bleed which
pattern of behavior a CCM would follow.
10.2.2 Risk of Hemorrhage
Despite evidence of de novo CCM formation,14,28 retrospective
studies assuming lesion presence since birth gave similar esti-
10.2.3 Morbidity of Hemorrhage
mates for first-ever hemorrhage rates as prospective studies, 0.1 Morbidity of superficial hemispheric CCM after a bleed usually
to 2.7%/lesion/y16,17,29,30,31 and 0.25 to 3.1%/person/y.16,23,26,29,30, manifests in epilepsy and only rarely in focal neurologic deficit,40,
31,32 A first, bleed may destabilize a CCM and increase the risk of 41 whereas deep eloquent lesions have a higher rate of permanent

further bleeding, which is supported by the majority of focal neurologic morbidity and mortality.34,40,41,42 A single bleed

82
Stereotactic Radiosurgery for Cavernomas

Fig. 10.1 (a) T1- and (b) T2-weighted axial


magnetic resonance images (MRIs) of a dorsal
ponto-mesencephalic cerebral cavernoma (CCM)
with evidence of recent hematoma and associ-
ated developmental venous anomalies (DVAs)
(arrow). T1-weighted (c) coronal and (d) sagittal
MRIs of the same DVA (arrow). (e) Anteroposte-
rior and (f) lateral views of the same DVA during
late venous phase of digital substraction angiog-
raphy. Angiographically occult CCM is not visible
(black dotted circle).

from a deep eloquent CCM leads to persisting neurologic deficit due to the low chance of causing persisting morbidity. For
in up to 40 to 60%, with a substantial risk of mortality,29,34,38,43 deep-seated lesions, surgical removal is generally recom-
and the chance for permanent disability cumulatively increases mended only in limited circumstances in experienced hands.
with each subsequent bleeding episode.38,42 Lesions should either reach the pial or ependymal surface or
should be approachable through a noneloquent surgical corri-
dor, and also have a history of repeated hemorrhages with pro-
10.3 Treatment Options for gressive neurologic deficit or cause significant mass effect. 36,45,
46,47 Surgery also has an adjuvant role in managing obstructive
Cavernomas hydrocephalus caused by growing or hemorrhagic CCM located
Three management options can be considered for a CCM: to highly eloquent regions when the symptoms arise from
microsurgical removal, SRS, and observation. Surgery for symp- raised intracranial pressure rather than from focal neurologic
tomatic superficial hemispheric CCM is usually safe and effec- deficit (▶ Fig. 10.2). Recent meta-analyses performed on surgi-
tive,44 whereas incidental hemispheric lesions can be observed cal series for brainstem or thalamus/basal ganglia CCM found

83
Stereotactic Radiosurgery for Cavernomas

Fig. 10.2 Computed tomography scan of the


same patient as in ▶ Fig. 10.1. (a) 2.5 weeks after
presentation demonstrating occlusive hydroce-
phalus. (b) Due to symptoms of raised intra-
cranial pressure, a third ventriculostomy was
performed resulting in improved clinical and
radiological states. Of note, at this stage micro-
surgical removal of cerebral cavernoma was not
necessary, as the primary problem was occlusive
hydrocephalus and not progressive focal neuro-
logic deficits due to brainstem compression. In
case of growing or clinical progression, micro-
surgery is the treatment of choice. However, in
case of hematoma resolution, stereotactic radio-
surgery is a good alternative for microsurgery in
this otherwise clinically intact patient.

10 to 14% persisting morbidity and 1.5 to 1.9% mortality, with actually show complete response, were they removed for analy-
an 89 to 91% resection rate. Importantly, the rebleed rate never sis. Alternatively, the scarring of the wall of such a low-pressure
goes to zero even after microsurgical removal due to the pro- lesion may sufficiently stabilize it to prevent a rebleed even
portion of residual lesions. Sixty-two percent of partially without full obliteration.
resected lesions rebled with an annual rate of 0.5 to 2.0% and
with 6% mortality.11,12 Thus, though an effective salvage treat-
ment, surgical removal as a prophylactic measure in patients
The Effect of Radiosurgery on Hemorrhage Rate
harboring deep-seated CCM with no or minimal neurologic def- The primary aim of SRS is to reduce the risk of future hemor-
icit is rarely considered.48,49 Thus, the question for these lesions rhage and consequential neurologic deterioration. Indeed, this
is whether to observe or treat them with SRS.7,10 Currently, is reported by the majority of SRS studies comparing pre- and
there is no consensus for the role of these three modalities in posttreatment bleed rates beyond a latency of 2 to 3 years. 7 The
CCM management. It is important to note that despite the over- first report of a large clinical series using a modern SRS techni-
lap between indications,2,3 the three are not competitive but que with sufficient follow-up time was published in 1995. 4 It
complement each other, and a decision should be made on an was found that the rebleed rate fell from 32%/patient/y pre-
individual basis taking into account not only CCM location and treatment to 8.8%/patient/y within the first 2 years after treat-
behavior, but the age and medical condition of the patient. ment and to 1.1%/patient/y thereafter. Recent updates from this
Moreover, the final treatment decision is also influenced by group including more patients and longer follow-up time 55 and
neurosurgeon’s experience and the preference of the fully other contemporary SRS series focusing on deep eloquent
informed patient. lesions,38,56,57,58,59,60 and studies analyzing mixed population of
lobar and deep CCM, confirmed this finding.41,61,62

10.3.1 Radiosurgery for Cavernomas


The Effect of Radiosurgery on Epilepsy
Rationale for Cavernoma Radiosurgery Although for patients with intractable epilepsy from a CCM,
It is well documented that radiation induces hyalinization and surgical resection of hemispheric CCM is the treatment of
thickening of the wall of the endothelium-lined pathological choice, as it is associated with low morbidity and high effective-
vessels of an AVM.50 The idea to use SRS for an AOVM was ini- ness.44 However, several conditions may warrant treatment
tially based on the assumption that the majority of these lesions alternatives, including eloquent location lesions, the patient’s
were partially thrombosed AVM; therefore, the vessels would medical condition, or the patient’s preference. For these cases,
be further obliterated by high-dose focused radiation, as SRS may be a real alternative. A retrospective multicenter study
observed in the pathological vessels of a true AVM. 51 Despite demonstrated that 53% of the patients suffering from long-last-
later histopathological studies finding most AOVM to be CCM,13 ing epilepsy refractory to medical therapy became seizure-free
early clinical studies found that these lesions had responded in (Engel classes IA and B) within a mean of 4 months after SRS;
a similar timescale to a true AVM, with reduction of rebleed 20% showed significant (class II) and only 26% little or no
rate within a 2-year latency period after treatment.4 Moreover, improvement (classes III and IV).63 Patients with CCM located in
histological studies of surgically resected previously irradiated the mesial temporal lobe did worse than those treated in other
CCM showed similar radiation-induced vasculopathy as seen in locations. More recent studies found similar results: 39 to 54%
AVM: fibrinoid necrosis, endothelial destruction, hyalinization, of the patients became seizure-free (class I), and 14 to 20.5%
marked fibrosis, and scar tissue formation.52,53,54 Most but not improved significantly (class II).62,64 A meta-analysis found that
all vessels were obliterated; there were also signs of neovascu- 31% of the patients became seizure-free and 35% improved sig-
larization similar to a subtotally obliterated AVM.54 Of note, nificantly after SRS.65 In the first instance, this seems inferior to
these specimens came from lesions that remained symptomatic surgical series, as class I response was achieved in 69% of the
after radiation, and those rendered silent by the treatment may surgical cases refractory to previous medical therapy. 66

84
Stereotactic Radiosurgery for Cavernomas

However, considering preintervention seizure duration, SRS lesions expected in a mixed population of CCM with more
seems to be as effective as surgery if applied early after seizure aggressive and benign nature. Importantly, the rate of further
onset. Although 90% of the patients treated with SRS improved bleed after the 2-year latency period is minimal even in this
with short history of epilepsy (≤ 3 years) and only 38.5% with population. Recent debate on the effect of SRS on CCM hemor-
longer lasting epileptic disease,64 81% improved with ≤ 1-year rhage rate is centered on whether the decrease of the rebleed
history and 70% with longer duration of epilepsy in the surgical rate within 2 years can really be attributed to the radiobiologi-
group.66 cal effect of the treatment or whether it simply reflects natural
history, as several observational studies raised the idea that
hemorrhages may occur in clusters.23,35,36 The contrary argu-
Critical Considerations in Cavernoma
ment is supported by the observations that lesions with long
Radiosurgery pretreatment history of repeated hemorrhages seem to main-
Unlike for an AVM, there is currently no radiological measure tain a high rate of hemorrhagic events for many years.37,38
for CCM cure after SRS, which certainly keeps skepticism alive. Another supporting argument is that the time course of reduc-
These lesions are angiographically occult, and MRI generally tion in rebleed rates after SRS parallels the time course of histo-
fails to demonstrate a definite change in the appearance of the logical changes known to develop after irradiation. Thus,
lesion after SRS. Although only a few of the irradiated CCM coincidence of the marked fall in rebleed rate within 2 years
enlarge41,56,67,68 and the proportion of true growth is somewhat after SRS with the natural history seems unlikely. As current
higher in an untreated population,14,28 CCMs after SRS appear radiosurgical literature lacks untreated control group, 1 this
heterogenous on MRI. Approximately half of the lesions debate will be speculative until such data arrive.
shrank,55,67 but postradiosurgery shrinkage may in part be due Early studies, often cited by critics of SRS, reported high radi-
to resolution of intralesional hematomas. Thus, it is impossible ation-associated complication rates. However, those studies
to tell from an individual lesion whether it is secured from fur- were from an era with poor delineation of the target with com-
ther bleed or not. puted tomography (CT) or less conformal MRI, and with the use
Due to the lack of an exact measure for cure, a prospective of higher-dose protocols.53,60,69,71 Clearly, these studies repre-
randomized controlled trial to clarify the conflicting issues sur- sent the early experimental phase of the collective learning
rounding SRS may appear attractive. However, it is unlikely to curve of CCM SRS. Contemporary treatment protocols, particu-
realize such a trial in the near future for numerous reasons, par- larly those using a gamma radiation–based instrument, include
ticularly due to the widely different immediate impact of the prescription dose less than 20 Gy (typically 12–15 Gy), highly
three options that would limit enrollment. We have therefore conformal MRI-based treatment planning, and without evi-
recently suggested a prospective international data collection dence of recent bleed (type II or III,14 at least 3 months after last
including all detected cases regardless of subsequent choice of hemorrhage) (▶ Fig. 10.3). The lesion is defined strictly within
management modality.7 Until such data arrive to answer critical the hemosiderin ring that is speculated to be a radiosensi-
voices addressing both effectiveness and safety of CCM SRS, it is tizer72; an associated DVA is preserved similarly to microsur-
crucial to define standard data collection and modern SRS treat- gery,43 as irradiating a DVA is associated with a high rate of
ment protocol. complication.73 Modern studies applying such treatment proto-
Although most of the published literature demonstrated a cols have reported low rates of adverse radiation effects result-
reduction in the rebleed rate of CCM with multiple hemor- ing in only low rate of mild persisting morbidity, and mortality
rhages after a 2-year latency period following SRS, this is not a related to the treated lesion was minimal.7
universal finding: A temporary increase in rebleed rate was also Utilization of SRS for this condition is expanding. Since the
reported,38,67,69 as well as reduction of rebleed rate after a introduction of modern CCM SRS in 1995, we treated 314
latency period longer than 2 years.53 We have alluded to the dif- lesions in 285 patients at the National Centre for Stereotactic
ficulties interpreting hemorrhagic events, particularly when Radiosurgery in Sheffield until 2012. Of these, 191 lesions were
attempts are made to account for prediagnosis clinical events; deep seated. Reflecting our aim to move toward earlier treat-
these exceptions may reflect the varied interpretation of what ment, numbers increased from an annual 3 to 4 treatments
counts as a hemorrhage and may well be due to different pa- before 2000 up to an average of 12 between 2000 until 2011,
tient selection. The key for proper interpretation of CCM SRS is and to 26 in 2012.7 When publishing our data in 2010, we had
the understanding of their natural history—to distinguish treated 46% of deep-seated CCM presenting in our catchment
between the risk of first and repeated hemorrhages, and to take area with SRS, but estimated to treat only 10 to 30% of newly
the anatomical position into account because lobar CCMs have diagnosed such lesions in the UK. We concluded that the geo-
a much lower annual incidence both of bleed and rebleed than graphical difference represented a reluctance to refer these
deep eloquent lesions. Analyzing mainly55 or exclusively38,57,58, lesions for SRS, owing to the perceived conflicting evidence on
70 results of SRS for deep eloquent CCM that had bled at least its safety and effectiveness.38 The fact that the number of
twice prior to treatment (i.e., proven to behave more aggres- treated deep-seated CCM almost doubled since 2010 suggests
sively), we find consequently a sharp decrease in the annual that the argument for early SRS treatment is winning at least in
rebleed rate from 30.5 to 32.5% to 0 to 2.4% within 2 years after the clinical practice in the UK.
SRS. When confining analysis exclusively to SRS for CCM that
had bled no more than once prior to treatment, during the first Critical Review of Radiosurgical Literature
2 years after SRS a higher rate of hemorrhage may be found
when compared with first ever hemorrhage rate.38 However, We have recently suggested standard data collection criteria to
this is still much lower than the rebleed rate of untreated the radiosurgical community, as the lack of quality evidence

85
Stereotactic Radiosurgery for Cavernomas

Fig. 10.3 (a) T1-weighted and (b) T2-weighted


axial, coronal, and sagittal, as well as (c) treat-
ment planning on proton density-weighted
magnetic resonance images (MRIs) of a type II14
right thalamic cerebral cavernoma (CCM) at the
time of stereotactic radiosurgery. (d) Treatment
planning T2-weighted MRI of a type II CCM
located in the left midbrain tegmentum. Red line:
lesion definition. Yellow line: 50% (prescription)
isodose line. Green line: 30% isodose line.

makes it currently impossible to lay down a universal indication should be reported separately to annual rebleed rate until treat-
algorithm for SRS in the management of CCM.7 Moreover, the ment. Hemorrhage rates should be calculated separately within
disappointingly heterogenous quality of literature on CCM SRS 2 years posttreatment and thereafter. In our view, it is crucial to
with distinct measures of natural history, posttreatment bleed offer separate analysis for superficial and deep-seated lesions,
rates, and morbidity provides ammunition for the critics who and similarly for lesions with 0, 1, or multiple bleeds due to
often cite early experimental and low-quality contemporary their disparate natural history. Although causal relationship
studies with predilection to support their negative view.2,9,10 with SRS is not proven for all cases, all lasting neurologic deteri-
Improved data collection with standardized reporting and oration unrelated to a posttreatment hemorrhage should be
treatment criteria seems to be the most realistic way to obtain considered as an adverse radiation effect to determine the max-
a better view on safety and effectiveness until high-level evi- imal potential morbidity of SRS. Due to delayed protection that
dence becomes available. is specific to this treatment modality, morbidity related to post-
First, the definition of clinical hemorrhage should be standar- treatment hemorrhage should also be recorded accurately.
dized in all future studies published in the CCM field, 15 leading With contemporary treatment protocol, a gamma radiation–
to a clear distinction between hemorrhagic and nonhemorrha- based instrument appears to be the most precise SRS treatment
gic clinical events, helping their separate documentation. 38 Ret- due to the highest conformity achieved with multiple isocen-
rospective annual first bleed rate for treated lesions (lesion/y) ters. The largest experience has been accumulated worldwide

86
Stereotactic Radiosurgery for Cavernomas

with Gamma Knife SRS over the last decades, but a Rotating studies represented early attempts, with poor definition of natu-
Gamma System also seems to be effective.41 ral history, selection criteria, and follow-up. A detailed extensive
A few systemic reviews dealing with CCM SRS have been pub- meta-analysis pooled all available SRS studies published until
lished; none of them meet the above criteria because they typi- 2009 without distinction on natural history, anatomical location,
cally pooled studies of heterogenous quality. Surgical reviews and SRS technique.65 As large, modern SRS series has been pub-
usually refer to early reports, thus supporting a negative view.8, lished since then: This study unavoidably underestimates its
11,39 Similar to all therapeutic interventions, SRS also had its effectiveness with overestimation of its morbidity. The other
technical evolution, and using early experimental studies as study specifically focused on SRS of brainstem CCMs, analyzing
arguments against SRS is similar to quoting poor surgical results five series.74 Of these, only three would meet strict methodical
from a premicroscopic era as arguments against surgery. A criteria of data analysis based on our current knowledge of
recent systemic review applying criteria of modern evidence- natural history and of modern CCM SRS techniques.57,58,70
based medicine found only one study comparing SRS with sur- ▶ Table 10.2 summarizes data from the most relevant contem-
gery and one comparing SRS with observation.1 However, both porary SRS series using our recently suggested criteria, focusing

Table 10.2 Summary of CCM series using modern gamma radiation–based stereotactic radiosurgery
Study Patients/lesions Deep Superficial Pre- Pre-TRT Post- Post- Permanent Post-TRT Mortality Treatment
(N) (n) (n) TRT 1st rebleed TRT TRT ARE bleed re- related to years
bleed (/y) bleed bleed (%) lated mor- treated
(/y) until 2 after 2 bidity (%) CCM (%)
y (/y) y (/y)

Kida & Ha- 152 87 65 N/A 31.8a 8b <5 N/A N/A 2 1991–2001
segawa,
200461†

Liu et al, 125 63 49 N/A 29.2 10.3 3.3 3.2 9.6 0 1993–2002
200562‡

Kida, 84 84 0 N/A N/A 7.1 1.8 N/A N/A 2.4 N/A


200956†

Wang et al, 96 13 83 N/A N/A 4.2 < 2.1 5.2 N/A 0 1995–2005
201064

Lunsford et 103 93 10 N/A 32.5 10.8 1.06 1 N/A 1 1988–2005


al, 201055††

Monaco et 68 68 0 N/A 32.38 8.2 1.37 1.5 N/A 2.9 1988–2005


al, 201070††

Nagy et al, 113/118 118 0 2.2c 30.5 5.1 1.3 7.3 7.3 0 1995–2008
201038 2.9 15 2.4

Lee et al, 49/50 50 0 N/A 31.3 3.3 1.74 4.1 N/A 0 1993–2010
201257‡

Jay et al, 16 16 0 N/A N/A 3.72 3.59 0 0 6.25 1998–2009


201275

Park & 20 20 0 N/A 39.5 8.2 0 5 0 0 2005–2010


Hwang,
201358

Liscák et al, 112 50 62 N/A N/A 3.2 0.5 0.9 3.6 2.7 1992–2000
201367

Frischer et 38 38 0 N/A 7.2 2.63 0.6 7.9 N/A N/A 1987–2011


al, 201359

Fedorcsák et 45/51 14 37 2d 21.7 7.1 0 7 7 0 2008–2012


al, 201441 0.3 0 0 0 0 0 0

Abbreviations: ARE, adverse radiation effect; CCM, cerebral cavernoma; N/A, not applicable.
Source: Data from 38,41,55–59,61,62,64,67,70,75
Note: Latest reports from each group are listed only. (All except one group used Leksell Gamma Knife; the one exception 41 used GammaART-6000).
†, ‡, †† Data from the same groups analyzing either both deep-seated and superficial, or brainstem lesions only.
aDuring 5 years prior to radiosurgery.
bFirst year after treatment.
cFirst line: “low-risk” group, second line: “high-risk” group.
dFirst line: deep-seated, second line: superficial.

87
Stereotactic Radiosurgery for Cavernomas

mainly on the effect of hemorrhage rate of SRS and on its to SRS and found that annual bleed rate from the pretreatment
safety.7 Only studies using a modern SRS technique and provid- 2.2% stabilized at 1.3% after a 2-year period of temporary
ing useful information with at least one of the relevant outcome increase to 5.1% (n = 77).38 As discussed earlier, this result pre-
parameters were considered.38,41,55–59,61,62,64,67,70,75 Of note, stud- sumably reflects the proportion of more aggressive lesions
ies specifically focusing on the effect of SRS on epilepsy are not treated before a second bleed and the delayed response to SRS.
listed. We must also acknowledge that all of these are retrospec-
tive observational studies that lack a control group. Morbidity after Radiosurgery
The rate and severity of morbidity in untreated cases are both
Hemorrhage significantly associated with hemorrhage, exacerbated by
For pragmatic reasons, the minimal definition of clinical hem- repeated hemorrhage, and are seen more readily when CCMs
orrhage is a new clinical event associated with imaging evi- are in deep eloquent locations. As the beneficial effect of SRS is
dence of hemorrhage.38,41,55,57,58,70 However, over half of the expected only after a latency period and the risk of hemorrhage
listed studies fail to define what they consider as hemorrhage. never seems to reach zero, posttreatment hemorrhage may also
Moreover, the lack of distinction between deep-seated and add to persisting morbidity. In those studies where this was
superficial CCMs55,61,62,64,67 and the lack of distinction between mentioned, 0 to 9.6% of patients suffered persisting neurologic
first and repeated bleed67 make it hard to obtain a proper view deficit due to hemorrhage after SRS (n = 442)38,41,58,62,67,75 and
on the real effect of SRS on hemorrhage rate. On this basis of all mortality (1.2%, n = 747) related to treated CCM also came
strict definition, the annual rebleed rate falls from 30.5 to 39.5% from hemorrhage.38,41,58,61,62,64,67,70,75 Once suffering from a
pretreatment to 3.3. to 15% within the first 2 years after SRS, bleed, the likelihood of permanent deficit seems to be the same
and to 0 to 2.4% thereafter in deep-seated lesions with multiple with posttreatment hemorrhages as with pretreatment hemor-
hemorrhages (n = 179).38,57,58,70 Only one study analyzed sepa- rhages, suggesting that the benefit of SRS is not to reduce the
rately those deep-seated lesions after a single or no bleed prior severity but the frequency of bleeds.38

Fig. 10.4 Proposed algorithm for the management of cerebral cavernomas. CCM, cerebral cavernoma; MRI, magnetic resonance imaging. *Surgery is
first option in most cases, but stereotactic radiosurgery is a valid alternative. **Both modalities appear to be effective, but currently there is no
evidence to demonstrate superiority of either. (From Nagy G, Kemeny AA. Stereotactic radiosurgery of intracranial cavernous malformations.
Neurosurg Clin N Am. 2013;24:584. Used with permission.)

88
Stereotactic Radiosurgery for Cavernomas

Adverse radiation effects may be temporary, as perilesional [8] Bertalanffy H, Benes L, Miyazawa T, Alberti O, Siegel AM, Sure U. Cerebral cav-
ernomas in the adult. Review of the literature and analysis of 72 surgically
edema causing either temporary neurologic deficits or remain-
treated patients. Neurosurg Rev 2002; 25: 1–53, discussion 54–55
ing clinically silent is typically seen within 12 months after SRS. [9] Steiner L, Karlsson B, Yen CP, Torner JC, Lindquist C, Schlesinger D. Radiosur-
Persisting adverse radiation effects typically present later, and gery in cavernous malformations: anatomy of a controversy. J Neurosurg
using modern treatment protocols (see above) their rates are 2010; 113: 16–21, discussion 21–22
low (0 to 7.9%, n = 324), resulting in only mild morbidity even in [10] Bertalanffy H, Gerganov VM. Microsurgical or radiosurgical management of
intracranial cavernomas. Acta Neurochir Suppl (Wien) 2013; 116: 103–106
deep-seated lesions.38,41,57,58,59,70,75 Persisting morbidity for
[11] Gross BA, Batjer HH, Awad IA, Bendok BR. Cavernous malformations of the
hemispheric lesions is negligible. basal ganglia and thalamus. Neurosurgery 2009; 65: 7–18, discussion 18–19
[12] Gross BA, Batjer HH, Awad IA, Bendok BR, Du R. Brainstem cavernous malfor-
mations: 1390 surgical cases from the literature. World Neurosurg 2013; 80:

10.4 Summary 89–93


[13] Tomlinson FH, Houser OW, Scheithauer BW, Sundt TM Jr, Okazaki H, Parisi JE.
Angiographically occult vascular malformations: a correlative study of fea-
Stereotactic radiosurgery for CCM is one of the most vibrant
tures on magnetic resonance imaging and histological examination. Neuro-
fields of contemporary SRS, which has gained wide acceptance surgery 1994; 34: 792–799, discussion 799–800
during the last two decades. This may be in large part due to [14] Zabramski JM, Wascher TM, Spetzler RF, et al. The natural history of familial
better imaging, the evolution of radiosurgical techniques, and cavernous malformations: results of an ongoing study. J Neurosurg 1994; 80:
our better understanding of the natural history despite their 422–432
[15] Al-Shahi Salman R, Berg MJ, Morrison L, Awad IA, Angioma Alliance Scientific
heterogenous and unpredictable nature. Stereotactic radiosur-
Advisory Board. Hemorrhage from cavernous malformations of the brain:
gery for this indication evolved from experimental attempts definition and reporting standards. Stroke 2008; 39: 3222–3230
treating aggressive inoperable lesions to becoming an attractive [16] Del Curling O Jr, Kelly DL Jr, Elster AD, Craven TE. An analysis of the natural
treatment modality, especially for deep-seated lesions soon history of cavernous angiomas. J Neurosurg 1991; 75: 702–708
after first presentation to prevent further clinical deteriora- [17] Robinson JR, Awad IA, Little JR. Natural history of the cavernous angioma. J
Neurosurg 1991; 75: 709–714
tion.6,7 Undoubtedly, particularly during the last few years, it
[18] Sage MR, Brophy BP, Sweeney C, et al. Cavernous haemangiomas (angiomas)
has been considered to complement microsurgery and wait- of the brain: clinically significant lesions. Australas Radiol 1993; 37: 147–155
and-watch policy (▶ Fig. 10.4).7 [19] Gross BA, Lin N, Du R, Day AL. The natural history of intracranial cavernous
We acknowledge that SRS in the management of CCM malformations. Neurosurg Focus 2011; 30: E24
[20] Batra S, Lin D, Recinos PF, Zhang J, Rigamonti D. Cavernous malformations:
remains controversial due to the lack of high-level evidence,
natural history, diagnosis and treatment. Nat Rev Neurol 2009; 5: 659–670
although dissenting voices are increasingly in the minority. [21] Al-Shahi R, Bhattacharya JJ, Currie DG, et al. Scottish Intracranial Vascular
There is now sufficient positive experience supporting the use Malformation Study Collaborators. Prospective, population-based detection
of SRS for CCMs located in the brainstem, thalamus, basal gan- of intracranial vascular malformations in adults: the Scottish Intracranial Vas-
glia, or internal capsule, once they become symptomatic. Fur- cular Malformation Study (SIVMS). Stroke 2003; 34: 1163–1169
[22] Al-Shahi Salman R, Hall JM, Horne MA, et al. Scottish Audit of Intracranial
thermore, due to the recognized cumulative morbidity of
Vascular Malformations (SAIVMs) collaborators. Untreated clinical course of
repeated hemorrhages on one hand and the low risk of radia- cerebral cavernous malformations: a prospective, population-based cohort
tion induced adverse effects on the other, we advocate an early study. Lancet Neurol 2012; 11: 217–224
intervention 3 to 6 months after the first bleed (once the pa- [23] Flemming KD, Link MJ, Christianson TJ, Brown RD Jr. Prospective hemorrhage
risk of intracerebral cavernous malformations. Neurology 2012; 78: 632–636
tient has recovered and hematoma resolved). It has been said
[24] Abdulrauf SI, Kaynar MY, Awad IA. A comparison of the clinical profile of cav-
that “controversy is the child of ignorance.” The CCM contro- ernous malformations with and without associated venous malformations.
versy may remain the subject of debate until after further rigor- Neurosurgery 1999; 44: 41–46, discussion 46–47
ous studies are conducted. [25] Lippitz B. Treatment of cavernoma: an evidence-based dilemma? Acta Neuro-
chir Suppl (Wien) 2013; 116: 99–101
[26] Porter PJ, Willinsky RA, Harper W, Wallace MC. Cerebral cavernous malfor-
mations: natural history and prognosis after clinical deterioration with or
References without hemorrhage. J Neurosurg 1997; 87: 190–197
[27] Clatterbuck RE, Eberhart CG, Crain BJ, Rigamonti D. Ultrastructural and im-
[1] Poorthuis M, Samarasekera N, Kontoh K, et al. Comparative studies of the di-
munocytochemical evidence that an incompetent blood-brain barrier is re-
agnosis and treatment of cerebral cavernous malformations in adults: sys-
lated to the pathophysiology of cavernous malformations. J Neurol Neurosurg
tematic review. Acta Neurochir (Wien) 2013; 155: 643–649
Psychiatry 2001; 71: 188–192
[2] Gross BA, Du R. Diagnosis and treatment of vascular malformations of the
[28] Pozzati E, Acciarri N, Tognetti F, Marliani F, Giangaspero F. Growth, subse-
brain. Curr Treat Options Neurol 2014; 16: 279
quent bleeding, and de novo appearance of cerebral cavernous angiomas.
[3] Samarasekera N, Poorthuis M, Kontoh K, et al. Guidelines for the management
Neurosurgery 1996; 38: 662–669, discussion 669–670
of cerebral cavernous malformations in adults. 2012. Available at: http://
[29] Fritschi JA, Reulen HJ, Spetzler RF, Zabramski JM. Cavernous malformations of
www.cavernoma.org.uk/opus473/final_CCM_guidelines_2.pdf. Accessed
the brain stem. A review of 139 cases. Acta Neurochir (Wien) 1994; 130: 35–
March 9, 2014
46
[4] Kondziolka D, Lunsford LD, Flickinger JC, Kestle JR. Reduction of hemorrhage
[30] Kim DS, Park YG, Choi JU, Chung SS, Lee KC. An analysis of the natural history
risk after stereotactic radiosurgery for cavernous malformations. J Neurosurg
of cavernous malformations. Surg Neurol 1997; 48: 9–17, discussion 17–18
1995; 83: 825–831
[31] Moriarity JL, Wetzel M, Clatterbuck RE, et al. The natural history of cavernous
[5] Brown RD Jr, Flemming KD, Meyer FB, Cloft HJ, Pollock BE, Link ML. Natural
malformations: a prospective study of 68 patients. Neurosurgery 1999; 44:
history, evaluation, and management of intracranial vascular malformations.
1166–1171, discussion 1172–1173
Mayo Clin Proc 2005; 80: 269–281
[32] Kondziolka D, Lunsford LD, Kestle JR. The natural history of cerebral caver-
[6] Niranjan A, Lunsford LD. Stereotactic radiosurgery guidelines for the manage-
nous malformations. J Neurosurg 1995; 83: 820–824
ment of patients with intracranial cavernous malformations. Prog Neurol
[33] Kupersmith MJ, Kalish H, Epstein F, et al. Natural history of brainstem caver-
Surg 2013; 27: 166–175
nous malformations. Neurosurgery 2001; 48: 47–53, discussion 53–54
[7] Nagy G, Kemeny AA. Stereotactic radiosurgery of intracranial cavernous mal-
[34] Aiba T, Tanaka R, Koike T, Kameyama S, Takeda N, Komata T. Natural history
formations. Neurosurg Clin N Am 2013; 24: 575–589
of intracranial cavernous malformations. J Neurosurg 1995; 83: 56–59

89
Stereotactic Radiosurgery for Cavernomas

[35] Barker FG II, Amin-Hanjani S, Butler WE, et al. Temporal clustering of hemor- [56] Kida Y. Radiosurgery for cavernous malformations in basal ganglia, thalamus
rhages from untreated cavernous malformations of the central nervous sys- and brainstem. Prog Neurol Surg 2009; 22: 31–37
tem. Neurosurgery 2001; 49: 15–24, discussion 24–25 [57] Lee CC, Pan DH, Chung WY, et al. Brainstem cavernous malformations: the
[36] Wang CC, Liu A, Zhang JT, Sun B, Zhao YL. Surgical management of brain-stem role of Gamma Knife surgery. J Neurosurg 2012; 117 (Suppl): 164–169
cavernous malformations: report of 137 cases. Surg Neurol 2003; 59: 444– [58] Park SH, Hwang SK. Gamma knife radiosurgery for symptomatic brainstem
454, discussion 454 intra-axial cavernous malformations. World Neurosurg 2013; 80: e261–e266
[37] Hasegawa T, McInerney J, Kondziolka D, Lee JY, Flickinger JC, Lunsford LD. [59] Frischer JM, Gatterbauer B, Holzer S, et al. Microsurgery and radiosurgery for
Long-term results after stereotactic radiosurgery for patients with cavernous brainstem cavernomas: effective and complementary treatment options.
malformations. Neurosurgery 2002; 50: 1190–1197, discussion 1197–1198 World Neurosurg 2014; 81:520–528
[38] Nagy G, Razak A, Rowe JG, et al. Stereotactic radiosurgery for deep-seated [60] Pollock BE, Garces YI, Stafford SL, Foote RL, Schomberg PJ, Link MJ. Stereo-
cavernous malformations: a move toward more active, early intervention. tactic radiosurgery for cavernous malformations. J Neurosurg 2000; 93:
Clinical article. J Neurosurg 2010; 113: 691–699 987–991
[39] Gross BA, Batjer HH, Awad IA, Bendok BR. Brainstem cavernous malforma- [61] Kida Y, Hasegawa T. Radiosurgery for cavernous malformations: results of
tions. Neurosurgery 2009; 64: E805–E818, discussion E818 long-term follow-up. In: Kondziolka D, ed. Radiosurgery. Vol 5. Basel, Swit-
[40] Robinson JR Jr, Awad IA, Magdinec M, Paranandi L. Factors predisposing to zerland: Karger; 2004:153–160
clinical disability in patients with cavernous malformations of the brain. Neu- [62] Liu KD, Chung WY, Wu HM, et al. Gamma knife surgery for cavernous heman-
rosurgery 1993; 32: 730–735, discussion 735–736 giomas: an analysis of 125 patients. J Neurosurg 2005; 102 (Suppl): 81–86
[41] Fedorcsák I, Nagy G, Dobai JG, Mezey G, Bognár L. [Radiosurgery of intracere- [63] Régis J, Bartolomei F, Kida Y, et al. Radiosurgery for epilepsy associated with
bral cavernomas—current Hungarian practice]. Ideggyogy Sz 2015; In press cavernous malformation: retrospective study in 49 patients. Neurosurgery
[Hungarian] 2000; 47: 1091–1097
[42] Tung H, Giannotta SL, Chandrasoma PT, Zee CS. Recurrent intraparenchymal [64] Wang P, Zhang F, Zhang H, Zhao H. Gamma knife radiosurgery for intracranial
hemorrhages from angiographically occult vascular malformations. J Neuro- cavernous malformations. Clin Neurol Neurosurg 2010; 112: 474–477
surg 1990; 73: 174–180 [65] Pham M, Gross BA, Bendok BR, Awad IA, Batjer HH. Radiosurgery for angio-
[43] Porter RW, Detwiler PW, Spetzler RF, et al. Cavernous malformations of the graphically occult vascular malformations. Neurosurg Focus 2009; 26: E16
brainstem: experience with 100 patients. J Neurosurg 1999; 90: 50–58 [66] Englot DJ, Han SJ, Lawton MT, Chang EF. Predictors of seizure freedom in the
[44] Lekovic GP, Porter RW, Spetzler RF. Supratentorial and infratentorial caver- surgical treatment of supratentorial cavernous malformations. J Neurosurg
nous malformations. In: Winn HR, ed. Youmans Neurological Surgery. Vol 4. 2011; 115: 1169–1174
6th ed. Philadelphia, PA: Elsevier Saunders; 2011:4134–4145 [67] Liscák R, Urgosik D, Simonova G, Vymazal J, Semnicka J. Gamma knife ra-
[45] Abla AA, Lekovic GP, Turner JD, de Oliveira JG, Porter R, Spetzler RF. Advances diosurgery of brain cavernomas. Acta Neurochir Suppl (Wien) 2013; 116:
in the treatment and outcome of brainstem cavernous malformation surgery: 107–111
a single-center case series of 300 surgically treated patients. Neurosurgery [68] Kim MS, Pyo SY, Jeong YG, Lee SI, Jung YT, Sim JH. Gamma knife surgery for
2011; 68: 403–414, discussion 414–415 intracranial cavernous hemangioma. J Neurosurg 2005; 102 (Suppl): 102–
[46] Dukatz T, Sarnthein J, Sitter H, et al. Quality of life after brainstem cavernoma 106
surgery in 71 patients. Neurosurgery 2011; 69: 689–695 [69] Amin-Hanjani S, Ogilvy CS, Candia GJ, Lyons S, Chapman PH. Stereotactic ra-
[47] Pandey P, Westbroek EM, Gooderham PA, Steinberg GK. Cavernous malforma- diosurgery for cavernous malformations: Kjellberg’s experience with proton
tion of brainstem, thalamus, and basal ganglia: a series of 176 patients. Neu- beam therapy in 98 cases at the Harvard Cyclotron. Neurosurgery 1998; 42:
rosurgery 2013; 72: 573–589, discussion 588–589 1229–1236, discussion 1236–1238
[48] Samii M, Eghbal R, Carvalho GA, Matthies C. Surgical management of brain- [70] Monaco EA, Khan AA, Niranjan A, et al. Stereotactic radiosurgery for the
stem cavernomas. J Neurosurg 2001; 95: 825–832 treatment of symptomatic brainstem cavernous malformations. Neurosurg
[49] Bradac O, Majovsky M, de Lacy P, Benes V. Surgery of brainstem cavernous Focus 2010; 29: E11
malformations. Acta Neurochir (Wien) 2013; 155: 2079–2083 [71] Mitchell P, Hodgson TJ, Seaman S, Kemeny AA, Forster DM. Stereotactic radio-
[50] Szeifert GT, Levivier M, Lorenzoni J, Nyáry I, Major O, Kemeny AA. Morpho- surgery and the risk of haemorrhage from cavernous malformations. Br J
logical observations in brain arteriovenous malformations after gamma knife Neurosurg 2000; 14: 96–100
radiosurgery. Prog Neurol Surg 2013; 27: 119–129 [72] St George EJ, Perks J, Plowman PN. Stereotactic radiosurgery XIV: the role of
[51] Kondziolka D, Lunsford LD, Coffey RJ, Bissonette DJ, Flickinger JC. Stereotactic the haemosiderin ‘ring’ in the development of adverse reactions following ra-
radiosurgery of angiographically occult vascular malformations: indications diosurgery for intracranial cavernous malformations: a sustainable hypothe-
and preliminary experience. Neurosurgery 1990; 27: 892–900 sis. Br J Neurosurg 2002; 16: 385–391
[52] Gewirtz RJ, Steinberg GK, Crowley R, Levy RP. Pathological changes in surgi- [73] Lindquist C, Guo WY, Karlsson B, Steiner L. Radiosurgery for venous angio-
cally resected angiographically occult vascular malformations after radiation. mas. J Neurosurg 1993; 78: 531–536
Neurosurgery 1998; 42: 738–742, discussion 742–743 [74] Lu XY, Sun H, Xu JG, Li QY. Stereotactic radiosurgery of brainstem cavernous
[53] Karlsson B, Kihlström L, Lindquist C, Ericson K, Steiner L. Radiosurgery for malformations: a systematic review and meta-analysis. J Neurosurg 2014;
cavernous malformations. J Neurosurg 1998; 88: 293–297 120: 982–987
[54] Nyáry I, Major O, Hanzély Z, Szeifert GT. Histopathological findings in a surgi- [75] Jay SM, Chandran H, Blackburn TP. Gamma knife stereotactic radiosurgery for
cally resected thalamic cavernous hemangioma 1 year after 40-Gy irradiation. thalamic & brainstem cavernous angiomas. Br J Neurosurg 2012; 26: 367–
J Neurosurg 2005; 102 (Suppl): 56–58 370
[55] Lunsford LD, Khan AA, Niranjan A, Kano H, Flickinger JC, Kondziolka D. Ster-
eotactic radiosurgery for symptomatic solitary cerebral cavernous malforma-
tions considered high risk for resection. J Neurosurg 2010; 113: 23–29

90
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

11 Stereotactic Radiosurgery for Dural Arteriovenous


Fistulas
David Hung-Chi Pan, Wen-Yuh Chung, Huai-Che Yang, and Cheng-Chia Lee

veins may eventually predispose to the risks of cerebral hemor-


Key Points rhage and/or other neurologic deficits.1
Not all DAVFs demonstrate such a progressive clinical course
● A dural arteriovenous fistula is considered an acquired dis- though. Although not frequently seen, some DAVFs can regress
ease. Dural arteriovenous fistulas with anterograde cortical and thrombose gradually resulting in a spontaneous cure.16,17
venous drainage have been clinically regarded as benign, The factors predisposing to DAVF progression or involution
whereas dural arteriovenous fistulas with retrograde cortical have not been clearly clarified.
venous drainage are considered aggressive in behavior.
● For dural arteriovenous fistulas without retrograde cortical
venous drainage or dural arteriovenous fistulas with benign 11.2 Angioarchitecture and
symptoms, radiosurgery may be indicated as an initial treat-
ment. An obliteration rate of more than 70% is expected for
Classifications of Dural
cavernous sinus dural arteriovenous fistulas, and 60% for the Arteriovenous Fistulas
non–cavernous sinus dural arteriovenous fistulas. Few com-
plications are reported. There are many different classifications of DAVFs. The most
● For aggressive dural arteriovenous fistulas with extensive ve- popular classifications are the Borden-Shucart and Cognard
nous reflux, immediate risks of hemorrhage, progressive classifications.12,15 Both systems classify the DAVFs based on
neurologic deficits, or severe venous hypertension, initial their angiographic venous drainage pattern. However, owing to
treatment with endovascular procedure including emboliza- the unique clinical characteristics of DAVFs involving the caver-
tion and angioplasty or with surgery is suggested. nous sinus (CS), Barrow’s classification, which describe the
feeding arteries to the CS DAVFs, is also commonly used to clas-
sify this particular type of DAVFs.18
The Borden-Shucart system distinguishes DAVFs depending
11.1 Introduction on the site of drainage and the presence of cortical venous
drainage.15 Tape I DAVFs drain directly into the sinus or menin-
Intracranial dural arteriovenous fistulas (DAVFs) are abnormal geal veins with antegrade flow, whereas type II DAVFs have ret-
arteriovenous communications within the dura, in which me- rograde flow through the sinus into the subarachnoid veins.
ningeal arteries shunt blood directly into the dural sinus or lep- Type III DAVFs directly drain into the subarachnoid veins in a
tomeningeal veins.1,2 The incidence of DAVFs has been retrograde fashion.
estimated at 5 to 20% of all intracranial vascular malforma- The system of Cognard similarly separates DAVFs depending
tions.3,4,5,6 Dural arteriovenous fistulas constitute only 6% of on the site of drainage and the presence of cortical venous
supratentorial vascular malformations, whereas they are 35% of drainage, but also considers the direction of flow through the
infratentorial malformations.7 The mean age of presentation for draining vein as well as the presence of cortical venous ecta-
a DAVF is 50 to 60 years of age with no sex preference, although sia.12 Cognard type I DAVFs have solely antegrade sinus drain-
there is a wide range seen.8,9 Unlike the more common intra- age, similar to the Borden-Shucart system. Cognard type II
cerebral or parenchymal arteriovenous malformations (AVMs), DAVFs demonstrate retrograde drainage and are subdivided
DAVFs are thought to be acquired due to inflammation, throm- depending on whether drainage is through the sinus (IIa), corti-
bosis, or trauma of the dural sinus. However, the exact etiology cal vein (IIb), or both (IIa + b). Cognard type III DAVFs drain
and underlying disease are difficult to trace in many cases and directly into cortical veins similar to Borden-Shucart, but Cog-
the DAVFs are considered idiopathic.10,11 Dural arteriovenous nard gives lesions with venous ectasia a separate designation of
fistulas most commonly occur in the regions of the cavernous type IV. Dural arteriovenous fistulas that drain into spinal peri-
sinus, transverse/sigmoid sinuses, tentorium/ orcula, or cerebral medullary veins are designated type V by Cognard. Cognard et
convexities with drainage to the superior sagittal sinus. 1,12,13,14 al present their series of 258 patients and demonstrate a strong
A thorough understanding of DVAF morphology requires a correlation between DAVF type and rate of aggressive clinical
detailed cerebral angiographic investigation. Drainage of the symptoms and risk of hemorrhage.
venous flow from a DAVF can be antegrade or retrograde According to Barrow’s classification,18 CS DAVFs are classified
through a dural sinus, through a cortical vein, or both. The pat- into direct (type A) and indirect (types B–D) types. Direct CS
tern of the venous drainage is not necessarily static though. DAVFs are high-flow shunts between the cavernous portion of
Gradual alternation in the venous flow from antegrade to retro- the internal carotid artery and the CS, usually caused by a trau-
grade and delayed recruitment of arterial feeders into the nidus matic laceration of the internal carotid artery or rupture of an
(sump effect) have been observed in some patients. 3,9,13 This is intracavernous carotid aneurysm. Indirect CS DAVFs are dural
hypothesized to occur as a result of progressive sinus hyperten- fistulas between the CS and meningeal branches of the internal
sion with redirection of the blood flow into cortical veins. 2,12,15 carotid artery (type B), the external carotid artery (type C), or
The gradual venous hypertension and reflux of the cortical both (type D).

91
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

Because a DAVF is considered an acquired disease, two poten-


11.3 Clinical Manifestations tial inciting factors for DAVF formation were found: 13.4% of the
The clinical presentation of a DAVF is dependent on its location patients had history of head trauma and 6% had prior head and
and pattern of the venous drainage. The most common loca- neck surgeries.5 Dural arteriovenous fistulas with anterograde
tions are the CS followed by transverse-sigmoid sinus (TSS) that cortical venous drainage have been clinically regarded as
together account for about 80% of the cases.5 Patients with CS benign, whereas DAVFs with retrograde cortical venous drain-
DAVFs often have ocular manifestations (exophthalmos, chemo- age (CVD) are considered aggressive in behavior.9,20,23 In 1990,
sis, visual impairment, and diplopia). The duration of clinical Awad et al reported a meta-analysis of 377 DAVFs and defined
symptoms prior to diagnosis of a CS DAVFs is about 4 months at 100 aggressive cases as those with hemorrhages or progressive
median. In the TSS DAVFs, pulsatile tinnitus and throbbing focal neurologic deficits; the other 277 DAVFs were defined as
headache were the most common symptoms.5 benign cases.1 They concluded that no location of the DAVFs
Similar to other cerebral arteriovenous malformations, was immune from the aggressive neurologic behavior. The fac-
DAVFs can hemorrhage, with an estimated annual risk of tors that predict aggressive neurologic presentation included
approximately 1.8%.19 van Dijk et al in 2002 reported that leptomeningeal venous drainage, variceal or aneurysmal
persistence of the cortical venous reflux in DAVFs yields an venous dilation, and galenic drainage of the DAVFs.
annual hemorrhage rate of 8.1% and a mortality rate of Beyond hemorrhagic episodes, some patients suffered persis-
10.4%.20 Duffau et al reported a high risk of early rebleeding tent or slowly progressive neurologic deficits, including symp-
(35% within 2 weeks) after the first episode of hemorrhage, toms of hemiparesis, hemiparesthesia, cerebellar sign,
with graver consequence from the second bleed. 21 Söderman dementia, and mental confusion. From the authors’ experience
et al in 2008 evaluated hemorrhage rate in their 85 cases of in analysis of 321 DAVF patients, the incidence of nonhemor-
DAVFs with retrograde cortical venous drainage. They found rhagic neurologic deficits is 4.4% (9 of 206 cases) in CS DAVFs
a lower hemorrhage rate compared with those of the other and 37.4% (43 of 115 cases) in NCS DAVFs.
previous reports. In their patients already presenting with an The relationship between duration of clinical symptoms and
intracranial hemorrhage, the annual risk for the recurrent angiographic types of DAVF was also analyzed. In 115 patients
hemorrhage was 7.4%, whereas in those patients not present- with NCS DAVFs, the average duration of symptoms before diag-
ing with a hemorrhage, the bleeding rate was approximately nosis in Borden type I (Cognard type I and type IIa) patients
1.5% per year.22 Pan et al in 20135 reported a Gamma Knife (n = 63) was 19.2 months (range 3–168 months). For Borden
radiosurgery (GKRS) series of 321 DAVF patients, 7 of the 206 type II (Cognard type IIb and type IIa + b) patients (n = 35), the
CS DAVF patients (3.4%) experienced a hemorrhage, and 16 of average duration of symptoms was 39.1 months (range 2–180
the 115 noncavernous sinus (NCS) DAVF patients (13.9%) had months). For Borden type III (Cognard type III, type IV, and type
a hemorrhagic event prior to diagnosis. The report also dem- V) patients (n = 17), the average duration of symptoms was 23.7
onstrated that arteriovenous shunts involving the anterior months (range 1–144 months). This longer duration of symp-
skull base, tentorium, or sphenoparietal sinus harbored a toms seen with higher-grade DAVFs (p < 0.0001 between Bor-
higher risk of hemorrhage (▶ Table 11.1).5 den type I and type II/III, independent t test) suggests that

Table 11.1 Incidence of intracerebral hemorrhage (ICH) and nonhemorrhagic neurologic deficit (NHND) before GKRS in 321 patients with DAVFs
Number ICH % NHND %

Cavernous sinus 206 7 3.4 9 4.4

Transverse-sigmoidal 72 8 11.1 27 37.5


sinus

Petrosal sinus 9 1 11.1 4 44.4

Superior sagittal sinus 8 0 — 3 37.5

Tentorium 9 2 22.2 4 44.4

Frontal base (anterior 6 3 50.0 3 50


fossa)

Sphenoparietal 4 2 50.0 0 —

Vein of Galen 2 0 — 1 50.0

Jugular foramen 2 0 — 0 —

Clivus 2 0 — 0 —

Foramen magnum 1 0 — 1 100.0

Total 321 23 7.2 52 16.2

Abbreviations: DAVFs, dural arteriovenous fistulas; GKRS, Gamma Knife radiosurgery; NHND, nonhemorrhagic neurologic deficits: hemiparesis,
hemiparesthesia, cerebellar sign, dementia, mental confusion.

92
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

DAVFs with retrograde flow may be present for longer time pri- University of Pittsburgh (UPMC). They found an 83% obliteration
or to the diagnosis. rate for patients who had up-front radiosurgery with emboliza-
tion, and a 67% obliteration rate in patients who underwent
radiosurgery alone.43 At the University of Virginia, Cifarelli et al
11.4 Treatment Options in 2010 reported a 65% obliteration rate in a series of 55 DAVF
patients.44 Radiosurgery is often combined with endovascular
The recommended therapeutic intervention for a DAVF is therapy to provide immediate relief of symptoms and possibly
dependent on the anticipated natural history and hemodynam- reduction of the hemorrhage risk.8,14,34,36,37,45,46 In some reports,
ic change of the lesion. For lesions with antegrade sinus drain- DAVF obliteration rates using radiosurgery alone are comparable
age (Borden type I) and benign clinical manifestations, to those using the combined method, as the rates of sympto-
intervention is usually palliative or observational unless the matic improvement.35,37,39 Complications directly related to the
patient’s symptoms are intolerable.6,9 For patients with throb- radiosurgical procedure are notcommonly found.
bing headache, pulsatile tinnitus, ophthalmologic deterioration,
progressive neurologic deficits, increased intracranial pressure,
or elevated risk of hemorrhage, therapeutic intervention is rec- 11.5 Radiosurgical Technique and
ommended.12,15,24,25
Advances in the field of interventional neuroradiology have
Planning
increased treatment options for patients with DAVFs. Oblitera-
tion of the fistula can be attempted through a transarterial or
11.5.1 Principle
transvenous route. Transarterial embolization alone rarely leads Stereotactic radiosurgery is characterized by a steep dose falloff
to a complete obliteration of the DAVF because there are usually of radiation to the target margin; therefore, it relatively spares
numerous arterial feeders to the nidus. The purpose of a trans- the radiation exposure to surrounding normal tissues. Its applica-
arterial approach is mainly for reduction of arterial feeders and tion has been improved by many neurosurgeons, neuroradio-
the palliative symptomatic relief.24 For curative treatment, addi- logists, radiation oncologists, and physicists to advance the
tional treatment through a retrograde transvenous approach treatment of intracranial vascular or neoplastic lesions. Although
may be necessary. In transvenous embolization, superselective stereotactic radiosurgery (SRS) facilities change over the ensuing
disconnection of the refluxing vein is preferred over sacrifice of decades, the basic concepts have not changed: the radiobiological
the dural sinus, although this sometimes becomes necessary to effects of radiosurgery on vascular lesions is due to endothelial
achieve a cure.24 Endovascular therapy can also be combined damage, undulation of internal elastic membrane, proliferative
with surgery or radiosurgery when it is not feasible to com- vasculopathy with narrowing the lumen, subendothelial cell pro-
pletely obliterate a DAVF alone. liferation, and finally, complete lumen obliteration.47
An open surgical approach is indicated for DAVFs with
aggressive features that are not amenable to comprehensive
endovascular treatment. Typically, lesions involving the anteri-
11.5.2 Stereotactic Frame Placement
or cranial fossa or tentorial incisura are often associated with Patients undergo stereotactic frame placement (Leksell frame,
hemorrhage; thus, surgical intervention is indicated. Surgical type G; Elekta Instruments AB, Stockholm, Sweden) supple-
strategies include ligation of the fistula at the junction with the mented by local anesthesia with conscious sedation as needed.
drainage vein, interruption of arterial feeders, coagulation and/ Prior to the frame placement, the scalp is prepared with Beta-
or excision of the fistula in the dura, and resection of the dine and alcohol, and the areas of the pin placements are infil-
involved sinus.17,26,27 Recent studies have suggested that dis- trated with a long-acting local anesthetics. For the CS DAVF, the
connection of the draining vein alone without resection of the target is usually central of the skull, and the axis of frame place-
sinus is equally efficacious as resection of the fistula. The former ment is in the midline without lateralization. On the other
can avoid risks of venous hypertension associated with the hand, the frame is usually lateralized to the side of the lesion
sinus removal, particularly where the sinus is patent. 28,29,30,31 when a DAVF is located in the region of the lateral sinus.
Reported morbidity and mortality of surgical intervention has
ranged from 0 to 13%.32
Stereotactic radiosurgery has long been used for treatment of
11.5.3 Radiographic Images
intraparenchymal AVMs; treatment of DAVFs would be a natu- After the frame fixation, all patients undergo thin-sliced stereo-
ral extension of this. In 1993, Chandler and Friedman first tactic magnetic resonance imaging (MRI) with and without
reported a case in which a DAVF located in the anterior fossa intravenous contrast administration. Precontrast MRI sequen-
was successfully treated with radiosurgery.33 Since then, radio- ces include fast spin-echo (FSE) axial and coronal T2-weighted
surgical treatment has been delivered for DAVFs in various loca- (3-mm sections) images, contrast-enhanced three-dimensional
tions, including the CS, TSS, superior sagittal sinus, tentorium, volumetric MRI, and time-of-flight MR angiography (MRA; 1.5-
and other locations.34–42 Guo et al35 and Pollock et al40 have mm sections). All patients subsequently undergo biplane X-ray
separately reported an approximately 80% obliteration rate for angiography as the next examination. For patients who cannot
CS DAVFs treated by Gamma Knife alone or combined with tolerate an MRI (e.g., those with undetermined implants, pace-
embolization. Söderman et al in 2006 reported on 49 patients makers, or old aneurysm clips), computed tomography (CT)
with 52 DAVFs, with a 68% obliteration rate and another 24% may be an alternative option. However, the image resolution of
with flow regression at 2 years.6 More recently, Yang et al in CT will be less distinctive, especially when the patient had
2010 reported a series of 40 DAVF patients treated at the undergone prior surgery or embolization.

93
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

11.5.4 Treatment Planning and Dose 6-month intervals. Cerebral X-ray angiography is usually per-
formed between 1 to 3 years after GKRS, if complete regres-
Prescription sion of the lesion has been shown on the MRI. For CS DAVFs,
Target localization of the DAVF is performed by integrating a noninvasive color Doppler ultrasonography (CDU) exam-
imaging data from a stereotactic noncontrast MRI, a thin-cut ined through eyeballs is performed every 3 months to evalu-
axial view time-of-flight MRA, and a cerebral X-ray angiogram. ate flow direction and velocity in the superior ophthalmic
Our treatment goal is to occlude the fistulous shunts com- vein (SOV). Frequently, normalization of the CDU is associ-
pletely. Proper delineation of the treatment target to include all ated with concomitant findings of complete obliteration of
abnormal arteriovenous shunts on the dural sinus wall is cru- DAVF on the MRI and cerebral angiography.50
cial for a successful treatment. The target volume is defined Patient outcomes after radiosurgery are grouped into four
along the involved dural sinus wall where the true AVF occurs.2, categories: (1) complete improvement, indicating complete
3,39,47,48,49 The remote arterial feeders and drainage veins distal symptomatic relief with complete obliteration of the DAVF on
to the sinus are excluded from the treatment volume, as they cerebral angiogram and/or MRA; (2) partial improvement, indi-
are not considered a part of the true nidus. cating partial resolution of clinical symptoms with > 50% regres-
Usually, the prescribed margin dose for DAVF radiosurgery is sion of the DAVF nidus on MRA; (3) stationary, indicating no
around 18 to 20 Gy (range 15–25 Gy). The target coverage is change of the DAVF nidus on the follow-up MRA; (4) progres-
within a 50 to 70% isodose line. For the treatment of CS DAVFs, sion, indicating enlargement or aggressive change of the DAVF
we preferred to use a large (14- or 18-mm type C and 16-mm nidus on MRA.
Perfexion Gamma Knife [Elekta Instruments AB]) collimator to
cover the margin of the CS. In our center’s CS DAVF series, the
11.6.1 Treatment Results of DAVF
average number of isocenters was 3 (range 1–14). For the NCS
DAVFs, a greater number of isocenters (several large and many Radiosurgery
8-mm small shots) were used to cover the treatment volume, In our series at the Taipei Veterans General Hospital, post-GKRS
with a mean number of isocenters of 13 (range 1–27). Care is follow-up studies were available in 156 (76%) of 206 patients
taken to protect the adjacent critical structures such as the with CS DAVFs and 108 (94%) of 115 patients with NCS DAVFs.
optic nerve and brainstem to receive radiation doses less than 8 The median follow-up period for the CS group was 20.8 months
to 9 Gy. (range 1–149 months); for the NCS group it was 28 months
(range 2–141 months).
11.6 Follow-up Program ▶ Table 11.2 summarizes the clinical outcomes in our 264
DAVF patients with follow-ups. For the CS DAVFs, 109 of the
After GKRS, both a clinical neurologic examination and radio- 156 patients (70%) showed complete improvement and 47
graphic imaging study (MRI with MRA) are performed at (30%) were partially improved. No lesions were stationary or
progressed after radiosurgery. For the NCS DAVFs, 64 of the 108
(59%) showed complete improvement, 40 (37%) were partially
Table 11.2 Clinical outcome after GKRS in 264 patients with DAVFs
available for neurologic and imaging follow-ups improved, 2 (2%) were stationary, 1 (1%) showed progression,
and 1(1%) patient died.
Outcome CS NCS
To evaluate the response to GKRS in DAVFs with different
Complete improvement 109 (70%) 64 (59%) venous drainage patterns, we further analyzed treatment
results of the 108 patients with NCS DAVFs based on the Borden
Partial improvement 47 (30%) 40 (37%)
classification (▶ Table 11.3). The results show that radiosurgery
Stationary (no change) — 2 (2%) was effective in treating Borden type I lesions, with a 72% com-
plete obliteration rate; another 28% had partial improvement.
Progression — 1 (1%)
However, for Borden type II and III lesions, a lower cure rate
Death — 1 (1%) was observed. Of the 48 Borden type II and III patients, com-
Total 156 108
plete obliteration was observed in 21 (44%) patients, with
another 48% showing partial improvement, 4% stationary, and
Abbreviations: CS, cavernous sinus; DAVFs, dural arteriovenous fistulas; 2% progression. There was one late mortality (2%) in the group
GKRS, Gamma Knife radiosurgery; NCS, noncavernous sinus. of type II–III DAVF patients.

Table 11.3 Clinical outcome after GKRS in 108 patients with NCS DAVFs stratified by Borden classification
Borden type Complete improvement Partial improvement Stationary Progression Death Total

I 43 17 0 0 0 60

II 12 18 1 1 0 32

III 9 5 1 0 1 16

Total 64 40 2 1 1 108

Abbreviations: GKRS, Gamma Knife radiosurgery; NCS DAVFs, noncavernous sinus dural arteriovenous fistulas.

94
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

Table 11.4 Time span between treatment and image-proven oblitera- patients had complete or near-complete resolution of their pre-
tion in dural arteriovenous fistula radiosurgery senting symptoms. Of the eight patients with angiographic fol-
low-up, all demonstrated complete obliteration. 14 In the UPMC
CS NCS
series published in 2010, at a median follow-up of 45 months
n Median time n Median time (range 23–116 months), 28 patients (harboring 32 DAVFs) had
(mo) (mo) obliteration confirmed by imaging. They found a 83% oblitera-
MRI/MRA 100 21.4 (5.1– 70 31.1 (4.7– tion rate in patients who had up-front SRS with embolization
149.5) 140.1) and a 67% obliteration rate in patients who only had SRS. Caver-
nous carotid fistulas were associated with higher rates of occlu-
Angiography 51 24.2 (4.2– 52 32.8 (7.3–
sion (p = 0.012) and symptom improvement (p = 0.010) than
100.7) 143.7)
were TSS-related fistulas.43 In the series of 55 patients treated
Abbreviations: CS, cavernous sinus; MRA, magnetic resonance angiog- at the University of Virginia, the compete obliteration rate of
raphy; MRI, magnetic resonance imaging; NCS, noncavernous sinus. DAVF was 65%.44 Although some of the patients in these pub-
lications had been treated with surgical resection or endovas-
cular embolization prior to radiosurgery, they were referred
The time span between radiosurgery and image-proven obliter- for radiosurgery for further management of the residual
ation is listed in ▶ Table 11.4. For patients with CS DAVFs, the DAVFs. From these studies, we can estimate an overall success
median obliteration time is 21.4 months in MRI/MRA, and 24.2 rate of complete obliteration associated with radiosurgery of
months in cerebral angiography. For patients with NCS DAVFs, the DAVFs at 65 to 77%, with a greater number of patients gain-
median obliteration time is 31.1 months in MRI/MRA, and 32.8 ing symptomatic relief from the radiosurgical treatment.
months in cerebral angiography. Several reports including ours
have noted that the time span between radiosurgery and DAVF
obliteration in some cases can be as short as within 6 months.6,35,
51,52,53 Because the fistulous vessels of DAVFs that lie along the
11.7 Complications
sinus wall are usually small, DAVFs seem to respond more The potential complications after radiosurgery include persis-
promptly to radiosurgery compared with intracerebral AVMs.39 tent venous hypertension, intracranial hemorrhage, cranial
In our patients, one case died due to a new intracerebral nerve dysfunction, sinus stenosis with thrombus formation, late
hemorrhage 59 months after the treatment. This is the only cystic expanding hematoma, and focal radiation-induced brain
mortality in our series. Postradiosurgical hemorrhage due to an edema.
uncontrollable venous hypertension was found in another pa- For a DAVF with retrograde cortical venous drainage, the risk
tient with an extensive, aggressive DAVF (Borden type II) involv- of intracranial hemorrhage after radiosurgery continues until
ing the TSS. This patient recovered from the hemorrhage and such venous reflux has ceased, which is equivalent to the clo-
improved after further combined treatment with endovascular sure of the AVF. Although the hemorrhage risk after radiosur-
embolization and repeated radiosurgery (▶ Fig. 11.1). In this gery in the latency period before DAVF obliteration is low,
series, 98% (260 of 264 patients) had a stable or improved clin- Söderman et al showed a 2.5% annual hemorrhage rate after
ical condition after radiosurgery. For the assessment of adverse GKRS,6 and our data showed a 0.6% hemorrhage rate.5
reactions to radiation on MRI, there was only one patient who In CS DAVFs, the normalization of the reverse pulsatile flow
developed radiation-induced brain edema 6 months after radio- in SOV is usually observed at 3 to 9 months after GKRS,50 indi-
surgery. The edema subsided gradually after steroid treatment. cating a gradual change of the velocity and direction of the
For some DAVFs with extensive involvement of dural sinuses venous flow. After the treatment, thrombosis of the SOV may
and cortical veins, repeated radiosurgery might be necessary be sometimes observed by MRI in patients with temporary
for the complete obliteration of the DAVFs. In our series, 5 CS worsening of symptoms and signs before the onset of clinical
DAVFs and 14 NCS DAVFs had required repeated radiosurgery 1 improvement.54 Lau et al reported such a case of concurrent
to 3 years after the first treatment. The method and dose selec- thrombus formation in the SOV and anterior cavernous sinus 1
tion during second radiosurgery were similar to the first month after radiosurgery. Barcia-Salorio et al also reported that
treatment. 2 of their 25 patients had experienced a temporary worsening
In the literature, cumulative reports have proven the efficacy of symptoms as the shunt occluded.51
of GKRS in treating DAVFs. Shin reported on two patients with In our series, there was one patient complicated with a late
tentorial DAVFs treated with greater than 20 Gy to the fistula, hemorrhagic cyst formation in the region of the DAVF, similar
with complete obliteration obtained in both patients at 27 and to those seen after intracranial AVM radiosurgery.55 The case is
29 months.41 Söderman reported on 49 patients with 52 DAVFs, the first DAVF patient with such a late complication of SRS
showing a 68% obliteration rate and another 24% with flow reported in the literature. The patient underwent craniotomy
regression at 2 years.6 O’Leary achieved a 77% complete obliter- to resect the chronic hemorrhagic cyst 6.5 years after the
ation rate with improvement in another 15% of patients.45 In radiosurgery.
Brown et al’s series, of the 50 patients with angiographic fol- Other radiation-related complications are rare. Until now,
low-up at the Mayo Clinic (Rochester, MN), 68% demonstrated there is no report of temporal lobe radiation necrosis, hypo-
complete obliteration, with another 14% showing near-total thalamic–pituitary axis dysfunction, or radiation-induced sec-
obliteration.8 In Koebbe’s UPMC series published in 2005, all 18 ondary brain tumor for DAVF patients treated with SRS.

95
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

Fig. 11.1 Common carotid angiogram in (a) lateral and (b) anteroposterior views shows a Cognard type IIa + b dural arteriovenous fistula (DAVF)
involving transverse-sigmoid sinus in a 64-year-old man presented with conscious change at admission. The patient underwent Gamma Knife
radiosurgery (GKRS) initially with a peripheral dose of 16 Gy and maximum dose of 29 Gy (mean 21.5 Gy) to the DAVF nidus. Radiation volume was
35.3 mL. (c) One week after GKRS, a right temporal lobe hematoma due to remote venous congestion of the patient was noted in computed
tomography. The patient was further treated with endovascular embolization immediately. Two years after the combined treatment, the residual
DAVF showing in (d) lateral and (e) anteroposterior common carotid angiogram was further treated by the repeated GKRS. Follow-up angiogram (f,g)
several years later showed almost complete obliteration of the DAVF. The patient’s neurologic status significantly improved.

approximately 1.5% annual hemorrhage rate among the


11.8 Special Considerations and patients without previous hemorrhage, and 7.4% among those
Debates with previous hemorrhage.22 However, the selection bias,
including sensitivity of current image modalities, compliance of
11.8.1 Natural History of DAVF patients’ follow-up, and presence of asymptomatic patients, still
existed. We do know the presence of cortical venous reflux
The underlying etiology and natural course of DAVF are not yet
entails a higher risk of intracranial hemorrhage, but it is not
very well understood, and the magnitude of the risk varies con-
clear whether the duration of the disease process has any
siderably between studies.19,20,21,22 Söderman et al reported an
impact on the risk.
85-patient series with 25-year follow-up that demonstrated

96
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

Several studies had noted that the period between radiosur- angiographic complete obliteration rate with preservation of
gery and DAVF obliteration in some cases could be short, as functioning dural sinuses.22,35,42,46
compared with results of AVM radiosurgery.6,35,51,56,57 Different Currently, our strategy of treatment for DAVF patients is as
opinions might exist that to what extent obliteration after follows:
radiosurgery could be attributed to the treatment, or the nature ● For Borden type I (Cognard types I, IIa) DAVFs with persistent

course of the disease, especially when there were several case benign symptoms (headache, pulsatile tinnitus, or eye symp-
reports showing spontaneous DAVF obliteration. 53,58,59,60 Never- toms): Radiosurgery may be indicated as an initial treatment.
theless, a fast closure of the fistulas will shorten the period that ● For Borden type II/III (Cognard types IIa + b, IIb, III) DAVFs

the patient suffers from throbbing headache, tinnitus, or oph- with retrograde CVD:
thalmologic symptoms. In addition, for the cases with cortical a) Symptomatic CVD25 (with hemorrhage, increased intracra-
venous reflux, multimodal treatment including radiosurgery nial pressure, or progressive neurologic deficits): Surgery
and endovascular procedure may reduce the exposed time at or endovascular procedures are indicated for the initial
risk of intracerebral hemorrhage or disease progression. treatment.
b) Asymptomatic CVD25 (with only headache, pulsatile tinni-
tus, or eye symptoms, but without hemorrhage or in-
11.8.2 Selection of Treatment Methods creased intracranial pressure): Endovascular procedure
may be the first- line management. Alternatively, radiosur-
for DAVFs gery can be considered as an initial treatment for patients
The management for a DAVF should be individualized, taking who are elderly, medically frail, or harboring complex
into consideration the clinical presentation of the patient, the DAVFs.
anticipated natural history of the lesion based on location and
angioarchitecture of the DAVFs, and the benefit and inherent In 2010, UPMC published their treatment experience of DAVF
risk of the treatment modality. It is generally agreed that DAVFs and suggested a potential treatment and follow-up algorithm
presenting with hemorrhage, progressive neurologic deficits, or for patients with DAVF, as shown in ▶ Fig. 11.2. For DAVFs with
increased intracranial pressure require prompt treatment good vascular access, SRS followed or preceded by embolization
by endovascular embolization, surgery, or a combination of has the best early and later obliteration rates. For patients with
these procedures, to provide immediate relief of the venous poor vascular access for embolization, SRS alone is usually an
congestion. effective option with low complication rates. They recommend
For Borden type II–III lesions with a single or few CVDs, or that all patients have annual MRI follow-up after SRS. Angiogra-
DAVFs with an isolated dural sinus and CVDs, complete obliter- phy remains the standard to access treatment responses in all
ation of the lesion may be achieved effectively by surgery or patients with aggressive DAVFs. However, for patients with less
endovascular intervention.61,62,63 However, when DAVFs involve aggressive DAVFs and symptomatic improvement, MRI/MRA
dural sinuses with multiple complex feeders and CVDs, surgical may be a sufficient method to access outcomes.43
and endovascular treatment can be technically challenging.
Lucus et al in 1997 reported a meta-analysis and concluded that
even with combined therapy of surgery and embolization, over 11.8.3 The Role of Sinus Recanalization
30% of DAVFs involving TSS will demonstrate residual filling or
persistent symptoms.64 The current application of SRS can pro-
in Treating DAVFs
vide an additional therapeutic method to improve the treat- Two hypotheses have been proposed for the initial pathophysi-
ment result. ology of DAVFs. One is physiological arteriovenous shunts are
When a treatment is indicated for Borden type I DAVFs, the open due to an increment of the sinus and venous pressure.2
therapeutic benefit should outweigh the risks of the treatment. The other one is angiogenesis when venous hypertension
Evidence had shown that injury or increased pressure in the induced by an obstruction of the venous outflow may reduce
dural sinus could trigger the development of DAVFs or cause cerebral perfusion and lead to hypoxia with de novo formation
neurologic deficits secondary to venous hypertension.2 Thus, of DAVFs.67 Based on these theories, a correction of the venous
sacrificing a functioning dural sinus in Borden type I DAVFs by hypertension in the sinus should reduce cerebral venous edema
transvenous intervention or surgery may not be justified. Fur- and reverse vicious cycle of creating DAVFs. Using endovascular
thermore, it is difficult to achieve a complete obliteration of balloon angioplasty or stent implantation inside the sinus may
Borden type I DAVFs by transarterial embolization alone due to correct such venous hypertension.
the frequently complex and torturous course of the arterial sup- This is especially important when facing the patients with
ply.24 Studies had shown that local ischemia caused by incom- complicated DAVFs, such as multiple sinuses stenosis, single
plete closure of the DAVFs after endovascular and/or surgical venous drainage, or DAVFs involving superior sagittal sinus
intervention can increase expression of various vascular growth with venous hypertension. Sinus occlusion by embolization or
factors, which can recruit new collaterals resulting in recanali- resection sometimes carries the risk of further damaging the
zaton of the DAVFs.65,66,67,68 Thus, the use of endovascular inter- venous drainage and cause disastrous results. Any therapeutic
vention or surgery as a first-line treatment for Borden type I intervention of DAVFs in these conditions should be performed
DAVFs with the intention of palliation rather than cure should carefully after recognizing that the fistulous portion of the sinus
carefully balance the risks and benefits of the procedure. Our does not have a drainage function.
study and others have shown that DAVFs with benign venous In these difficult conditions, recanalization of the sinus could
drainage can be safely treated using radiosurgery with a high be the only way to restore venous outflow and correct venous

97
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

Fig. 11.2 Potential treatment algorithm for ma-


nagement of dural arteriovenous fistula. DAVFs,
dural arteriovenous fistulas; MRI, magnetic reso-
nance imaging; SRS, stereotactic radiosurgery.
(Adapted from Yang et al.43)

hypertension. Recently, we began to use balloon inflation or headache, pulsatile tinnitus, or ocular symptoms. For aggressive
stent placement combined with GKRS to treat some DAVFs with DAVFs with extensive CVD, immediate risks of hemorrhage,
partially obstructed sinuses. The idea of angioplasty is also pro- progressive neurologic deficits, or severe venous hypertension,
posed in some reports.69–74 In 2000, Murphy et al reported a initial treatment with endovascular procedure including embo-
case of TSS DAVF treated with transluminal angioplasty and lization and angioplasty or with surgery is suggested. In such
stent placement in a partially thrombosed fistulous sinus. 73 cases, radiosurgery may serve as a secondary treatment for fur-
Other authors subsequently described that stent placement for ther management of the residual fistulas. The latent period for
DAVFs is a promising technique and should be considered a the effects of radiation to occur and the longer time for cure
first-line treatment.70,71 compared with surgery and endovascular therapy remains a
In our experience, the use of balloon dilation or stent im- major drawback for radiosurgery. However, the gradual obliter-
plantation did improve the direction and flow of venous ation of a DAVF after radiosurgery can avoid immediate risk
return, and might facilitate DAVF obliteration if subsequent of aggravated venous hypertension or infarction, which some-
GKRS was performed. However, the study on the efficacy and times complicates endovascular embolization and surgery. It
safety of the angioplasty combined with GKRS is still ongoing is believed that using a multidisciplinary approach to DAVF
and needs more case population for consolidating the clinical management yields better results.
result.

References
11.9 Summary [1] Awad IA, Little JR, Akarawi WP, Ahl J. Intracranial dural arteriovenous malfor-
Stereotactic radiosurgery using the Gamma Knife is a safe and mations: factors predisposing to an aggressive neurological course. J Neuro-
surg 1990; 72: 839–850
effective alternative treatment for DAVFs. This method provides
[2] Hamada Y, Goto K, Inoue T, et al. Histopathological aspects of dural arterio-
a minimally invasive therapeutic modality for patients who venous fistulas in the transverse-sigmoid sinus region in nine patients. Neu-
harbor less aggressive DAVFs, but who suffer from intolerable rosurgery 1997; 40: 452–456, discussion 456–458

98
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

[3] Awad IA. The diagnosis and management of intracranial dural arteriovenous disruption without nidus excision. Neurosurgery 1998; 42: 796–804, discus-
malformations. Contemporary Neurosurgery 1991; 13: 1–5 sion 804–805
[4] Newton TH, Cronqvist S. Involvement of dural arteries in intracranial arterio- [30] Thompson BG, Doppman JL, Oldfield EH. Treatment of cranial dural arteriove-
venous malformations. Radiology 1969; 93: 1071–1078 nous fistulae by interruption of leptomeningeal venous drainage. J Neurosurg
[5] Pan DH, Wu HM, Kuo YH, Chung WY, Lee CC, Guo WY. Intracranial dural arte- 1994; 80: 617–623
riovenous fistulas: natural history and rationale for treatment with stereotac- [31] van Dijk JM, TerBrugge KG, Willinsky RA, Wallace MC. Selective disconnec-
tic radiosurgery. Prog Neurol Surg 2013; 27: 176–194 tion of cortical venous reflux as treatment for cranial dural arteriovenous fis-
[6] Söderman M, Edner G, Ericson K, et al. Gamma knife surgery for dural arterio- tulas. J Neurosurg 2004; 101: 31–35
venous shunts: 25 years of experience. J Neurosurg 2006; 104: 867–875 [32] Kakarla UK, Deshmukh VR, Zabramski JM, Albuquerque FC, McDougall CG,
[7] Aminoff MJ. Vascular anomalies in the intracranial dura mater. Brain 1973; Spetzler RF. Surgical treatment of high-risk intracranial dural arteriovenous
96: 601–612 fistulae: clinical outcomes and avoidance of complications. Neurosurgery
[8] Brown RD Jr, Flemming KD, Meyer FB, Cloft HJ, Pollock BE, Link ML. Natural 2007; 61: 447–457, discussion 457–459
history, evaluation, and management of intracranial vascular malformations. [33] Chandler HC Jr, Friedman WA. Successful radiosurgical treatment of a dural
Mayo Clin Proc 2005; 80: 269–281 arteriovenous malformation: case report. Neurosurgery 1993; 33: 139–141,
[9] Satomi J, van Dijk JM, Terbrugge KG, Willinsky RA, Wallace MC. Benign cranial discussion 141–142
dural arteriovenous fistulas: outcome of conservative management based on [34] Friedman JA, Pollock BE, Nichols DA, Gorman DA, Foote RL, Stafford SL. Re-
the natural history of the lesion. J Neurosurg 2002; 97: 767–770 sults of combined stereotactic radiosurgery and transarterial embolization
[10] Chaudhary MY, Sachdev VP, Cho SH, Weitzner I Jr, Puljic S, Huang YP. Dural for dural arteriovenous fistulas of the transverse and sigmoid sinuses. J Neu-
arteriovenous malformation of the major venous sinuses: an acquired lesion. rosurg 2001; 94: 886–891
AJNR Am J Neuroradiol 1982; 3: 13–19 [35] Guo WY, Pan DH, Wu HM, et al. Radiosurgery as a treatment alternative for
[11] Houser OW, Campbell JK, Campbell RJ, Sundt TM Jr. Arteriovenous malforma- dural arteriovenous fistulas of the cavernous sinus. AJNR Am J Neuroradiol
tion affecting the transverse dural venous sinus—an acquired lesion. Mayo 1998; 19: 1081–1087
Clin Proc 1979; 54: 651–661 [36] Lewis AI, Tomsick TA, Tew JM Jr. Management of tentorial dural arteriovenous
[12] Cognard C, Gobin YP, Pierot L, et al. Cerebral dural arteriovenous fistulas: clin- malformations: transarterial embolization combined with stereotactic radia-
ical and angiographic correlation with a revised classification of venous tion or surgery. J Neurosurg 1994; 81: 851–859
drainage. Radiology 1995; 194: 671–680 [37] Link MJ, Coffey RJ, Nichols DA, Gorman DA. The role of radiosurgery and par-
[13] Cognard C, Houdart E, Casasco A, Gabrillargues J, Chiras J, Merland JJ. Long- ticulate embolization in the treatment of dural arteriovenous fistulas. J Neu-
term changes in intracranial dural arteriovenous fistulae leading to worsen- rosurg 1996; 84: 804–809
ing in the type of venous drainage. Neuroradiology 1997; 39: 59–66 [38] Maruyama K, Shin M, Kurita H, Tago M, Kirino T. Stereotactic radiosurgery for
[14] Koebbe CJ, Singhal D, Sheehan J, et al. Radiosurgery for dural arteriovenous dural arteriovenous fistula involving the superior sagittal sinus. Case report. J
fistulas. Surg Neurol 2005; 64: 392–398, discussion 398–399 Neurosurg 2002; 97 (Suppl): 481–483
[15] Borden JA, Wu JK, Shucart WA. A proposed classification for spinal and cranial [39] Pan DH, Chung WY, Guo WY, et al. Stereotactic radiosurgery for the treat-
dural arteriovenous fistulous malformations and implications for treatment. J ment of dural arteriovenous fistulas involving the transverse-sigmoid sinus. J
Neurosurg 1995; 82: 166–179 Neurosurg 2002; 96: 823–829
[16] Davies MA, Ter Brugge K, Willinsky R, Wallace MC. The natural history and [40] Pollock BE, Nichols DA, Garrity JA, Gorman DA, Stafford SL. Stereotactic radio-
management of intracranial dural arteriovenous fistulae. Part 2: aggressive surgery and particulate embolization for cavernous sinus dural arteriovenous
lesions. Interv Neuroradiol 1997; 3: 303–311 fistulae. Neurosurgery 1999; 45: 459–466, discussion 466–467
[17] Liu JK, Dogan A, Ellegala DB, et al. The role of surgery for high-grade intracra- [41] Shin M, Kurita H, Tago M, Kirino T. Stereotactic radiosurgery for tentorial du-
nial dural arteriovenous fistulas: importance of obliteration of venous out- ral arteriovenous fistulae draining into the vein of Galen: report of two cases.
flow. J Neurosurg 2009; 110: 913–920 Neurosurgery 2000; 46: 730–733, discussion 733–734
[18] Barrow DL, Spector RH, Braun IF, Landman JA, Tindall SC, Tindall GT. Classifi- [42] Wu HM, Pan DH, Chung WY, et al. Gamma Knife surgery for the management
cation and treatment of spontaneous carotid-cavernous sinus fistulas. J Neu- of intracranial dural arteriovenous fistulas. J Neurosurg 2006; 105 (Suppl):
rosurg 1985; 62: 248–256 43–51
[19] Brown RD Jr, Wiebers DO, Nichols DA. Intracranial dural arteriovenous fistu- [43] Yang HC, Kano H, Kondziolka D, et al. Stereotactic radiosurgery with or with-
lae: angiographic predictors of intracranial hemorrhage and clinical outcome out embolization for intracranial dural arteriovenous fistulas. Neurosurgery
in nonsurgical patients. J Neurosurg 1994; 81: 531–538 2010; 67: 1276–1283, discussion 1284–1285
[20] van Dijk JM, terBrugge KG, Willinsky RA, Wallace MC. Clinical course of crani- [44] Cifarelli CP, Kaptain G, Yen CP, Schlesinger D, Sheehan JP. Gamma knife radio-
al dural arteriovenous fistulas with long-term persistent cortical venous re- surgery for dural arteriovenous fistulas. Neurosurgery 2010; 67: 1230–1235,
flux. Stroke 2002; 33: 1233–1236 discussion 1235
[21] Duffau H, Lopes M, Janosevic V, et al. Early rebleeding from intracranial dural [45] O’Leary S, Hodgson TJ, Coley SC, Kemeny AA, Radatz MW. Intracranial dural
arteriovenous fistulas: report of 20 cases and review of the literature. J Neu- arteriovenous malformations: results of stereotactic radiosurgery in 17 pa-
rosurg 1999; 90: 78–84 tients. Clin Oncol (R Coll Radiol) 2002; 14: 97–102
[22] Söderman M, Pavic L, Edner G, Holmin S, Andersson T. Natural history of du- [46] Pan HC, Sun MH, Yang DY, et al. Multidisciplinary treatment of cavernous si-
ral arteriovenous shunts. Stroke 2008; 39: 1735–1739 nus dural arteriovenous fistulae with radiosurgery and embolization. J Clin
[23] Davies MA, TerBrugge K, Willinsky R, Coyne T, Saleh J, Wallace MC. The valid- Neurosci 2005; 12: 744–749
ity of classification for the clinical presentation of intracranial dural arterio- [47] Schneider BF, Eberhard DA, Steiner LE. Histopathology of arteriovenous mal-
venous fistulas. J Neurosurg 1996; 85: 830–837 formations after gamma knife radiosurgery. J Neurosurg 1997; 87: 352–357
[24] Sarma D, ter Brugge K. Management of intracranial dural arteriovenous [48] Graeb DA, Dolman CL. Radiological and pathological aspects of dural arterio-
shunts in adults. Eur J Radiol 2003; 46: 206–220 venous fistulas. Case report. J Neurosurg 1986; 64: 962–967
[25] Zipfel GJ, Shah MN, Refai D, Dacey RG Jr, Derdeyn CP. Cranial dural arteriove- [49] Nishijima M, Takaku A, Endo S, et al. Etiological evaluation of dural arteriove-
nous fistulas: modification of angiographic classification scales based on new nous malformations of the lateral and sigmoid sinuses based on histopatho-
natural history data. Neurosurg Focus 2009; 26: E14 logical examinations. J Neurosurg 1992; 76: 600–606
[26] Kawaguchi T, Hosoda K, Shibata Y, Kidoguchi K, Koyama J, Tamaki N. Direct [50] Chiou HJ, Chou YH, Guo WY, et al. Verifying complete obliteration of carotid
surgical removal of the dural arteriovenous fistulas involving transverse-sig- artery-cavernous sinus fistula: role of color Doppler ultrasonography. J Ultra-
moid sinuses. J Clin Neurosci 2002; 9 (Suppl 1): 16–18 sound Med 1998; 17: 289–295
[27] Sundt TM Jr, Piepgras DG. The surgical approach to arteriovenous malfor- [51] Barcia-Salorio JL, Soler F, Barcia JA, Hernández G. Stereotactic radiosurgery for
mations of the lateral and sigmoid dural sinuses. J Neurosurg 1983; 59: the treatment of low-flow carotid-cavernous fistulae: results in a series of 25
32–39 cases. Stereotact Funct Neurosurg 1994; 63: 266–270
[28] Collice M, D’Aliberti G, Talamonti G, et al. Surgical interruption of leptome- [52] Halbach VV, Higashida RT, Hieshima GB, Reicher M, Norman D, Newton TH.
ningeal drainage as treatment for intracranial dural arteriovenous fistulas Dural fistulas involving the cavernous sinus: results of treatment in 30 pa-
without dural sinus drainage. J Neurosurg 1996; 84: 810–817 tients. Radiology 1987; 163: 437–442
[29] Hoh BL, Choudhri TF, Connolly ES Jr, Solomon RA. Surgical management of [53] Olutola PS, Eliam M, Molot M, Talalla A. Spontaneous regression of a dural ar-
high-grade intracranial dural arteriovenous fistulas: leptomeningeal venous teriovenous malformation. Neurosurgery 1983; 12: 687–690

99
Stereotactic Radiosurgery for Dural Arteriovenous Fistulas

[54] Lau LI, Wu HM, Wang AG, Yen MY, Hsu WM. Paradoxical worsening with [66] Kojima T, Miyachi S, Sahara Y, et al. The relationship between venous hyper-
superior ophthalmic vein thrombosis after gamma knife radiosurgery for du- tension and expression of vascular endothelial growth factor: hemodynamic
ral arteriovenous fistula of cavernous sinus: a case report suggesting the and immunohistochemical examinations in a rat venous hypertension model.
mechanism of the phenomenon. Eye (Lond) 2006; 20: 1426–1428 Surg Neurol 2007; 68: 277–284, discussion 284
[55] Lee CC, Pan DH, Ho DM, et al. Chronic encapsulated expanding hematoma [67] Tirakotai W, Bertalanffy H, Liu-Guan B, Farhoud A, Sure U. Immunohisto-
after gamma knife stereotactic radiosurgery for cerebral arteriovenous mal- chemical study in dural arteriovenous fistulas and possible role of local hypo-
formation. Clin Neurol Neurosurg 2011; 113: 668–671 xia for the de novo formation of dural arteriovenous fistulas. Clin Neurol Neu-
[56] Hasuo K, Mizushima A, Matsumoto S, et al. Type D dural carotid-cavernous rosurg 2005; 107: 455–460
fistula. Results of combined treatment with irradiation and particulate embo- [68] Zhu Y, Lawton MT, Du R, et al. Expression of hypoxia-inducible factor-1 and
lization. Acta Radiol 1996; 37: 294–298 vascular endothelial growth factor in response to venous hypertension. Neu-
[57] Onizuka M, Mori K, Takahashi N, et al. Gamma knife surgery for the treat- rosurgery 2006; 59: 687–696, discussion 687–696
ment of spontaneous dural carotid-cavernous fistulas. Neurol Med Chir (To- [69] Choi BJ, Lee TH, Kim CW, Choi CH. Reconstructive treatment using a stent
kyo) 2003; 43: 477–482, discussion 482–483 graft for a dural arteriovenous fistula of the transverse sinus in the case of
[58] Luciani A, Houdart E, Mounayer C, Saint Maurice JP, Merland JJ. Spontaneous hypoplasia of the contralateral venous sinuses: technical case report. Neuro-
closure of dural arteriovenous fistulas: report of three cases and review of surgery 2009; 65: E994–E996, discussion E996
the literature. AJNR Am J Neuroradiol 2001; 22: 992–996 [70] Levrier O, Métellus P, Fuentes S, et al. Use of a self-expanding stent with bal-
[59] Pritz MB, Pribram HF. Spontaneous closure of a high-risk dural arteriovenous loon angioplasty in the treatment of dural arteriovenous fistulas involving
malformation of the transverse sinus. Surg Neurol 1991; 36: 226–228 the transverse and/or sigmoid sinus: functional and neuroimaging-based
[60] Saito A, Furuno Y, Nishimura S, Kamiyama H, Nishijima M. Spontaneous clo- outcome in 10 patients. J Neurosurg 2006; 104: 254–263
sure of transverse sinus dural arteriovenous fistula: case report. Neurol Med [71] Liebig T, Henkes H, Brew S, Miloslavski E, Kirsch M, Kühne D. Reconstructive
Chir (Tokyo) 2008; 48: 564–568 treatment of dural arteriovenous fistulas of the transverse and sigmoid sinus:
[61] Heros RC. Gamma knife surgery for dural arteriovenous fistulas. J Neurosurg transvenous angioplasty and stent deployment. Neuroradiology 2005; 47:
2006; 104: 861–863, discussion 865–866 543–551
[62] Jiang C, Lv X, Li Y, Zhang J, Wu Z. Endovascular treatment of high-risk tentori- [72] Malek AM, Higashida RT, Balousek PA, et al. Endovascular recanalization with
al dural arteriovenous fistulas: clinical outcomes. Neuroradiology 2009; 51: balloon angioplasty and stenting of an occluded occipital sinus for treatment
103–111 of intracranial venous hypertension: technical case report. Neurosurgery
[63] van Rooij WJ, Sluzewski M, Beute GN. Dural arteriovenous fistulas with corti- 1999; 44: 896–901
cal venous drainage: incidence, clinical presentation, and treatment. AJNR [73] Murphy KJ, Gailloud P, Venbrux A, Deramond H, Hanley D, Rigamonti D. En-
Am J Neuroradiol 2007; 28: 651–655 dovascular treatment of a grade IV transverse sinus dural arteriovenous fistu-
[64] Lucas CdeP, Caldas JG, Prandini MN. Do leptomeningeal venous drainage and la by sinus recanalization, angioplasty, and stent placement: technical case
dysplastic venous dilation predict hemorrhage in dural arteriovenous fistula? report. Neurosurgery 2000; 46: 497–500, discussion 500–501
Surg Neurol 2006; 66 (Suppl 3): S2–S5, discussion S5–S6 [74] Yeh PS, Wu TC, Tzeng WS, Lin HJ. Endovascular angioplasty and stent place-
[65] Klisch J, Kubalek R, Scheufler KM, Zirrgiebel U, Drevs J, Schumacher M. Plas- ment in venous hypertension related to dural arteriovenous fistulas and ve-
ma vascular endothelial growth factor and serum soluble angiopoietin recep- nous sinus thrombosis. Clin Neurol Neurosurg 2010; 112: 167–171
tor sTIE-2 in patients with dural arteriovenous fistulas: a pilot study. Neuro-
radiology 2005; 47: 10–17

100
12 Stereotactic Radiosurgery for
Part IV Meningiomas 102

Benign Tumor Indications 13 Stereotactic Radiosurgery for Pituitary


Adenomas 107

14 Stereotactic Radiosurgery for


Chordomas 120

15 Stereotactic Radiosurgery for


Chondrosarcomas 128

16 Stereotactic Radiosurgery for Glomus


Tumors 132

17 Stereotactic Radiosurgery for


Nonvestibular Schwannomas 137

18 Stereotactic Radiosurgery for


Hemangioblastomas 146

19 Stereotactic Radiosurgery for Vestibular


Schwannomas 150

IV
Stereotactic Radiosurgery for Meningiomas

12 Stereotactic Radiosurgery for Meningiomas


William Friedman and Frank J. Bova

12.2 Radiosurgery Technique


Key Points
Although the details of radiosurgical treatment techniques dif-
● Although resection is the gold standard for meningioma fer somewhat from system to system, the basic paradigm is
treatment, some common meningioma locations (e.g., cav- quite similar everywhere. Below is a brief description of a typi-
ernous sinus or petroclival region) are not readily amenable cal radiosurgical treatment at the University of Florida.
to a complete resection because of location and the proxim- Almost all radiosurgical procedures in adults are performed
ity of vital neural and vascular structures. on an outpatient basis. The patient reports to the neurosurgical
● Local tumor control rates for benign (i.e., World Health clinic the day before treatment for a detailed history and physi-
Organization [WHO] grade I meningiomas) are high, but cal, as well as an in-depth review of the treatment options. If
long-term control for atypical and malignant meningiomas is radiosurgery is deemed appropriate, the patient is sent to the
less favorable. radiology department for a volumetric magnetic resonance
● Stereotactic radiosurgery for meningiomas conveys a long- imaging (MRI) scan. A radiosurgical plan can be generated, in
term morbidity rate of 10% or less. advance, using this MRI study. The next morning, the patient
arrives at 7:00 am. A stereotactic head ring is applied under
local anesthesia. No skin shaving or preparation is required.
Subsequently, stereotactic computed tomography (CT) scanning
12.1 Introduction is performed. One-millimeter slices are obtained throughout
the entire head. The patient is then transported to an outpa-
Stereotactic radiosurgery (SRS) is a minimally invasive tient holding area where he and his family have breakfast and
treatment modality that delivers a large, single dose of radi- relax until the treatment-planning process is complete.
ation to a specific intracranial target while sparing sur- The stereotactic CT scan and the nonstereotactic volumetric
rounding tissue. Unlike conventional fractionated MRI scan are transferred via Ethernet to the treatment-plan-
radiotherapy, SRS does not maximally exploit the higher ning computer. The CT images are quickly processed so that
radiosensitivity of brain lesions relative to normal brain each pixel has an anteroposterior, lateral, and vertical stereotac-
(therapeutic ratio). Its selective destruction is dependent tic coordinate, all related to the head ring previously applied to
mainly on sharply focused high-dose radiation and a steep the patient’s head. Using image fusion software, the nonstereo-
dose gradient away from the defined target. The biological tactic MRI is fused, pixel for pixel, with the stereotactic CT. The
effect is irreparable cellular damage (probably via DNA preplan performed the day before is, likewise, fused to the ster-
strand breaks) and delayed vascular occlusion within the eotactic CT. Final dosimetry then begins and continues until the
high-dose target volume. Because a therapeutic ratio is not neurosurgeon, radiation therapist, and radiation physicist are
required, traditionally radioresistant lesions can be treated. satisfied that an optimal dose plan has been developed. A varie-
However, because destructive doses are used, any normal ty of options are available for optimizing the dosimetry. The
structure included in the target volume is subject to fundamental goal is to deliver a radiation field that is precisely
damage. conformal to the lesion shape, while delivering a minimal dose
The basis for SRS was conceived over 60 years ago by Lars of radiation to all surrounding neural structures.
Leksell.1 He proposed the technique of focusing multiple beams With linac systems, the target dose distribution can be tail-
of external radiation on a stereotactically defined intracranial ored by varying collimator sizes, eliminating undesirable arcs,
target. The averaging of these intersecting beams results in very manipulating arc angles, using multiple isocenters, and differ-
high doses of radiation to the target volume, but innocuously entially weighting the isocenters.9 In our center, multiple iso-
low doses to nontarget tissues along the path of any given centers are used to achieve highly conformal dose distributions,
beam. His team’s implementation of this concept culminated in exactly analogous to the Gamma Knife technique. Some linac
the development of the Gamma Knife. systems use an alternative approach that relies upon a com-
An alternate radiosurgical solution using a linear accelera- puter-driven multileaf collimator to generate nonspherical
tor (linac) was first described in 1984 by Betti and Derechin- beam shapes which are conformal to the beam’s eye view of the
sky.2 Colombo et al described such a system in 1985, 3 and tumor. The multileaf collimator can be adjusted statically or
linacs have subsequently been modified in various ways to dynamically as the linac rotates. Intensity modulation can be
achieve the precision and accuracy required for radiosurgical used to achieve dose distributions that are close to those seen
applications.4–7 In 1986, a team composed of neurosurgeons, with multiple isocenters and treatment time can be reduced.
radiation physicists, and computer programmers began When dose planning is complete, the patient then is attached
development of the University of Florida linac-based radio- to the radiosurgical machine. It takes approximately 10 minutes
surgery system.8 This system has been used to treat over to treat each isocenter. For meningiomas, we almost always use
3,800 patients at the University of Florida since May 1988, a peripheral dose of 12.5 Gy at the 70% isodose line. The head
and is in use at multiple sites worldwide. Many other com- ring is removed, and after a short observation period, the pa-
mercial versions of radiosurgical systems are currently tient is discharged. Patients with benign meningiomas undergo
available. MRI follow-up yearly for 5 years, then every 3 years. Patients

102
Stereotactic Radiosurgery for Meningiomas

with atypical or malignant meningiomas need more frequent 12.3.1 Major Series
follow-up.
The first significant report on radiosurgery for meningiomas
was published by Kondziolka et al in 1991.14 They described 50
patients treated with the new University of Pittsburgh Gamma
12.3 Meningiomas Knife unit. Two-year actuarial tumor control was 96%. Three
Meningiomas are the most common benign primary brain patients had experienced radiation-induced neurologic deficit
tumor, with an incidence of approximately 7/100,000 in the (6%). As of 2008, over 64,000 patients with meningiomas had
general population. Surgery has long been thought to be the been treated with Gamma Knife units and many more with
treatment of choice for symptomatic lesions and is often cura- linac systems. The originally reported numbers on tumor con-
tive. Many meningiomas, however, occur in locations where trol and complications have held up fairly well.15
attempted surgical cure may be associated with morbidity or In 2005, Malik et al16 reported on 309 meningiomas treated
mortality, such as the cavernous sinus or petroclival region.10,11 with the Sheffield Gamma Knife unit. Five-year actuarial control
In addition, many of these tumors occur in the elderly, where was 87% for benign meningiomas, 49% for atypical tumors, and
the risks of general anesthesia and surgery are known to be 0% for malignant tumors. Complications occurred in 3% of the
increased. Hence, there is interest in alternative treatments, patients.
including radiotherapy and radiosurgery, either as a primary or In 2008, Kondziolka et al17 updated the Pittsburgh experi-
adjuvant approach. ence: 972 patients with 1,045 meningiomas had been treated.
Simpson, in a classic paper, described the relationship The overall control rates for benign meningiomas were 93 and
between completeness of surgical resection and tumor recur- 97% for those with previous histological confirmation. Fifty-
rence.12 A grade I resection, which is complete tumor remov- three patients followed for 10 years had a control rate of 91%.
al with excision of the tumor’s dural attachment and Overall morbidity was 8%. Atypical tumor control was 50% and
involved bone, has a 10% recurrence rate. A grade II resection, malignant tumor control was 17%.
complete resection of the tumor and coagulation of its dural In 2012, Pollock et al18,19 provided updates on the Mayo Clinic
attachment, has up to a 20% recurrence rate. Grade III resec- (Rochester, MN) Gamma Knife experience: 416 patients had
tion is complete tumor removal without dural resection or been treated. The 5- and 10-year local controls rates were 96%
coagulation. Grade IV resection is subtotal and grade V resec- and 89%; 11% of patients experienced a radiation-induced com-
tion is simple decompression. Recurrence rates in the grade plication. The median peripheral dose was 16 Gy. Risk factors
IV and V groups basically reflect the natural history of the for complications included higher tumor volume and parasagit-
tumor, with high rates of recurrence over time. Unfortu- tal/falx/convexity location.
nately, some common meningioma locations, such as the cav- In 2012, Santacroce et al20 reported a retrospective observa-
ernous sinus or petroclival region, are not readily amenable tional analysis of 4,565 consecutive patients harboring 5,300
to a complete dural resection or coagulation strategy because benign meningiomas treated at 15 Gamma Knife centers in
of location and the proximity of vital neural and vascular Europe. Median tumor dose was 14 Gy. Only tumors with > 2-
structures. In addition, relatively high complication rates year follow-up were included in the analysis. Five- and 10-year
have been described for meningioma surgery in some loca- actuarial control rates were 95 and 89%, respectively. Perma-
tions and in the elderly. nent morbidity was seen in 7% of patients.
Pollock and colleagues13 analyzed 198 patients with men-
ingiomas < 35 mm in diameter, treated with either surgical 12.3.2 Location-Specific Series
resection or Gamma Knife radiosurgery. Tumor recurrence
was more frequent in the surgical resection group (12% vs. Ding et al21 reported on 65 patients with 90 benign parasagittal
2%). No statistically significant difference was detected in the or falcine meningiomas (▶ Fig. 12.1). The median prescription
3- and 7-year actuarial progression-free survival rate dose was 15 Gy. Actuarial tumor control was 85% at 3 years and
between patients with Simpson grade 1 resections and those 70% at 5 years. Morbidity was 8.2%. The authors felt that these
who underwent radiosurgery. Progression-free survival rates control rates and complications rates compared favorably with
with radiosurgery were superior to Simpson grade 2, 3, and 4 surgery for tumors in this location.
resections. Complications were much lower in the radiosur- Flannery et al22 reported on 168 petroclival meningiomas
gery group. and cerebellopontine angle meningiomas (▶ Fig. 12.2). The
Meningiomas have many ideal characteristics for radio- median dose was 13 Gy. Five- and 10-year tumor control rates
surgery: They are easily visualized on MRI. They are gener- were 91 and 86%, respectively. Morbidity was 8%. Salvage ther-
ally distinct and separate from surrounding brain. It appears apy included surgery4 and repeat radiosurgery.8
that radiation provides tumor control which is at least Zenonos et al23 reported on 24 patients with foramen mag-
equivalent to surgery. And as discussed above, these tumors num meningiomas. The median tumor dose was 13 Gy. At a
are frequently in locations where complete surgical resec- median follow-up of 47 months, all tumors were controlled. No
tion is impossible or risky. Below, we review the major ser- patient suffered a radiation-induced complication. The authors
ies in the literature on radiosurgery for meningiomas. We present a management paradigm calling for radiosurgery of
look at the largest overall series, then at some specific small, minimally symptomatic tumors or for residual tumor
reports categorized by meningioma location. We conclude after far lateral surgical approaches.
with a short review of the relatively small linac radiosur- Kondziolka and colleagues24 reported on 125 patients with
gery experience. convexity meningiomas. The mean marginal dose was 14.2 Gy.

103
Stereotactic Radiosurgery for Meningiomas

Fig. 12.1 (a) Axial enhanced computed tomography radiosurgical image. One isocenter was used, elongated axially by arc elimination, for this 4.1-cc
tumor; 15 Gy was delivered to the 80% isodose line. (b) Axial enhanced magnetic resonance image 12 years later shows tumor shrinkage.

Fig. 12.2 (a) Axial enhanced radiosurgical magnetic resonance image (MRI); 11 isocenters were used to treat this 10.6-cc tumor; 12.5 Gy was
delivered to the 70% isodose line (shown in red). (b) Axial enhanced MRI image 8 years later shows tumor shrinkage.

Actuarial tumor control at 3 and 5 years was 95 and 86%. Over- Lee et al25 reported on 159 such patients. Actuarial tumor
all morbidity was 9.6%. The authors felt that radiosurgery for control was 93% at 5 and 10 years. Morbidity was 9%. Kimball
this location was most valuable for patients with tumors in crit- et al26 reported on 55 patients treated at University of Flori-
ical locations or in poor medical condition. da. Actuarial tumor control was 100% at 5 years and 98% at
A number of authors have reported on radiosurgery for 10 years. Sixty-five percent of patients had improvement in
cavernous sinus meningiomas (▶ Fig. 12.3), a location almost their presenting cranial neuropathies and one patient was
universally recognized as inappropriate for surgery. In 2002, made worse. Spiegelmann et al27 reported on 102 patients

104
Stereotactic Radiosurgery for Meningiomas

Fig. 12.3 (a) Axial enhanced radiosurgical treat-


ment magnetic resonance image (MRI). Five
isocenters were used to treat this 8.8-cc tumor;
12.5 Gy was delivered to the 70% isodose line
(shown in red). (b) Axial MRI 12-years later shows
tumor shrinkage.

treated in Tel Aviv. Gross tumor control was 98%. Complica- As of April 2014, the University of Florida experience with
tions were seen in five patients. Pollock et al 28 reported on meningiomas included 619 patients.
115 patients treated with the Mayo Clinic Gamma Knife.
Tumor control was 99% at 5 years and 93% at 10 years; 12% of
patients had complications. Williams et al 29 reported on 138 12.5 Summary
“parasellar” meningiomas treated in Charlottesville, Virginia.
An extensive literature, involving thousands of patients, consis-
Tumor control was 95% at 5 years and 69% at 10 years; 10% of
tently demonstrates excellent long-term tumor control after
patients experienced complications.
radiosurgery for benign meningiomas. Morbidity is generally in
the range of 10% long term. It is frequently minor (i.e., cranial
12.4 Linear Accelerator neuropathy, seizure, headache). It compares especially well to
surgical morbidity for tumor locations known to be high risk for
Radiosurgery surgery (cavernous sinus, petroclival, parasagittal, foramen
magnum).
Although at least an order of magnitude smaller than the
published Gamma Knife experience, multiple linac radiosur-
gical series have been published. 30–35 Hakim et al were the References
only ones of this early group to report actuarial statistics. 36 A
[1] Leksell L. The stereotaxic method and radiosurgery of the brain. Acta Chir
total of 127 with 155 meningiomas were treated. Actuarial
Scand 1951; 102: 316–319
tumor control for patients with benign tumors was 89.3% at [2] Betti OO, Derechinsky VE. Hyperselective encephalic irradiation with a linear
5 years. Six patients (4.7%) had permanent radiation-induced accelerator. Acta Neurochir Suppl (Wien) 1984; 33: 385–390
complications. [3] Colombo F, Benedetti A, Pozza F, et al. External stereotactic irradiation by lin-
The University of Florida report on linac radiosurgery treat- ear accelerator. Neurosurgery 1985; 16: 154–160
[4] Hartmann GH, Schlegel W, Sturm V, Kober B, Pastyr O, Lorenz WJ. Cerebral
ment of meningiomas is one of the largest published. 37,38 Two
radiation surgery using moving field irradiation at a linear accelerator facility.
hundred ten patients were treated from May 1989 through Int J Radiat Oncol Biol Phys 1985; 11: 1185–1192
December 2001. All patients had follow-up for a minimum of 2 [5] McGinley PH, Butker EK, Crocker IR, Landry JC. A patient rotator for stereotac-
years and no patients were lost to follow-up. Actuarial local tic radiosurgery. Phys Med Biol 1990; 35: 649–657
[6] Podgorsak EB, Olivier A, Pla M, Lefebvre PY, Hazel J. Dynamic stereotactic ra-
control for benign tumors was 100% at 1 and 2 years, and 96%
diosurgery. Int J Radiat Oncol Biol Phys 1988; 14: 115–126
at 5 years. Actuarial local control for atypical tumors was 100% [7] Winston KR, Lutz W. Linear accelerator as a neurosurgical tool for stereotactic
at 1 year, 92% at 2 years, and 77% at 5 years. Actual control for radiosurgery. Neurosurgery 1988; 22: 454–464
malignant tumors was 100% at 1 and 2 years, but only 19% at 5 [8] Friedman WA, Bova FJ. The University of Florida radiosurgery system. Surg
years. Permanent radiation-induced complications occurred in Neurol 1989; 32: 334–342
[9] Friedman WA, Buatti JM, Bova FJ, Mendenhall WM. LINAC Radiosurgery–A
3.8%, all of which involved malignant tumors. These tumor con-
Practical Guide. New York, NY: Springer-Verlag; 1998.
trol and treatment morbidity rates compare well with all other [10] Sekhar LN, Jannetta PJ, Burkhart LE, Janosky JE. Meningiomas involving the
published series. clivus: a six-year experience with 41 patients. Neurosurgery 1990; 27: 764–
Kimball et al26 reported on the University of Florida experi- 781, discussion 781
ence with cavernous sinus meningiomas. Fifty-five patients [11] Sekhar LN, Altschuler EM. Meningiomas of the cavernous sinus. In: Al-Mefty
O, ed. Meningiomas. New York, NY: Raven Press; 1991:445–460
were treated. Actuarial tumor control was 100% at 5 years and
[12] Simpson D. The recurrence of intracranial meningiomas after surgical treat-
98% at 10 years. Sixty-five percent of patients reported ment. J Neurol Neurosurg Psychiatry 1957; 20: 22–39
improvement in their cranial nerve dysfunction, only one pa- [13] Pollock BE, Stafford SL, Utter A, Giannini C, Schreiner SA. Stereotactic radio-
tient was worse. Spiegelmann et al27 reported on their experi- surgery provides equivalent tumor control to Simpson Grade 1 resection for
patients with small- to medium-size meningiomas. Int J Radiat Oncol Biol
ence with 117 cavernous sinus meningiomas. Tumor control
Phys 2003; 55: 1000–1005
was 98%; 37% of patients had improved cranial neuropathies; [14] Kondziolka D, Lunsford LD, Coffey RJ, Flickinger JC. Stereotactic radiosurgery
five patients were worse. of meningiomas. J Neurosurg 1991; 74: 552–559

105
Stereotactic Radiosurgery for Meningiomas

[15] Sheehan JP, Williams BJ, Yen CP. Stereotactic radiosurgery for WHO grade I [27] Spiegelmann R, Cohen ZR, Nissim O, Alezra D, Pfeffer R. Cavernous sinus men-
meningiomas. J Neurooncol 2010; 99: 407–416 ingiomas: a large LINAC radiosurgery series. J Neurooncol 2010; 98: 195–202
[16] Malik I, Rowe JG, Walton L, Radatz MW, Kemeny AA. The use of stereotactic [28] Pollock BE, Stafford SL, Link MJ, Garces YI, Foote RL. Single-fraction radiosur-
radiosurgery in the management of meningiomas. Br J Neurosurg 2005; 19: gery of benign cavernous sinus meningiomas. J Neurosurg 2013; 119: 675–
13–20 682
[17] Kondziolka D, Mathieu D, Lunsford LD, et al. Radiosurgery as definitive ma- [29] Williams BJ, Yen CP, Starke RM, et al. Gamma Knife surgery for parasellar
nagement of intracranial meningiomas. Neurosurgery 2008; 62: 53–58, dis- meningiomas: long-term results including complications, predictive factors,
cussion 58–60 and progression-free survival. J Neurosurg 2011; 114: 1571–1577
[18] Pollock BE, Stafford SL, Link MJ, Brown PD, Garces YI, Foote RL. Single-fraction [30] Valentino V, Schinaia G, Raimondi AJ. The results of radiosurgical manage-
radiosurgery of benign intracranial meningiomas. Neurosurgery 2012; 71: ment of 72 middle fossa meningiomas. Acta Neurochir (Wien) 1993; 122:
604–612, discussion 613 60–70
[19] Pollock BE, Stafford SL, Link MJ, Garces YI, Foote RL. Single-fraction radiosur- [31] Villavicencio AT, Black PM, Shrieve DC, Fallon MP, Alexander E, Loeffler JS. Li-
gery for presumed intracranial meningiomas: efficacy and complications nac radiosurgery for skull base meningiomas. Acta Neurochir (Wien) 2001;
from a 22-year experience. Int J Radiat Oncol Biol Phys 2012; 83: 1414–1418 143: 1141–1152
[20] Santacroce A, Walier M, Régis J, et al. Long-term tumor control of benign in- [32] Engenhart R, Kimmig BN, Höver KH, et al. Stereotactic single high dose radia-
tracranial meningiomas after radiosurgery in a series of 4565 patients. Neu- tion therapy of benign intracranial meningiomas. Int J Radiat Oncol Biol Phys
rosurgery 2012; 70: 32–39, discussion 39 1990; 19: 1021–1026
[21] Ding D, Xu Z, McNeill IT, Yen CP, Sheehan JP. Radiosurgery for parasagittal [33] Spiegelmann R, Nissim O, Menhel J, Alezra D, Pfeffer MR. Linear accelerator
and parafalcine meningiomas. J Neurosurg 2013; 119: 871–877 radiosurgery for meningiomas in and around the cavernous sinus. Neurosur-
[22] Flannery TJ, Kano H, Lunsford LD, et al. Long-term control of petroclival men- gery 2002; 51: 1373–1379, discussion 1379–1380
ingiomas through radiosurgery. J Neurosurg 2010; 112: 957–964 [34] Chuang CC, Chang CN, Tsang NM, et al. Linear accelerator-based radiosurgery
[23] Zenonos G, Kondziolka D, Flickinger JC, Gardner P, Lunsford LD. Gamma Knife in the management of skull base meningiomas. J Neurooncol 2004; 66: 241–
surgery in the treatment paradigm for foramen magnum meningiomas. J 249
Neurosurg 2012; 117: 864–873 [35] Deinsberger R, Tidstrand J, Sabitzer H, Lanner G. LINAC radiosurgery in skull
[24] Kondziolka D, Madhok R, Lunsford LD, et al. Stereotactic radiosurgery for con- base meningiomas. Minim Invasive Neurosurg 2004; 47: 333–338
vexity meningiomas. J Neurosurg 2009; 111: 458–463 [36] Hakim R, Alexander E III, Loeffler JS, et al. Results of linear accelerator-based
[25] Lee JY, Niranjan A, McInerney J, Kondziolka D, Flickinger JC, Lunsford LD. Ster- radiosurgery for intracranial meningiomas. Neurosurgery 1998; 42: 446–
eotactic radiosurgery providing long-term tumor control of cavernous sinus 453, discussion 453–454
meningiomas. J Neurosurg 2002; 97: 65–72 [37] Friedman WA, Murad GJ, Bradshaw P, et al. Linear accelerator surgery for
[26] Kimball MM, Friedman WA, Foote KD, Bova FJ, Chi YY. Linear accelerator ra- meningiomas. J Neurosurg 2005; 103: 206–209
diosurgery for cavernous sinus meningiomas. Stereotact Funct Neurosurg [38] Shafron DH, Friedman WA, Buatti JM, Bova FJ, Mendenhall WM. Linac radio-
2009; 87: 120–127 surgery for benign meningiomas. Int J Radiat Oncol Biol Phys 1999; 43: 321–
327

106
Stereotactic Radiosurgery for Pituitary Adenomas

13 Stereotactic Radiosurgery for Pituitary Adenomas


Cheng-Chia Lee and Jason P. Sheehan

30 to 80%. Because of the rare but well-documented occurrence


Key Points of late recurrence following endocrine remission, long-term
and rigorous clinical and radiographic follow-up is necessary
● Stereotactic radiosurgery offers a high rate of tumor control for all pituitary adenoma patients treated with radiosurgery.
for functioning and nonfunctioning pituitary adenomas.
● A higher margin dose is required to achieve endocrine remis-
sion in functioning adenomas. 13.2 History
● The rate of endocrine remission after radiosurgery appears
Radiotherapy for pituitary disorders had been introduced in
faster than that achieved with fractionated radiotherapy.
1909 for acromegaly.4,5,6 The initial procedure described by
● The most frequent complication following radiosurgery of a
Griming would deliver (from the oral cavity) 200 roentgens to
pituitary adenoma is hypopituitarism. However, radiation-in-
the sella over a period of 1 month with sessions of 1 hour twice
duced hypopituitarism occurs in only a minority of patients.
per week. On the other hand, Beclere had excellent idea to
rotate the patient so that “the hypophysis was treated by cross-
fire through four to five different areas on the frontotemporal
region.”4 However, radiation was elapsed for nearly 30 years
13.1 Introduction until the 1950s, when several authors published their systemic
Pituitary adenomas represent one of the most common types of study on radiotherapy of pituitary adenomas,7,8,9 and concluded
intracranial tumors.1,2 They are quite common among the gen- that radiotherapy to pituitary fossa tumors is most effective at
eral population and constitute 10 to 20% of all intracranial dosage levels of about 4,000 roentgen. With the advancement
tumors.3 They are classified by size, with microadenomas < 1 cm of modern radiological facilities, radiotherapy plays an impor-
in size and macroadenomas at least 1 cm in size, and by hormo- tant role in treating residual or recurrent pituitary adenomas.
nal secretory status, with hormone hypersecretion from func- The Gamma Knife was subsequently employed by Leksell to
tioning lesions and absence of abnormal hormonal production treat the first pituitary adenoma patient in 1968. Initially con-
comprising nonfunctioning lesions. Although their macroscopic ceived by Lars Leksell in 1951, SRS delivers a single high, con-
appearance and anatomical location are relatively homogene- centrated dose of radiation to the target. According to Leksell’s
ous, pituitary tumors have the potential to generate a wide series in 1980, 95 patients with Cushing disease and Nelson
variety of clinical sequelae. Nonfunctioning pituitary adenomas syndrome have been treated by gamma radiation.10,11 Small
enlarge progressively in the pituitary fossa and often extend and spatially well-defined volumes of tissue in the pituitary
beyond the sellar turcica. The enlargement of the tumor may gland can be selectively destroyed. With the smallest collimator,
cause mass effects, including visual field deficits resulting from a kind of intracellular “microradiosurgery” is possible. In a
optic apparatus compression, and hypopituitarism resulting review of 37 patients with an observation time of more than
from compression of the normal pituitary gland. For function- one year, complete remission was obtained in 29 cases. This is a
ing pituitary adenomas, prolactinomas result in amenorrhea– promising result. Since then, radiosurgical devices and techni-
galactorrhea syndrome; growth hormone (GH)-secreting ques have developed significantly with thousands of pituitary
adenomas cause acromegaly in adult and gigantism in child; adenomas treated in the interim.
adrenocorticotropic hormone (ACTH)-secreting adenomas Around the time of Leksell’s development of the Gamma
cause Cushing syndrome. Knife, innovative work in the field of radiosurgery using heavy
Treatment options for pituitary tumors include medical ther- particles from cyclotrons was conducted by Raymond Kjellberg
apy, microscopic or endoscopic surgical resection, radiosurgery, and Jacob Fabrikant. Later, in 1983 at a hospital in Buenos Aires,
radiotherapy, or observation depending on the biochemical Betti and Derechinsky developed the concept of a modified lin-
profile and clinical status of the patient. Stereotactive radiosur- ear accelerator (linac) for SRS.12,13 Their system relied upon a
gery (SRS) is used to treat a number of so-called benign intra- 10-MV linac, and it utilized a chair for the patient which was
cranial tumors, and pituitary adenomas represent one such based upon the Talairach stereotactic frame.14 Other innovative
example. Radiosurgery is most commonly as adjunctive treat- developments in linac-based SRS devices followed shortly
ments following incomplete surgical resection leaving residual thereafter from Winston and Lutz in Boston, Massachusetts,
tumor, tumor recurrence, or failure of medical therapy. In this Hartman and Sturm in Heidelberg, Germany, Barcia-Salorio in
chapter, we present the principal and method of the radiosur- Valencia, Spain, Colombo in Vincenza, Italy, and Podgorsak in
gery for pituitary tumors, including nonfunctioning adenomas, Montreal, Canada.12–18
ACTH-secreting adenomas, GH-secreting adenomas, and prolac-
tinomas. Although postradiosurgery radiographic tumor control
for nonfunctioning adenomas is excellent, typically around 90%, 13.3 Patient Selection and Pre-SRS
the rates of biochemical remission for functioning adenomas
are lower than the tumor control rates. The highest endocrine
Evaluation
remission rates are achieved in patients with Cushing disease Initial diagnosis of pituitary adenomas was based on the combi-
and the lowest in those with prolactinomas; rates range from nation of clinical features of different types of pituitary

107
Stereotactic Radiosurgery for Pituitary Adenomas

adenomas, and biochemical assessment according to recom- supplemented by local anesthesia and conscious sedation as
mended endocrine guidelines. All patients suspected of harbor- needed.22 Prior to frame placement, the scalp is prepared
ing a pituitary tumor should undergo a complete endocrine with alcohol, and the areas of the pin placements are infil-
evaluation, neuroimaging study, and ophthalmic examination trated with a long-acting local anesthetics. The use the angle
before SRS. Endocrine studies should be done through each fac- of the optic apparatus as the axis of frame is usually helpful.
et of the hypothalamic–pituitary–end organ axis, including GH, This angle approximates a line joining the lateral canthus and
insulin-like growth factor-1 (IGF-1), ACTH, serum cortisol, pro- the top of the pinna, and it makes identification of the optic
lactin, T4 or free T4, thyroxin-stimulating hormone (TSH), lutei- nerves, chasm, and tracts easier by having an image that
nizing hormone (LH), follicle-stimulating hormone (FSH), and demonstrates the entire optic apparatus in a single MRI
testosterone (men) levels. Imaging studies consisted of mag- slice.23
netic resonance imaging (MRI) with and without contrast using
thin slices, and volume acquisition through the region of the
sella turcica. The ophthalmologic evaluation included visual
13.5.2 Radiographic Images
acuity and visual field testing. Afterward, all patients underwent thin-sliced stereotactic MRI
If a patient has a neurologic deficit attributable to an adeno- with and without intravenous contrast administration. Imaging
ma, surgery is the initial treatment of choice for all tumors studies were also obtained for all patients using standard pitui-
except a prolactinoma. Transsphenoidal surgery (endoscopic or tary MRI sequences at the University of Virginia (Charlottesville,
microscopic) allows for the most rapid relief of mass effect and VA). Precontrast sequences included coronal and sagittal T1-
reduction in excessive hormone levels in patients with Cushing weighted (1-mm sections), fast spin-echo (FSE) axial. and coro-
disease and acromegaly. Prior surgical resection also facilitates nal T2-weighted (1-mm sections) images. The postcontrast
subsequent radiosurgery; radiosurgeons can always prescribe a sequences included coronal T1-weighted (1-mm sections), sag-
higher dose with safe margin, which are especially important ittal FSE T1-weighted (1-mm sections), and coronal spoiled gra-
for hormone remission of functioning adenomas. dient echo (SPGR) T1-weighted images. Since 2010, dynamic
MRI sequences were utilized as part of radiosurgical planning
sequencing.24,25,26
13.4 Principles of Stereotactic For patients who cannot tolerate an MRI, computed tomogra-
phy (CT) may be an alternative option. However, the accuracy
Radiosurgery would be less and the lesions are usually blurred, especially if
The use of a single high dose of ionizing beams to treat intra- the patient underwent prior surgical resection. Positron emis-
cranial disorders was a creative concept. Radiosurgery is sion tomography (PET) imaging may also be of value for detect-
characterized by a steep dose falloff, thereby relatively spar- ing hypersecretory adenomas; such images can be overlaid on
ing radiation exposure to surrounding normal tissues. Its stereotactic tomographic images (e.g., MRI or CT). 27
application has changed the direction of many fields such as
neurosurgery and radiation oncology. Significant contribu-
tions have been made by numerous neurosurgeons, radiation
13.5.3 Treatment Planning
oncologists, and physicists to advance the field of SRS. How- Regardless of the radiosurgical modality used, SRS requires
ever, despite all the changes in SRS over the ensuing decades, clear and accurate imaging of that target. Advances in radio-
the fundamental concepts have not changed: The radiobio- graphic imaging over the past 20 years have increased the effi-
logical effects of radiosurgery is due to a vascular effect or a cacy and safety of radiosurgical treatment of pituitary lesions.
cytotoxic effect.19 The current treatment planning for a pituitary adenoma is usu-
From its inception, radiosurgery has been a multidiscipli- ally performed using computer-based software. The target vol-
nary discipline. The definition sanctioned by the American ume and the surrounding structures are contoured. A dose plan
Association of Neurological Surgeons, Congress of Neurologi- can be rendered to deliver an ideal dose to the target and a safe
cal Surgeons, and American Society for Radiation Oncology dose to adjacent critical structures. Conformality, dose uniform-
calls for delivery of radiosurgery by a team consisting of a ity, and gradient index should be assessed and adjusted made
neurosurgeon, radiation oncologist, and medical physicist. 20 to optimize the dose plan.
Similarly, the American College of Radiology recommended a When treating a tumor near the optic apparatus, the adjust-
similar multidisciplinary approach to ensure quality of care ment of the frame angle may be appropriate (▶ Fig. 13.1).
and went so far as to specify specific responsibilities during
the SRS process for the individual members of the multidisci-
plinary team.21
13.5.4 Dose Selection
In general, single-session radiosurgical margin doses vary
from 12 to 18 Gy for nonfunctioning adenomas, and from 15
13.5 Radiosurgical Technique and to 30 Gy for functioning adenomas. 28–33 Care should be taken
to avoid high doses or “hot spots” to critical neurovascular
Planning structures such as the cranial nerves (CNs) or carotid artery
within the cavernous sinus region. Radiosurgery can be
13.5.1 Stereotactic Frame Replacement hypofractionated in two to five sessions to deliver a more
Patients underwent stereotactic frame placement (Leksell optimal dose plan tailored to the constraints of a particular
frame; Elekta Instruments AB, Stockholm, Sweden) case.

108
Stereotactic Radiosurgery for Pituitary Adenomas

Fig. 13.1 Case illustrating the dosing plan of a pituitary adenoma. A prescription dose of 20 Gy (inner green isodose line) was delivered to the margin
of the adenoma and lower isodose lines are shown in yellow and the outer green lines. Evaluation of the dose-volume histogram for radiation exposure
to the optic apparatus (in blue) should be performed. In this case, the chiasm received a maximal dose of 5.9 Gy, and right optic nerve a maximum
dose of 6.3 Gy.

13.5.5 Dose Limitations to Critical radiosurgical doses of between 19 and 30 Gy to this region with
a low risk of appreciable side effects.36,39,40,41,42 Injury to the
Structures cavernous segment of the carotid artery is rare after SRS, with
Visual deterioration following SRS is rare and can be avoided if only one isolated case report.43 In cases where the tumor
the dose to the optic apparatus is restricted to ≤ 8 Gy, although appears to extend into the cavernous sinus, shielding can be
reports of 10 to 12 Gy have been described by some groups. 34 used to lower radiation doses to critical structures.
Transitionally, a distance of 3 mm or more between the rostral There does appear to be a direct correlation between the
extent of the adenoma and the optic apparatus is desirable. The effect on adenoma volume and the endocrine remission rate
absolute distance between the optic apparatus is not the limit- following radiosurgery.30,32,36 Fortunately, because most pitui-
ing factor, but rather defines how steeply the radiation gradient tary adenomas are small enough to be well suited for SRS, dose-
must be constructed so that a tolerable dose is delivered to the volume considerations are not usually a limiting factor. Because
optic apparatus while still delivering an effective dose to the the systemic effects of functioning adenomas can be so devas-
adenoma. If an acceptable gradient cannot be constructed, then tating, it seems intuitive to deliver a reasonably high dose
alternative treatment should be considered. Modern radiosur- (≥ 20 Gy to the margin) to allow rapid hormonal normalization
gical devices may allow a distance of as little as 1 to 2 mm. 35,36, and control of tumor growth. For improved rates of hormonal
37 Ultimately, the tolerable absolute dose permitted likely varies normalization in functioning adenomas, margin dose of 25 or
from patient to patient; it is affected by factors such as previous 30 Gy may even be chosen. However, it is not known to what
damage to the optic apparatus by pituitary adenoma compres- degree a higher margin dose (e.g., 20 vs. 30 Gy) will result in
sion, ischemic changes, type and timing of previous interven- delayed hypopituitarism. In cases of functioning adenomas
tions (e.g., fractionated radiotherapy and surgery), the patient’s with radiologically identifiable targets in the cavernous sinus,
age, and the presence or absence of other comorbidities.(e.g., radiosurgical plans can be devised with higher range treatment
diabetes or hypertension).29,38 doses while shielding much of the normal stalk, gland, and
The majority of CNs in the cavernous sinus appear to be more optic apparatus. Nonfunctioning pituitary adenomas appear to
resistant to radiation effects than the optic nerve, but reports of require a lower radiosurgery treatment dose than functioning
cranial neuropathy, particularly after repeat radiosurgery, are adenomas.33,43 The lowest effective dose for a nonfunctioning
well documented.29 Although the tolerable limit to the caver- tumor is not unknown, but many centers deliver 12 to 15 Gy to
nous sinus nerve is unknown, reports have detailed effective the margin.

109
Stereotactic Radiosurgery for Pituitary Adenomas

trial evaluating the role of SRS for 512 patients with nonfunc-
13.6 Outcomes of Radiosurgery tioning pituitary adenomas (median follow-up of 36 months;
for Nonfunctioning Pituitary range 1–223 months), an overall tumor control rate of 93% was
reported.33 Favorable outcomes of tumor control and neuro-
Adenomas logic preservation were more commonly seen in patients older
Because the endocrine presentation is silent, the primary radio- than 50 years, those with a tumor volume < 5 cc, and those
surgical goal for nonfunctioning adenomas is radiological without prior fractionated radiotherapy.33
tumor control. Stereotactic radiosurgery resulted in tumor con-
trol rates of 83 to 100% with a median 46-month follow-up, and 13.7 Outcomes of Radiosurgery
new-onset hypopituitarism following radiosurgery was
observed in 0 to 40% with a median of 9%. ▶ Table 13.1 lists the for Functioning Pituitary
major radiosurgical series for nonfunctioning adenoma patients
since 2002. In our experience at the University of Virginia with
Adenomas
140 nonfunctioning pituitary adenomas patients, we have Unlike nonfunctioning adenomas, the primary radiosurgical
reported an approximately 90% tumor control and delayed goal for functioning adenomas is both endocrine remission and
hypopituitarism in 30% of patients.44 New or worsening CN def- radiological tumor control. Radiological tumor control usually
icits were observed in 14% of patients. In a recent multicenter accompanies endocrine remission, but some adenomas exhibit

Table 13.1 Major radiosurgery series for nonfunctioning pituitary adenomas


Study N Mean or median FU (mo) Mean or median margin Tumor control (%)
dose (Gy)

Feigl et al, 200276 61 55 15 94

Sheehan et al, 200231 42 31 16 98

Wowra et al, 2002 77 30 58 16 93

Petrovich et al, 200378 52 34 15 100

Losa et al, 200479 54 41 17 96

Muacevic et al, 200480 51 22 17 95

Kajiwara et al, 200581 14 32 13 93

Picozzi et al, 200582 51 41 17 97

Iwai et al, 200583 28 36 12 93

Mingione et al, 200684 100 46 19 92

Voges et al, 200685 37 57 13 100

Liscák et al, 200786 140 60 20 100

Pollock et al, 200826 62 64 16 97

Hoybye et al, 200987 23 78 20 100

Kobayashi et al, 200988 71 50 NA 97

Castro et al, 201089 14 42 13 100

Hayashi et al, 201090 43 36 18 100

Gopalan et al, 201191 48 95 18 83

Iwata et al, 201192 100 33 21 Gy/3 fr, 25 Gy/5 fr 98

Park et al, 201124 125 62 13 90

El-Shehaby et al, 201293 21 44 12 85

Runge et al, 201294 65 83 13 98

Starke et al, 201244 140 50 18 90

Wilson et al, 201295 51 50 14 100

Sheehan et al, 201333 512 36 16 93

Abbreviations: fr, fraction; FU, follow-up; Gy: gray; NA, not available.

110
Stereotactic Radiosurgery for Pituitary Adenomas

tumor control yet fail to achieve endocrine remission. Radiosur- remission in Cushing disease compared with those used to con-
gery plays an important role in the treatment of persistent trol the growth of nonfunctioning adenomas. Delayed endo-
Cushing disease, acromegaly, and part of prolactinomas refrac- crine recurrence after radiosurgery-induced remission can
tory to surgical and/or medical management. occur. For example, in a radiosurgical series of 90 patients with
For Cushing disease, endocrine remission is typically defined Cushing disease with a mean follow up of 45 months, recur-
as a normal 24-hour urine free cortisol (UFC) and serum corti- rence of Cushing disease occurred in 10 patients at a mean time
sol. Most radiosurgical series for Cushing disease demonstrate of 27 months after initial remission.29 An illustrative case with
endocrine remission in the majority of patients after radiosur- Cushing disease is shown in ▶ Fig. 13.2.
gery; the mean remission rate across major series is 51%. The For acromegaly, endocrine remission varied widely (range 0–
mean time interval after radiosurgery to endocrine remission 82%) across series, but the mean remission rate for acromegalic
in successfully treated cases is 12 months.29 ▶ Table 13.2 details patients after radiosurgery was 44.7%. Endocrine remission for
recent major radiosurgical series for Cushing disease. Higher acromegaly was typically defined as a normalization of IGF-1 or
radiosurgical doses typically are required to attain endocrine nadir serum GH < 2.5 µg/dL. Some radiosurgical series use the

Table 13.2 Major radiosurgical series for Cushing disease


Study N Mean or median FU (mo) Mean or median margin Endocrine remission (%)
dose (Gy)

Izawa et al, 200096 12 26 24 17

Sheehan et al, 200030 43 39 20 63

Shin et al, 200097 6 88 32 50

Hoybye et al, 200198 18 17 NA 44

Feigl et al, 200276 4 55 15 60

Kobayashi et al, 200299 20 64 29 23

Laws et al, 2002100 40 NA 20 74

Pollock et al, 200263 9 42 20 78

Choi et al, 2003101 7 43 29 56

Petrovich et al, 200378 4 34 15 NA

Witt et al, 200337 8 24 24 0

Wong et al, 2003102 5 38 NA 100

Devin et al, 2004103 35 42 15 49

Kajiwara et al, 200581 2 39 26 50

Voges et al, 200685 17 59 16 53

Castinetti et al, 2007104 40 55 30 43

Jagannathan et al, 200729 90 45 23 54

Petit et al, 2008105 33 62 20 52

Pollock et al, 2008106 8 73 20 87

Tinnel et al, 200862 12 37 25 50

Castinetti et al, 2009107 18 94 28 50

Kobayashi et al, 200988 30 64 29 35

Wan et al, 2009108 68 67 23 28

Hayashi et al, 201090 13 36 25 38

Sheehan et al, 201132 82 31 24 54

Wein et al, 2012109 17 23 18 59

Grant et al, 2013110 15 40 35 73

Sheehan et al, 201370 96 48 22 70

Abbreviations: FU, follow-up; NA, not available.

111
Stereotactic Radiosurgery for Pituitary Adenomas

Fig. 13.2 A 34-year-old woman with Cushing disease underwent two transsphenoidal surgeries before radiosurgery. The sella was empty, and there
was no visible tumor on magnetic resonance imaging. Persistent high serum cortisol (40 µg/dL) and urine free cortisol (UFC; 324 µg/24 h) levels were
noted. The patient underwent whole-sellar stereotactic radiosurgery (SRS) with a margin dose of 25 Gy. Endocrine remission was achieved 22 months
after whole-sellar SRS. The serum cortisol and UFC were gradually decreased, and suppressive medications were halted. However, the patient
eventually developed partial hypopituitarism at 56 months after SRS. TSS, transsphenoidal surgery; GKS, Gamma Knife radiosurgery.

results of an oral glucose tolerance test (OGTT) to define endo- prolactinomas.53 Although some reports demonstrated an
crine remission for acromegaly. ▶ Table 13.3 provides an over- acceptable outcome with a large sample size, SRS used as the
view of recent major radiosurgical series for acromegalic primary treatment is not typical in most medical centers. 53
patients. From prognostic factor analysis, patients with a func- Because prolactinomas usually respond well to medical ther-
tioning adenoma volume of < 3 cc at the time of radiosurgery apy, SRS is reserved for patients who have medicaly refrac-
have been noted to have significantly higher chance of endo- tory prolactinomas. Thus, prolactinoma patients typically
crine remission following radiosurgery.32 Thus, maximum safe undergoing SRS demonstrate a more aggressive adenoma
surgical resection prior to radiosurgery increases the chance of phenotype. At the University of Virginia, only 26% of patients
endocrine remission. At the University of Virginia, the median with medically refractory prolactinomas achieved normaliza-
time to endocrine remission after radiosurgery for acromegaly tion of prolactin at a mean of 24.5 months after SRS; those
was 24 months, and this was longer than for comparable Cush- off antisecretory medication at the time of SRS were more
ing disease patients.32,45 Although drawn from retrospective likely to achieve endocrine remission.38 These results are also
studies, there appears to be compelling evidence to temporarily consistent with the study of Landolt et al. 48
halt pituitary suppressive medications around the time of
radiosurgery for patients with acromegaly; this approach por-
tends a greater rate of endocrine remission after SRS. 46 13.8 Complications
▶ Fig. 13.3 illustrates an acromegaly case who underwent SRS
and achieved successful remission within 27 months. The side effects of ionizing radiation are classified as acute
For prolactinoma treated with a margin dose of 13 to (within days), early delayed (within weeks), and late delayed
30 Gy, the remission rate varied from 0 to 84%. 38,47–52 Patients (within months to years) toxicity.
who were treated with a higher radiation radiosurgical dose
had a significantly higher remission rate. 37,38,49,50 In a cohort
of prolactinoma patients, Witt reported there was no remis-
13.8.1 Acute Radiation Injury
sion with a margin dose under 19 Gy. 37 Pan et al reported a Acute toxicity of SRS is rare compared with conventional
52% remission rate and a higher margin dose of 30 Gy in a radiotherapy. Some acute radiation injuries such as skin
retrospective study of 128 patients in whom Gamma Knife changes and hair loss were rarely present in a current clinical
radiosurgery (GKRS) was used as an up-front treatment for setting.

112
Stereotactic Radiosurgery for Pituitary Adenomas

Table 13.3 Major radiosurgical series for acromegaly


Study N Mean or median FU Mean or median Endocrine remission
(mo) margin dose (Gy) (%)

Izawa et al, 200096 29 26 24 41

Shin et al, 200097 6 43 34 67

Zhang et al, 200052 68 34 31 37

Fukuoka et al, 2001111 9 42 20 50

Ikeda et al, 2001112 17 56 25 82

Feigl et al, 200276 9 55 15 60

Pollock et al, 200263 26 42 20 42

Attanasio et al, 2003113 30 46 20 23

Choi et al, 2003101 9 43 29 50

Muramatsu et al, 2003114 4 30 28 50

Petrovich et al, 200378 5 34 15 NA

Witt et al, 200337 4 24 24 25

Castinetti et al, 2005115 82 50 25 17

Gutt et al, 200554 44 23 18 48

Kajiwara et al, 200581 2 54 14 0

Koybayashi et al, 2005116 67 63 19 5

Jezkova et al, 2006117 96 54 35 50

Voges et al, 200685 64 54 17 38

Pollock et al, 2007118 46 63 20 50

Roberts et al, 2007119 9 25 21 44

Vik-Mo et al, 2007120 61 66 27 17

Jagannathan et al, 200828 95 57 22 53

Losa et al, 2008121 83 69 22 60

Pollock et al, 2008106 27 47 20 67

Tinnel et al, 200862 9 35 25 44

Castinetti et al, 2009107 43 102 24 42

Ronchi et al, 2009122 35 120 20 46

Wan et al, 2009108 103 67 21 37

Hayashi et al, 201090 25 36 25 40

Iwai et al, 2010123 26 84 20 38

Poon et al, 2010124 40 74 20–35 75

Franzin et al, 2012125 103 71 23 61

Liu et al, 201249 40 72 21 48

Grant et al, 2013110 13 40 35 61

Lee et al, 2014126 136 62 25 67

Abbreviations: FU, follow-up; NA, not available.

113
Stereotactic Radiosurgery for Pituitary Adenomas

Fig. 13.3 Stereotactic radiosurgery was performed for a 39-year-old man with residual adenoma after surgical resection. Although the patient failed
to discontinue the long-acting octreotide once, endocrine remission was achieved 23 months after SRS. He developed new hormone deficiencies,
including hypogonadism at 25 months, hypothyroidism at 41 months, and hypoadrenalism at 44 months after SRS. m, months; TSS, transsphenoidal
surgery; GKS, Gamma Knife radiosurgery.

13.8.2 Early Delayed Radiation Injury patient is not afflicted with hypopituitarism does not practi-
cally exist. Furthermore, an optimal radiosurgical dose to
The early delayed radiation injuries include hypopituitarism the target lesion should not be compromised for the sake of
and hypothalamic dysfunction, radiation necrosis, new-onset avoiding hypopituitarism. The clinical consequences of mac-
visual deterioration, or other CN dysfunctions. Complications roscopic tumor progression, recurrence, or persistent hor-
resulting from SRS vary depending on tumor size, the extension mone hypersecretion far outweigh those of radiosurgery-
of tumor, and radiation doses. induced hypopituitarism, which can readily be managed with
medical therapy by neuroendocrinologists.
Hypopituitarism and Hypothalamic On the other hand, very few patients have new-onset diabe-
tes insipidus after SRS, and the incidence of diabetes insipidus
Dysfunction
after radiosurgery is generally related to changes in the tumor
Hypothalamic–pituitary dysfunction is the most common complex that mechanically impact the pituitary. Other hypo-
intermediate-to-late complication of SRS of pituitary adeno- thalamic dysfunctions, including poor control of body tempera-
mas. Approximately 30 to 50% of patients develop a new hor- ture, and changes in sleep and appetite, are exceedingly rare
mone deficiency within 5 to 10 years after radiosurgery. 28,29, event after SRS.
32,33,54 Using SRS for pituitary adenoma at the University of

Virginia, 30% patients developed a delayed onset of hypopi-


tuitarism within 3 years. The thyroid function was affected
Cranial Neuropathy Including Optic Neuritis
most, then gonadotrophic hormone, ACTH, and growth hor- The second most common radiosurgery-related complica-
mone.55 There appeared to be two potent and independent tion following treatment of pituitary adenomas is cranial
variables, margin dose to the tumor and suprasellar exten- neuropathies. Multiple CNs, including II, III, IV, V, and VI, by
sion,55 which were predictive of the risk of developing a hor- virtue of their location in the parasellar and suprasellar
mone deficiency. Although an ideal radiosurgical dose plan regions are at risk of inadvertent injury from radiosurgical
has a steep gradient index that minimizes the dose to normal treatment. Most radiosurgery series report neurologic defi-
pituitary tissue and therefore reduces the risk of treatment- cit rates of < 5%, with optic neuropathy as the most common
induced hypopituitarism, a true safe dose below which the owing to its high sensitivity to radiation-induced damage. 33

114
Stereotactic Radiosurgery for Pituitary Adenomas

In a recent study of 217 pituitary adenoma patients who


underwent radiosurgery, 9 patients (4%) developed new or
13.10 Special Considerations and
worsened CN dysfunction. Of those patients with radiosur- Debates in the Management of
gery-induced cranial neuropathies, six (67%) experienced
complete resolution over a median follow-up of 32 months. Pituitary Adenomas
As such, the radiosurgical maximum dose to the optic appa-
ratus should be kept below the limit threshold of 8 to 12 Gy
13.10.1 Comparison of External Beam
to minimize the risk of optic nerve damage. Careful dose Radiation Therapy versus Radiosurgery
planning with contouring of critical structures and shielding
As a result of the reported higher complications rates as well
of the same can often achieve a solution that delivers an
as the longer and lower success rates particularly for endo-
optimal dose to the target and a typically safe dose to the
crine remission of functioning adenomas after EBRT, the cur-
critical structures.
rent role of adjuvant, postsurgical management of recurrent
or residual pituitary adenomas has largely shifted away from
Radiation Necrosis external beam radiotherapy (EBRT) to radiosurgery. Radiosur-
gery provides certain advantages over EBRT, including
Radiation necrosis rarely occurs in the SRS for pituitary adeno-
increased convenience for the patient due to the relative ease
ma. Neurologic deterioration due to radiation necrosis is often
of single-session radiosurgery treatment compared with
delayed, occurring several months to several years after ioniz-
EBRT and a better ability to spare normal pituitary and neu-
ing radiation to the sellar region. For patients with pituitary
ral structures due to steeper gradient indices. Additionally,
adenoma, radiation necrosis can occur in the tumors that
late-responding tissue, such as pituitary adenoma cells, have
extends to cavernous sinus; temporal lobe necrosis has also
a greater radiobiological response than early-responding tis-
been described in very few cases.28
sue to higher radiation doses in fewer fractions—of which
radiosurgery represents the most extreme end of the spec-
13.8.3 Late Delayed Radiation Effects trum, as it is usually delivered in one session. Furthermore,
the rate of endocrine remission following radiosurgery is
In the era of conventional radiotherapy, late delayed radia- unequivocally more rapid than after EBRT. 64 The faster endo-
tion complications included radiation-induced secondary crine remission achieved with radiosurgery as compared with
brain tumors, and cerebrovascular disturbance in the treat- EBRT can yield substantial benefits to patients with function-
ment of sellar tumors. 56,57,58,59 In the current clinical set- ing adenomas.
ting, the risk of a radiation-induced secondary tumor is Just as with radiosurgery, the most frequently encountered
quite low when radiosurgery is used in the treatment of complication following EBRT is delayed hypopituitarism,
sellar tumors. This may be due to the longer latency period; although the reported rates, ranging from 50 to 100%
however, radiobiologically, it is possible that the sharp dose depending on the duration and quality of endocrine follow-
falloff achieved with SRS results in very few cells at risk for up, are significantly higher than in the radiosurgery litera-
malignant transformation as compared with broader-field ture.65 In most EBRT series, the rates of optic neuropathy are
radiotherapy approaches. 60 This hypothesis is supported by comparable to those found in radiosurgery series. However,
a long-term follow-up study by Rowe and colleagues, 61 in at a total radiation dose of 65 Gy, the 5-year risk of visual def-
which only one new primary intracranial tumor was icits after EBRT is up to 50% with reports of optic neuropathy
reported among 4,877 patients treated by SRS. Fortunately, at doses as low as 46 Gy in 1.8-Gy fractions. 66 More severe
there are currently no reported cases in the literature of complications, which occur at a reportedly higher frequency
radiation-induced neoplasm following radiosurgery for following EBRT than radiosurgery, include a 10-year risk of
pituitary tumors. As for cerebrovascular complications, we radiation-induced neoplasia of 2.7% and a 5-year stroke risk
found only one case report of a patient who had a cerebral of 4%, presumably from radiation-induced carotid stenosis or
infarction with internal carotid artery occlusion after SRS occlusion.57 It is important to note that EBRT, which was
for a pituitary tumor. 43 developed prior to radiosurgery, has been used to treat pitui-
tary adenoma patients for a longer period; this has resulted
in more extended follow-up intervals in the EBRT literature
13.9 Prognosis than in the radiosurgery literature. However, the radiosurgi-
Late biochemical recurrence of secretory pituitary adenomas is cal literature has become quite mature as of late, and the
fortunately rare after successful radiosurgery-induced endo- severe complications associated with ischemic stroke and
crine remission. However, in some radiosurgical series, late radiation-induced neoplasia well documented with EBRT do
recurrence rates of up to 20% have been reported. Several Cush- not seem to be observed with radiosurgery. Nevertheless,
ing disease radiosurgery series have described late biochemical these severe complications are at least theoretically possible
recurrence.29,62 The overall incidence of late biochemical with radiosurgery, too.
recurrence is relatively low.29,63 Ultimately, the existence and Although radiosurgery has displaced EBRT as the preferred
potential for biochemical recurrence despite successful endo- adjuvant treatment modality for pituitary adenoma patients,
crine remission underscores the importance of long-term there remain cases in which EBRT is favored. For large pitui-
endocrine follow-up after radiosurgery for functioning pitui- tary adenomas, typically > 3 cm in diameter, tumors with
tary adenomas. irregular anatomy, including diffuse local infiltration and

115
Stereotactic Radiosurgery for Pituitary Adenomas

suprasellar or brainstem extension, and lesions in very close 13.10.4 The Radioresistant Effects of
proximity to neural structures highly sensitive to radiation
(e.g., the optic apparatus), EBRT may represent a safer treat- Antisecreting Medications
ment option than radiosurgery. External beam radiotherapy The negative effect of somatostatin analogs on the results of
provides reasonable tumor control, with rates exceeding 90% SRS for acromegaly was first reported in 2000. 48,63 Landolt
in most series for nonfunctioning adenomas, but a lower and and colleagues detailed the influence of octreotide on the
later rate of endocrine remission for functioning lesions, with results of SRS in 31 patients. They found that patients
a differential response based on the adenoma subtype. 59,67 treated with octreotide at the time of SRS achieved a nor-
External beam radiotherapy may also be used for patients mal level of growth hormone and IGF-1 after a significantly
with pituitary carcinoma. longer interval than patients who did not receive the
drug.48 Pollock et al also reported similar results in 2002. 63
The similar condition also occurred in the patients with
13.10.2 Role of Up-front Radiosurgery prolactinomas: The remission rate was in patients receiving
As a general principle, the use of radiosurgery in the ma- an antisecretory medication at the time of radiosurgery. 53
nagement of pituitary adenomas should be reserved for As a result of these studies, we discontinued antisecreting
recurrent or residual lesions and for patients with function- medications for 6 weeks before SRS, and resumed them 2 to
ing adenomas who remain symptomatic from persistent 6 weeks after SRS. Similar findings of deleterious effects
hormone hypersecretion despite surgical intervention. The from ketoconazole after SRS for patients with Cushing dis-
literature does not support the routine use of up-front ease have been found. 70 Larger, randomized clinical trials
radiosurgery for pituitary adenomas, especially for func- are still necessary to confirm the negative relationship
tioning adenomas. However, radiosurgery may be used as between hormone-suppressive medications and outcome of
an up-front treatment in rare and unusual circumstances. In radiosurgery.
a multicenter study that enrolled patients from the Univer-
sity of Virginia, the University of Pittsburgh, and Taipei Vet-
eran General Hospital,68 41 patients with nonfunctioning 13.10.5 Whole-Sellar Stereotactic
adenoma underwent GKRS as primary management due to Radiosurgery of MRI-Indeterminate
advanced age, multiple comorbidities, and psychiatric disor-
ders. The overall tumor control rate was 92.7%, and the Functioning Adenomas
actuarial tumor control rate was 94 and 85% at 5 and 10 Although being the best neuroimaging study for defining pitui-
years postradiosurgery, respectively. Radiosurgery could tary adenomas, MRI has been reported to fail to detect between
also be considered as an up-front treatment in a patient 36 and 64% of patients with ACTH-secreting pituitary
with an adenoma that residues largely in the cavernous adenomas.71
sinus and for whom resection is unlikely to produce sub- The patients frequently have residual tumor with micro-
stantial reduction in the overall tumor volume. scopic infiltration of the venous sinuses or adjacent dura and
are neither readily evident on neuroimaging studies nor sur-
gically accessible. As a result, recurrence rates for Cushing
13.10.3 Various Histological Entities of disease have been reported to range from 5 to 27% with a
Nonfunctioning Adenomas versus the mean follow-up period ranging from 6.7 to 9.6 years. 72,73,74,75
Based on the considerations above, total and partial hypo-
Efficacy of SRS physectomies appear to be a reasonable approach for
Silent corticotroph pituitary adenoma represents a rare entity selected patients. At the University of Virginia, we delivered
of nonfunctioning adenoma. Prior studies suggest that it may radiosurgery to the entire sella for symptomatic patients with
appear more aggressive with a high recurrent rate. At the Uni- a medically refractory, invasive, or imaging-negative func-
versity of Virginia, 27 consecutive patients (16% of a whole ser- tioning pituitary adenomas. Adopting a similar rationale of
ies of nonfunctioning adenoma) with silent corticotroph hypophysectomy, whole-sellar SRS attempts to treat recur-
pituitary adenoma were identified based upon a histopatholog- rent or persistent diseases by delivering focused high-dose
ical analysis. The actuarial progression-free survival was 97, 95, radiation to the entire sellar and parasellar contents because
and 89% in the nonfunctioning group and 84, 52, and 52% in the microscopic invasion can neither be localized on imaging
silent corticotroph pituitary adenoma group at 3, 5, and 8 years, studies nor identified by surgeons intraoperatively. Endocrine
respectively.69 Hence, silent corticotroph pituitary adenoma remission of 54, 78, and 87% was achieved within 2, 4, and 6
represents an aggressive entity with a higher recurrence rate years after SRS, respectively. We observed hypopituitarism in
after SRS. In view of the increased risk of recurrence or progres- 42.2% of the patients, with 3.1% eventually developing panhy-
sion, prescribing higher than the dose generally used for non- popituitarism during the follow-up period. Compared with
functioning adenomas (13–18 Gy) may be reasonable. This the morbidities of persistent hypersecretion from functioning
provides a critical implication that an increased margin dose adenomas, patients with postradiosurgery hypopituitarism
may be used to treat patients bearing silent ACTH adenoma in can be readily and fully treated with hormonal replacement.
the future clinical practice. However, a multicenter study or Therefore, whole-sellar SRS could be an effective treatment
clinical trial may be of paramount importance to confirm this for functioning adenoma patients without a discrete adeno-
finding. ma on MRI.

116
Stereotactic Radiosurgery for Pituitary Adenomas

[22] Walton L, Hampshire A, Roper A, et al. Development of a relocatable frame


13.11 Conclusions technique for gamma knife radiosurgery. Technical note. J Neurosurg 2000;
93 (Suppl 3): 198–202
Radiosurgery plays an important role in the contemporary ma- [23] Hayashi M, Taira T, Chernov M, et al. Gamma knife surgery for cancer pain-
nagement of patients with a pituitary adenoma. Stereotactic pituitary gland-stalk ablation: a multicenter prospective protocol since 2002.
radiosurgery is typically used in patients with substantial resid- J Neurosurg 2002; 97 (Suppl): 433–437
ual tumor or recurrence after surgical resection of nonfunction- [24] Park KJ, Kano H, Parry PV, et al. Long-term outcomes after gamma knife ster-
eotactic radiosurgery for nonfunctional pituitary adenomas. Neurosurgery
ing adenomas. Stereotactic radiosurgery is also employed for
2011; 69: 1188–1199
patients with functioning adenomas that fail to achieve endo- [25] Patil CG, Lad SP, Harsh GR, Laws ER Jr, Boakye M. National trends, complica-
crine remission after prior resection. Neurologic function after tions, and outcomes following transsphenoidal surgery for Cushing’s disease
radiosurgery is usually preserved or at times improved, even from 1993 to 2002. Neurosurg Focus 2007; 23: E7
[26] Pollock BE, Cochran J, Natt N, et al. Gamma knife radiosurgery for patients
when the treated adenoma extends into the cavernous sinus.
with nonfunctioning pituitary adenomas: results from a 15-year experience.
Delayed postradiosurgical hypopituitarism is the most common Int J Radiat Oncol Biol Phys 2008; 70: 1325–1329
complication, but is manageable with appropriate hormone [27] Levivier M, Massager N, Wikler D, Devriendt D, Goldman S. Integration of
replacement. Life-long neuroimaging and endocrine follow-up functional imaging in radiosurgery: the example of PET scan. Prog Neurol
is recommended for patients with pituitary adenoma treated Surg 2007; 20: 68–81
[28] Jagannathan J, Sheehan JP, Pouratian N, Laws ER Jr, Steiner L, Vance ML. Gam-
with radiosurgery.
ma knife radiosurgery for acromegaly: outcomes after failed transsphenoidal
surgery. Neurosurgery 2008; 62: 1262–1269, discussion 1269–1270
[29] Jagannathan J, Sheehan JP, Pouratian N, Laws ER, Steiner L, Vance ML. Gamma

References Knife surgery for Cushing’s disease. J Neurosurg 2007; 106: 980–987
[30] Sheehan JM, Vance ML, Sheehan JP, Ellegala DB, Laws ER Jr. Radiosurgery for
[1] Laws ER Jr, Sheehan JP. Pituitary Surgery: A Modern Approach. Basel and New Cushing’s disease after failed transsphenoidal surgery. J Neurosurg 2000; 93:
York: Karger, 2006 738–742
[2] Laws ER Jr, Vance ML. Radiosurgery for pituitary tumors and craniopharyng- [31] Sheehan JP, Kondziolka D, Flickinger J, Lunsford LD. Radiosurgery for residual
iomas. Neurosurg Clin N Am 1999; 10: 327–336 or recurrent nonfunctioning pituitary adenoma. J Neurosurg 2002; 97
[3] Vance ML. Treatment of patients with a pituitary adenoma: one clinician’s ex- (Suppl): 408–414
perience. Neurosurg Focus 2004; 16: E1 [32] Sheehan JP, Pouratian N, Steiner L, Laws ER, Vance ML. Gamma Knife surgery
[4] Béclère A. The radiotherapeutic treatment of tumours of the hypophysis, gi- for pituitary adenomas: factors related to radiological and endocrine out-
gantism, and acromegaly. Arch Roentgen Ray 1910; 14: 142–150 comes. J Neurosurg 2011; 114: 303–309
[5] Gramegna A. Un cas d’acromégalie traité par la radiothérapie Rev Neurol [33] Sheehan JP, Starke RM, Mathieu D, et al. Gamma Knife radiosurgery for the
(Paris) 1909; 17: 15–17 management of nonfunctioning pituitary adenomas: a multicenter study. J
[6] Lindholm J, Nielsen EH. Craniopharyngioma: historical notes. Pituitary 2009; Neurosurg 2013; 119: 446–456
12: 352–359 [34] Leber KA, Berglöff J, Pendl G. Dose-response tolerance of the visual pathways
[7] Bachman AL, Harris W. Roentgen therapy for pituitary adenoma; correlation and cranial nerves of the cavernous sinus to stereotactic radiosurgery. J Neu-
of tumor dose with response in 64 cases. Radiology 1949; 53: 331–341 rosurg 1998; 88: 43–50
[8] Buschke F. Radiotherapy of pituitary adenomas. West J Surg Obstet Gynecol [35] Lim YL, Leem W, Kim TS, Rhee BA, Kim GK. Four years’ experiences in the
1950; 58: 271–278 treatment of pituitary adenomas with gamma knife radiosurgery. Stereotact
[9] Sheline GE, Goldberg MB, Feldman R. Pituitary irradiation for acromegaly. Ra- Funct Neurosurg 1998; 70 (Suppl 1): 95–109
diology 1961; 76: 70–75 [36] Sheehan JP, Jagannathan J, Pouratian N, Steiner L. Stereotactic radiosurgery
[10] Leksell L. Stereotactic radiosurgery. J Neurol Neurosurg Psychiatry 1983; 46: for pituitary adenomas: a review of the literature and our experience. Front
797–803 Horm Res 2006; 34: 185–205
[11] Rähn T, Thorén M, Hall K, Backlund EO. Stereotactic radiosurgery in Cushing’s [37] Witt TC. Stereotactic radiosurgery for pituitary tumors. Neurosurg Focus
syndrome: acute radiation effects. Surg Neurol 1980; 14: 85–92 2003; 14: E10
[12] Betti O, Derechinsky V. [Multiple-beam stereotaxic irradiation]. Neurochirur- [38] Pouratian N, Sheehan J, Jagannathan J, Laws ER Jr, Steiner L, Vance ML. Gam-
gie 1983; 29: 295–298 ma knife radiosurgery for medically and surgically refractory prolactinomas.
[13] Betti OO. [History of radiosurgery]. Cancer Radiother 1998; 2: 101–104 Neurosurgery 2006; 59: 255–266, discussion 255–266
[14] Mehta MP. The physical, biologic, and clinical basis of radiosurgery. Curr Probl [39] Kuo JS, Chen JC, Yu C, et al. Gamma knife radiosurgery for benign cavernous
Cancer 1995; 19: 265–329 sinus tumors: quantitative analysis of treatment outcomes. Neurosurgery
[15] Colombo F, Benedetti A, Pozza F, et al. External stereotactic irradiation by lin- 2004; 54: 1385–1393, discussion 1393–1394
ear accelerator. Neurosurgery 1985; 16: 154–160 [40] Liu AL, Wang C, Sun S, Wang M, Liu P. Gamma knife radiosurgery for tumors
[16] Hartmann GH, Schlegel W, Sturm V, Kober B, Pastyr O, Lorenz WJ. Cerebral involving the cavernous sinus. Stereotact Funct Neurosurg 2005; 83: 45–51
radiation surgery using moving field irradiation at a linear accelerator facility. [41] Nakamura N, Shin M, Tago M, et al. Gamma knife radiosurgery for cavernous
Int J Radiat Oncol Biol Phys 1985; 11: 1185–1192 hemangiomas in the cavernous sinus. Report of three cases. J Neurosurg
[17] Lutz W, Winston KR, Maleki N. A system for stereotactic radiosurgery with a 2002; 97 (Suppl): 477–480
linear accelerator. Int J Radiat Oncol Biol Phys 1988; 14: 373–381 [42] Peker S, Kiliç T, Sengöz M, Pamir MN. Radiosurgical treatment of cavernous
[18] Podgorsak EB, Olivier A, Pla M, Hazel J, de Lotbinière A, Pike B. Physical as- sinus cavernous haemangiomas. Acta Neurochir (Wien) 2004; 146: 337–341,
pects of dynamic stereotactic radiosurgery. Appl Neurophysiol 1987; 50: discussion 340
263–268 [43] Lim YJ, Leem W, Park JT, Kim TS, Rhee BA, Kim GK. Cerebral infarction with
[19] Garcia-Barros M, Paris F, Cordon-Cardo C, et al. Tumor response to radio- ICA occlusion after Gamma Knife radiosurgery for pituitary adenoma: a case
therapy regulated by endothelial cell apoptosis. Science 2003; 300: 1155– report. Stereotact Funct Neurosurg 1999; 72 (Suppl 1): 132–139
1159 [44] Starke RM, Williams BJ, Jane JA Jr, Sheehan JP. Gamma Knife surgery for pa-
[20] Barnett GH, Linskey ME, Adler JR, et al. American Association of Neuro- tients with nonfunctioning pituitary macroadenomas: predictors of tumor
logical Surgeons. Congress of Neurological Surgeons Washington Com- control, neurological deficits, and hypopituitarism. J Neurosurg 2012; 117:
mittee Stereotactic Radiosurgery Task Force. Stereotactic radiosurgery— 129–135
an organized neurosurgery-sanctioned definition. J Neurosurg 2007; [45] Lee CC, Vance ML, Lopes MB, Xu Z, Chen CJ, Sheehan J. Stereotactic radiosur-
106: 1–5 gery for acromegaly: outcomes by adenoma subtype. Pituitary 2014
[21] Seung SK, Larson DA, Galvin JM, et al. American College of Radiology (ACR) [46] Loeffler JS, Shih HA. Radiation therapy in the management of pituitary adeno-
and American Society for Radiation Oncology (ASTRO) Practice Guideline for mas. J Clin Endocrinol Metab 2011; 96: 1992–2003
the Performance of Stereotactic Radiosurgery (SRS). Am J Clin Oncol 2013; [47] Jezková J, Hána V, Krsek M, et al. Use of the Leksell gamma knife in the treat-
36: 310–315 ment of prolactinoma patients. Clin Endocrinol (Oxf) 2009; 70: 732–741

117
Stereotactic Radiosurgery for Pituitary Adenomas

[48] Landolt AM, Haller D, Lomax N, et al. Octreotide may act as a radioprotective [74] Sonino N, Zielezny M, Fava GA, Fallo F, Boscaro M. Risk factors and long-term
agent in acromegaly. J Clin Endocrinol Metab 2000; 85: 1287–1289 outcome in pituitary-dependent Cushing’s disease. J Clin Endocrinol Metab
[49] Liu X, Kano H, Kondziolka D, et al. Gamma knife stereotactic radiosurgery for 1996; 81: 2647–2652
drug resistant or intolerant invasive prolactinomas. Pituitary 2013; 16: 68– [75] Yap LB, Turner HE, Adams CB, Wass JA. Undetectable postoperative cortisol
75 does not always predict long-term remission in Cushing’s disease: a single
[50] Pan L, Zhang N, Wang EM, Wang BJ, Dai JZ, Cai PW. Gamma knife radiosur- centre audit. Clin Endocrinol (Oxf) 2002; 56: 25–31
gery as a primary treatment for prolactinomas. J Neurosurg 2000; 93 (Suppl [76] Feigl GC, Bonelli CM, Berghold A, Mokry M. Effects of gamma knife radiosur-
3): 10–13 gery of pituitary adenomas on pituitary function. J Neurosurg 2002; 97
[51] Tanaka S, Link MJ, Brown PD, Stafford SL, Young WF Jr, Pollock BE. Gamma (Suppl): 415–421
knife radiosurgery for patients with prolactin-secreting pituitary adenomas. [77] Wowra B, Stummer W. Efficacy of gamma knife radiosurgery for nonfunc-
World Neurosurg 2010; 74: 147–152 tioning pituitary adenomas: a quantitative follow up with magnetic reso-
[52] Zhang N, Pan L, Wang EM, Dai JZ, Wang BJ, Cai PW. Radiosurgery for growth nance imaging-based volumetric analysis. J Neurosurg 2002; 97 (Suppl):
hormone-producing pituitary adenomas. J Neurosurg 2000; 93 (Suppl 3): 6– 429–432
9 [78] Petrovich Z, Yu C, Giannotta SL, Zee CS, Apuzzo ML. Gamma knife radiosur-
[53] Landolt AM, Lomax N. Gamma knife radiosurgery for prolactinomas. J Neuro- gery for pituitary adenoma: early results. Neurosurgery 2003; 53: 51–59, dis-
surg 2000; 93 (Suppl 3): 14–18 cussion 59–61
[54] Gutt B, Wowra B, Alexandrov R, et al. Gamma-knife surgery is effective in [79] Losa M, Valle M, Mortini P, et al. Gamma knife surgery for treatment of resid-
normalising plasma insulin-like growth factor I in patients with acromegaly. ual nonfunctioning pituitary adenomas after surgical debulking. J Neurosurg
Exp Clin Endocrinol Diabetes 2005; 113: 219–224 2004; 100: 438–444
[55] Xu Z, Lee Vance M, Schlesinger D, Sheehan JP. Hypopituitarism after stereo- [80] Muacevic A, Wowra B, Siefert A, Tonn JC, Steiger HJ, Kreth FW. Microsurgery
tactic radiosurgery for pituitary adenomas. Neurosurgery 2013; 72: 630–637, plus whole brain irradiation versus Gamma Knife surgery alone for treatment
636–637 of single metastases to the brain: a randomized controlled multicentre phase
[56] Brada M, Ashley S, Ford D, Traish D, Burchell L, Rajan B. Cerebrovascular mor- III trial. J Neurooncol 2008; 87: 299–307
tality in patients with pituitary adenoma. Clin Endocrinol (Oxf) 2002; 57: [81] Kajiwara K, Saito K, Yoshikawa K, et al. Image-guided stereotactic radiosur-
713–717 gery with the CyberKnife for pituitary adenomas. Minim Invasive Neurosurg
[57] Brada M, Burchell L, Ashley S, Traish D. The incidence of cerebrovascular acci- 2005; 48: 91–96
dents in patients with pituitary adenoma. Int J Radiat Oncol Biol Phys 1999; [82] Picozzi P, Losa M, Mortini P, et al. Radiosurgery and the prevention of re-
45: 693–698 growth of incompletely removed nonfunctioning pituitary adenomas. J Neu-
[58] Brada M, Ford D, Ashley S, et al. Risk of second brain tumour after conserva- rosurg 2005; 102 (Suppl): 71–74
tive surgery and radiotherapy for pituitary adenoma. BMJ 1992; 304: 1343– [83] Iwai Y, Yamanaka K, Yoshioka K. Radiosurgery for nonfunctioning pituitary
1346 adenomas. Neurosurgery 2005; 56: 699–705, discussion 699–705
[59] Minniti G, Osti M, Jaffrain-Rea ML, Esposito V, Cantore G, Maurizi Enrici R. [84] Mingione V, Yen CP, Vance ML, et al. Gamma surgery in the treatment of non-
Long-term follow-up results of postoperative radiation therapy for Cushing’s secretory pituitary macroadenoma. J Neurosurg 2006; 104: 876–883
disease. J Neurooncol 2007; 84: 79–84 [85] Voges J, Kocher M, Runge M, et al. Linear accelerator radiosurgery for pitui-
[60] Buatti JM, Friedman WA, Meeks SL, Bova FJ. The radiobiology of radiosurgery tary macroadenomas: a 7-year follow-up study. Cancer 2006; 107: 1355–
and stereotactic radiotherapy. Med Dosim 1998; 23: 201–207 1364
[61] Rowe J, Grainger A, Walton L, Silcocks P, Radatz M, Kemeny A. Risk of malig- [86] Liscák R, Vladyka V, Simonová G, et al. Arteriovenous malformations after
nancy after gamma knife stereotactic radiosurgery. Neurosurgery 2007; 60: Leksell gamma knife radiosurgery: rate of obliteration and complications.
60–65, discussion 65–66 Neurosurgery 2007; 60: 1005–1014, discussion 1015–1016
[62] Tinnel BA, Henderson MA, Witt TC, et al. Endocrine response after gamma [87] Höybye C, Rähn T. Adjuvant Gamma Knife radiosurgery in non-functioning
knife-based stereotactic radiosurgery for secretory pituitary adenoma. Ster- pituitary adenomas; low risk of long-term complications in selected patients.
eotact Funct Neurosurg 2008; 86: 292–296 Pituitary 2009; 12: 211–216
[63] Pollock BE, Nippoldt TB, Stafford SL, Foote RL, Abboud CF. Results of stereotac- [88] Kobayashi T. Long-term results of stereotactic gamma knife radiosurgery for
tic radiosurgery in patients with hormone-producing pituitary adenomas: pituitary adenomas. Specific strategies for different types of adenoma. Prog
factors associated with endocrine normalization. J Neurosurg 2002; 97: 525– Neurol Surg 2009; 22: 77–95
530 [89] Castro DG, Cecílio SA, Canteras MM. Radiosurgery for pituitary adenomas:
[64] Landolt AM, Haller D, Lomax N, et al. Stereotactic radiosurgery for recurrent evaluation of its efficacy and safety. Radiat Oncol 2010; 5: 109
surgically treated acromegaly: comparison with fractionated radiotherapy. J [90] Hayashi M, Chernov M, Tamura N, et al. Gamma Knife robotic microradiosur-
Neurosurg 1998; 88: 1002–1008 gery of pituitary adenomas invading the cavernous sinus: treatment concept
[65] Becker G, Kocher M, Kortmann RD, et al. Radiation therapy in the multimodal and results in 89 cases. J Neurooncol 2010; 98: 185–194
treatment approach of pituitary adenoma. Strahlenther Onkol 2002; 178: [91] Gopalan R, Schlesinger D, Vance ML, Laws E, Sheehan J. Long-term outcomes
173–186 after Gamma Knife radiosurgery for patients with a nonfunctioning pituitary
[66] Emami B, Lyman J, Brown A, et al. Tolerance of normal tissue to therapeutic adenoma. Neurosurgery 2011; 69: 284–293
irradiation. Int J Radiat Oncol Biol Phys 1991; 21: 109–122 [92] Iwata H, Sato K, Tatewaki K, et al. Hypofractionated stereotactic radiotherapy
[67] Estrada J, Boronat M, Mielgo M, et al. The long-term outcome of pituitary irra- with CyberKnife for nonfunctioning pituitary adenoma: high local control
diation after unsuccessful transsphenoidal surgery in Cushing’s disease. N with low toxicity. Neuro-oncol 2011; 13: 916–922
Engl J Med 1997; 336: 172–177 [93] El-Shehaby AM, Reda WA, Tawadros SR, Abdel Karim KM. Low-dose Gamma
[68] Lee CC, Kano H, Yang HC, et al. Initial Gamma Knife radiosurgery for nonfunc- Knife surgery for nonfunctioning pituitary adenomas. J Neurosurg 2012; 117
tioning pituitary adenomas. J Neurosurg 2014; 120: 647–654 (Suppl): 84–88
[69] Xu Z, Ellis S, Lee C-C, et al. Silent corticotroph adenomas following Gamma [94] Runge MJ, Maarouf M, Hunsche S, et al. LINAC-radiosurgery for nonsecreting
Knife radiosurgery. Int J Radiat Oncol Biol Phys 2014 pituitary adenomas. Long-term results. Strahlenther Onkol 2012; 188: 319–
[70] Sheehan JP, Xu Z, Salvetti DJ, Schmitt PJ, Vance ML. Results of gamma knife 325
surgery for Cushing’s disease. J Neurosurg 2013; 119: 1486–1492 [95] Wilson PJ, De-Loyde KJ, Williams JR, Smee RI. A single centre’s experience of
[71] Lüdecke DK, Flitsch J, Knappe UJ, Saeger W. Cushing’s disease: a surgical view. stereotactic radiosurgery and radiotherapy for non-functioning pituitary ad-
J Neurooncol 2001; 54: 151–166 enomas with the Linear Accelerator (Linac). J Clin Neurosci 2012; 19: 370–
[72] Chee GH, Mathias DB, James RA, Kendall-Taylor P. Transsphenoidal pituitary 374
surgery in Cushing’s disease: can we predict outcome? Clin Endocrinol (Oxf) [96] Izawa M, Hayashi M, Nakaya K, et al. Gamma knife radiosurgery for pituitary
2001; 54: 617–626 adenomas. J Neurosurg 2000; 93 (Suppl 3): 19–22
[73] Invitti C, Pecori Giraldi F, de Martin M, Cavagnini F, Study Group of the Italian [97] Shin M, Kurita H, Sasaki T, et al. Stereotactic radiosurgery for pituitary adeno-
Society of Endocrinology on the Pathophysiology of the Hypothalamic-Pitui- ma invading the cavernous sinus. J Neurosurg 2000; 93 (Suppl 3): 2–5
tary-Adrenal Axis. Diagnosis and management of Cushing’s syndrome: results [98] Höybye C, Grenbäck E, Rähn T, Degerblad M, Thorén M, Hulting AL. Adreno-
of an Italian multicentre study. J Clin Endocrinol Metab 1999; 84: 440–448 corticotropic hormone-producing pituitary tumors: 12- to 22-year follow-up

118
Stereotactic Radiosurgery for Pituitary Adenomas

after treatment with stereotactic radiosurgery. Neurosurgery 2001; 49: 284– [112] Ikeda H, Jokura H, Yoshimoto T. Transsphenoidal surgery and adjuvant gam-
291, discussion 291–292 ma knife treatment for growth hormone-secreting pituitary adenoma. J Neu-
[99] Kobayashi T, Kida Y, Mori Y. Gamma knife radiosurgery in the treatment of rosurg 2001; 95: 285–291
Cushing disease: long-term results. J Neurosurg 2002; 97 (Suppl): 422–428 [113] Attanasio R, Epaminonda P, Motti E, et al. Gamma-knife radiosurgery in acro-
[100] Laws ER, Reitmeyer M, Thapar K, Vance ML. Cushing’s disease resulting from megaly: a 4-year follow-up study. J Clin Endocrinol Metab 2003; 88: 3105–
pituitary corticotrophic microadenoma. Treatment results from transsphe- 3112
noidal microsurgery and gamma knife radiosurgery. Neurochirurgie 2002; [114] Muramatsu J, Yoshida M, Shioura H, et al. [Clinical results of LINAC-based
48: 294–299 stereotactic radiosurgery for pituitary adenoma]. Nippon Igaku Hoshasen
[101] Choi JY, Chang JH, Chang JW, Ha Y, Park YG, Chung SS. Radiological and hor- Gakkai Zasshi 2003; 63: 225–230
monal responses of functioning pituitary adenomas after gamma knife radio- [115] Castinetti F, Taieb D, Kuhn JM, et al. Outcome of gamma knife radiosurgery in
surgery. Yonsei Med J 2003; 44: 602–607 82 patients with acromegaly: correlation with initial hypersecretion. J Clin
[102] Wong GK, Leung CH, Chiu KW, et al. LINAC radiosurgery in recurrent Cush- Endocrinol Metab 2005; 90: 4483–4488
ing’s disease after transsphenoidal surgery: a series of 5 cases. Minim Inva- [116] Kobayashi T, Mori Y, Uchiyama Y, Kida Y, Fujitani S. Long-term results of gam-
sive Neurosurg 2003; 46: 327–330 ma knife surgery for growth hormone-producing pituitary adenoma: is the
[103] Devin JK, Allen GS, Cmelak AJ, Duggan DM, Blevins LS. The efficacy of linear disease difficult to cure? J Neurosurg 2005; 102 (Suppl): 119–123
accelerator radiosurgery in the management of patients with Cushing’s dis- [117] Jezková J, Marek J, Hána V, et al. Gamma knife radiosurgery for acromegaly—
ease. Stereotact Funct Neurosurg 2004; 82: 254–262 long-term experience. Clin Endocrinol (Oxf) 2006; 64: 588–595
[104] Castinetti F, Nagai M, Dufour H, et al. Gamma knife radiosurgery is a success- [118] Pollock BE, Jacob JT, Brown PD, Nippoldt TB. Radiosurgery of growth hor-
ful adjunctive treatment in Cushing’s disease. Eur J Endocrinol 2007; 156: mone-producing pituitary adenomas: factors associated with biochemical re-
91–98 mission. J Neurosurg 2007; 106: 833–838
[105] Petit JH, Biller BM, Yock TI, et al. Proton stereotactic radiotherapy for persis- [119] Roberts BK, Ouyang DL, Lad SP, et al. Efficacy and safety of CyberKnife radio-
tent adrenocorticotropin-producing adenomas. J Clin Endocrinol Metab surgery for acromegaly. Pituitary 2007; 10: 19–25
2008; 93: 393–399 [120] Vik-Mo EO, Oksnes M, Pedersen PH, et al. Gamma knife stereotactic radiosur-
[106] Pollock BE, Brown PD, Nippoldt TB, Young WF Jr. Pituitary tumor type affects gery for acromegaly. Eur J Endocrinol 2007; 157: 255–263
the chance of biochemical remission after radiosurgery of hormone-secreting [121] Losa M, Gioia L, Picozzi P, et al. The role of stereotactic radiotherapy in pa-
pituitary adenomas. Neurosurgery 2008; 62: 1271–1276, discussion 1276– tients with growth hormone-secreting pituitary adenoma. J Clin Endocrinol
1278 Metab 2008; 93: 2546–2552
[107] Castinetti F, Nagai M, Morange I, et al. Long-term results of stereotactic radio- [122] Ronchi CL, Attanasio R, Verrua E, et al. Efficacy and tolerability of gamma
surgery in secretory pituitary adenomas. J Clin Endocrinol Metab 2009; 94: knife radiosurgery in acromegaly: a 10-year follow-up study. Clin Endocrinol
3400–3407 (Oxf) 2009; 71: 846–852
[108] Wan H, Chihiro O, Yuan S. MASEP gamma knife radiosurgery for secretory pi- [123] Iwai Y, Yamanaka K, Yoshimura M, Kawasaki I, Yamagami K, Yoshioka K. Gam-
tuitary adenomas: experience in 347 consecutive cases. J Exp Clin Cancer Res ma knife radiosurgery for growth hormone-producing adenomas. J Clin Neu-
2009; 28: 36 rosci 2010; 17: 299–304
[109] Wein L, Dally M, Bach LA. Stereotactic radiosurgery for treatment of Cushing [124] Poon TL, Leung SC, Poon CY, Yu CP. Predictors of outcome following Gamma
disease: an Australian experience. Intern Med J 2012; 42: 1153–1156 Knife surgery for acromegaly. J Neurosurg 2010; 113 (Suppl): 149–152
[110] Grant RA, Whicker M, Lleva R, Knisely JP, Inzucchi SE, Chiang VL. Efficacy and [125] Franzin A, Spatola G, Losa M, Picozzi P, Mortini P. Results of gamma knife ra-
safety of higher dose stereotactic radiosurgery for functional pituitary adeno- diosurgery in acromegaly. Int J Endocrinol 2012; 2012: 342034
mas: a preliminary report. World Neurosurg 2014; 82:195–201 [126] Lee CC, Vance ML, Xu Z, et al. Stereotactic radiosurgery for acromegaly. J Clin
[111] Fukuoka S, Ito T, Takanashi M, Hojo A, Nakamura H. Gamma knife radiosur- Endocrinol Metab 2014; 99: 1273–1281
gery for growth hormone-secreting pituitary adenomas invading the caver-
nous sinus. Stereotact Funct Neurosurg 2001; 76: 213–217

119
Stereotactic Radiosurgery for Chordomas

14 Stereotactic Radiosurgery for Chordomas


Dan Kunaprayoon, Ananth Charya, Jay S. Loeffler, and Helen A. Shih

chordomas was higher in whites (91.2%) as compared with


Key Points blacks (2.2%) and other races/ethnicities (6.5%). Furthermore,
the annual incidence rate of chordomas in whites was four-fold
● Chordomas are rare, indolent, but locally aggressive tumors greater than blacks. The average age of diagnosis for chordomas
of the skull base and spine with a propensity for recurrence. is typically in the sixth decade, with a worse prognosis associ-
● Stereotactic radiosurgery allows for increased relative biolog- ated with diagnosis at 70 years or greater. Chordomas have a
ical effectiveness compared with conventionally fractionated lower incidence in ages < 40, with < 5% of cases occurring in chil-
radiotherapy, which can be particularly meaningful for a dren or adolescents.3 Survival analysis of 594 patients with cra-
disease requiring a high fractionated dose for response. nial chordomas within the SEER database demonstrated 3-, 5-,
● Stereotactic radiosurgery for chordomas has been used in and 10-year relative survival rates of 80.9, 73.5, and 58.7%,
different settings, most prominently in the management of respectively.6 Both males and females had similar 5-year (71.9%
patients with recurrent or residual disease. vs. 75.4%) and 10-year (57.8% vs. 59.3%) survival probabilities.
● The wide range of local control rates reported reflects the Individuals older than 50 years and less than 10 years at diag-
variability of patient populations and treatment settings. nosis face worse prognosis as compared with individuals of
● High margin dose and smaller tumor volumes are associated ages 11 to 49 years.
with improved local control.
● Dosimetric advantages of stereotactic radiosurgery allow for
favorable trade-offs between tumor control and adverse radi- 14.3 Pathogenesis
ation events.
Chordomas arise from the remnants of the notochord that con-
tinue to reside within the vertebrae and axial skeleton after
involution of the embryonic notochord. Human embryo and
14.1 Introduction fetus studies in addition to cell-differentiation-tracking experi-
ments indicate these notochordal remnants preferentially
Chordomas are rare, indolent, but locally aggressive tumors of locate to anatomical sites that correspond to the typical loca-
the skull base and spine that arise from the embryological rem- tions of chordoma lesions.2 Moreover, molecular phenotyping
nants of the notochord with dural epithelial–mesenchymal ori- experiments demonstrate similar morphology between noto-
gin.1 These tumors are often not clinically evident until late chordal nest cells and those obtained from chordomas.
stages due to invasion and local destruction of nearby tissues. The notochordal hypothesis is further supported by the dem-
Indistinct margins, local invasiveness, high tumor burden, and onstration of transcription factor T (brachyury) gene duplica-
proximity to critical bony, vascular, and neural structures create tion in individuals with familial chordoma.7 Brachyury is
significant challenges in the clinical management of these neo- expressed in normal, undifferentiated embryonic notochord in
plasms.2 Chordomas may occur anywhere along the spine and the axial skeleton and is known to be overexpressed in cases of
skull base, but most commonly arise from the spheno-occipital sporadic chordoma as well. Individuals with familial chordoma
or sacrococcygeal regions. The distribution between cranial, possess unique duplications, which consist solely of the bra-
mobile spine, and sacral regions is approximately even, with chyury gene, within the 6q27 chromosomal regions as demon-
frequencies of 30.0, 29.2, and 32.8%, respectively.3 With skull strated by high-resolution-array comparative genomic
base presentations, chordomas frequently cause destruction of hybridization.8
the clivus with subsequent invasion of the posterior fossa and
compression of the brainstem or the nasopharynx. 4 Cranial
chordomas may present with cranial neuropathies, headache, 14.4 Histopathology and
endocrinopathy, or hemorrhage.5 Although considered to be of
low metastatic potential, late-stage diagnosis of chordomas Radiographic Features
increases the likelihood of distant metastases.1 Five percent of
First recognized by Virchow in 1857, chordomas display charac-
chordomas metastasize, with likely sites including the brain,
teristic vacuolated physaliferous cells and display variable levels
bones, lung, and skin at the time of initial diagnosis.2
of histological atypia. However, the association between histo-
pathological features and biological activity remains to be eluci-
dated.2 Chordomas can classically be categorized into three
14.2 Epidemiology different histological variants as follows: (1) classical, which
Recent population-based studies using data from the Surveil- exhibit lobulated masses comprising of groups of cells sepa-
lance, Epidemiology, and End Results (SEER) program of the rated by fibrous septa with pathognomonic vacuolated cyto-
National Cancer Institute (NCI) estimated that chordomas plasm; (2) chondroid, which display characteristics of both
account for 1 to 4% of primary bone malignancies. Chordomas chordoma and chondrosarcoma; and (3) dedifferentiated. Tradi-
are rare malignancies with an incidence rate of 0.089 per tionally, chordomas are recognized by positive immunohisto-
100,000.3 Incidence rates are higher in men as compared with chemical staining for cytokeratin markers such as S-100 and
women (0.10 per 100,000 vs. 0.06 per 100,000). Prevalence of epithelial membrane antigen (MUC-1). Staining for brachyury

120
Stereotactic Radiosurgery for Chordomas

may provide a more sensitive and specific biomarker for detect- tumor margins.14 External beam radiotherapy with convention-
ing combined with traditional assays for cytokeratin staining.2 al fractionation involves daily treatments, five times per week,
Radiographically, chordomas typically appear as midline over approximately 7 to 8 weeks to deliver an effective cumula-
lesions with destructive bony involvement. On magnetic reso- tive radiation dose. External beam radiotherapy for skull base
nance imaging (MRI) and computed tomography (CT), these chordomas employs both image-guided intensity-modulated
lesions appear as soft-tissue masses.9 Specifically, on MRI, these radiotherapy and conventional fractionated stereotactic radio-
tumors display as isointense or hypointense to the surrounding therapy to minimize radiation dose to nearby structures while
musculature, which is indicative of calcification and bony inva- optimizing target conformality.15 Despite the advancements in
sion on T1-weighted images.2 On T2-weighted images, chordo- delivery with better conformality indices, photons still have a
mas display high signal, inhomogeneous texture. With relatively high exit dose (beyond the tumor target), resulting in
gadolinium, patchy to dense enhancement can be seen.9 Chor- significant normal-tissue collateral irradiation.
domas have decreased uptake of radioisotope on bone scan as
compared with other bone malignancies.
14.5.3 Charged Particle Therapy
Proton and carbon radiotherapy permits higher radiation doses
14.5 Treatment Options to tumors with less exposure to surrounding critical structures
due to their physical beam characteristics.16–20 Due to their
14.5.1 Surgery mass and charge, accelerated particles have a characteristic
Surgical management is the traditional initial management of beam profile such that the integral dose in normal tissue is less
choice for skull base chordomas. A variety of broad surgical both proximally and distally to tumor targets in addition to
approaches are employed, which are dependent on tumor loca- achieving a concentration of dose within the target.16,17 These
tion and surgical preference.10 These include transsphenoidal, facilities are complex and expensive to maintain. There are a
transmaxillary, transnasal, high anterior cervical retropharyng- limited number of treatment centers that constrain this as a
eal, and transoral approaches, as well as minimally invasive resource for patient care.
endoscopic methods.2 An aggressive surgical approach achiev-
ing maximal radical resection must be balanced with reducing
operative morbidity and preserving critical neuroanatomical
14.5.4 Stereotactic Radiosurgery
structures, such as the brainstem, cranial nerves, pituitary, and Stereotactic radiosurgery (SRS) allows for several advantages
cavernous sinus. In light of the above, surgical approaches may compared with conventional radiotherapy. The use of high dose
be directed at ameliorating brainstem and optic nerve tumor per fraction is associated with an increased relative biological
compression and reducing tumor volume to set the stage for effectiveness (RBE), especially important in the treatment of
future radiotherapy.11 Even when gross total resection is chordomas, which are slow growing and have traditionally
achieved, local tumor recurrence rates can be high. In a series of been viewed as relatively radioresistant tumors.5 Stereotactic
74 patients who underwent surgical resection for skull base radiosurgery can be delivered as a single fraction or few frac-
chordomas, gross total resection was achieved in 53 patients. tions (typically 2–5 sessions). Although there are no long-term
Among these patients, the 10-year recurrence-free survival was prospective data guiding the use of fractionated stereotactic
31%.10 Furthermore, in patients who underwent surgical resec- radiotherapy versus single-fraction radiosurgery, tumor vol-
tion without subsequent adjuvant radiotherapy, local recur- ume, target conformality, setup uncertainty, and dose gradient
rence may be as high as 70%.1 In light of the above, the are taken into account during planning to determine number of
paradigmatic management strategy for skull base chordomas fractions for treatment. Stereotactic radiosurgery offers a clear
typically involves both maximal resection and adjuvant radio- practical and logistical advantage over several weeks of treat-
therapy. Exceptions are small lesions that can be gross totally ment courses of conventional photon or particle therapies.
resected, with unlikely residual microscopic disease. These Several factors should be considered when undergoing SRS
patients can be curatively managed with surgery alone. planning. High-resolution thin-slice MRI with long TR (repeti-
tion time) and contrast offers better target definition. This is
especially important in the case of recurrence or prior failed
14.5.2 Conventional Radiation Therapy radiotherapy. However, MRI alone does not define calcification,
En bloc resection is achievable in a low percentage of patients cortical bone, or destructive edges well, and should be comple-
with skull base chordomas, and as a result, adjuvant radiother- mented by high-resolution CT with attention to bone and soft-
apy reduces the local recurrence compared with surgery alone. tissue windows that may decrease out-of-field tumor recur-
Postoperative irradiation is limited by adjacent anatomical rence.21 To avoid geographic misses, some groups suggest liber-
structures; the tolerance dose of the brainstem and cranial al inclusion of the whole bony clivus and consideration of a
nerves is lower than the effective treatment dose necessary for boost dose.13 Beam-blocking techniques and conformal plan-
local tumor control. Chordomas are ineffectively treated with ning with multiple isocenters with automatic positioning sys-
radiation doses < 60 Gy.1 Patient series employing conventional tems should be employed to decrease risks of complications,
radiotherapy with doses between 40 and 60 Gy demonstrate especially when the target volume is located close to critical
local control rates nearing 50%.2,12 Others have advocated for structures such as the brainstem or optic nerves.
doses approaching 80 Gy.13 Technological advances in photon Stereotactic radiosurgery can be delivered with different
radiotherapy allow for greater targeting accuracy with highly techniques. These include Gamma Knife (GK), CyberKnife (CK),
conformal techniques to allow for steep dose reductions at the and linear accelerator (linac)-based treatments. The majority of

121
Stereotactic Radiosurgery for Chordomas

the reported series utilizes GK, which uses a hemispheric array both univariate and multivariate analyses, corresponding to 5-
of cobalt-60 sources delivering photon radiation converging on and 10-year long-term control rates of 87 and 76% in tumors <
a focal point. Patients are immobilized with a fixed frame to 20 cc and 51 and 51% in tumors ≥ 20 cc.25 This corroborates ear-
achieve positioning accuracy < 0.5 mm. Dose is typically pre- lier experience with proton therapy associating tumor volume <
scribed to the 50% isodose line, maximizing dose at the center 25 cc with long-term control.32
of the target and minimizing dose at the target edge with a Other reports portray weaker association between tumor
sharp dose falloff.14 volume and long-term control. The North American Gamma
The CK consists of a robotic, image-guided stereotactic sys- Knife Consortium aggregated outcomes of 71 patients from six
tem with a linac mounted on a fully articulated robotic arm academic medical centers who underwent GKSRS for intracra-
capable of 6 degrees of freedom without external frame–based nial chordomas. They reported 5-year long-term control rates
fixation.22 As a result, an advantage of the CK system is the of 82% for volumes < 7 cc and 57% for volumes ≥ 7 cc that was
capacity to treat tumors with complex shapes or difficult loca- significant on univariate but not multivariate analysis. 28 Koga et
tions that are otherwise challenging to treat using frame-based al evaluated 14 patients with skull base chordomas or chondro-
systems. Similar to other treatment modalities, radiation dose sarcomas undergoing GK and also did not find tumor volume a
to the tumor margin is planned based on volume, location, and significant factor for long-term control on univariate or multi-
a history of prior radiotherapy.23 The treatment-planning sys- variate analysis.33 Martin et al did not find any of their factors,
tem of the CK affords the flexibility to select either isocentric- including tumor volume, to reach statistical significance on
or nonisocentric-based methods. However, nonisocentric radia- multivariate analysis in their series of 28 patients with chordo-
tion delivery characteristically results in greater homogeneity mas or chondrosarcomas undergoing GK as primary or adjuvant
in coverage and steeper dose falloff outside the target mar- treatment.34 Nonetheless, smaller residual tumor volumes may
gins.22 Thus, nearby critical structures can be delineated and be indicative of more complete up-front resection. They are also
spared based on dose-weighted contouring of the tumor vol- potentially better candidates for repeat radiosurgery with fewer
ume. Moreover, the CK affords the ability to deliver fractionated side effects depending on degree of overlap.27
radiosurgical regimens, which further reduces the radiation risk Although dose and volume are thought to be the main
to critical structures adjacent to the lesion. parameters, other factors have been found to be associated with
Linac SRS employs photons generated from a traditional linac tumor control and survival. Younger age was reported as a
produced by accelerating electrons onto a metal target. Treat- favorable predictor for both survival and tumor control on uni-
ment can be planned using dynamic conformal arcs or with variate and multivariate analysis, which has not been seen in
multiple beams with the goal of producing a highly conformal other studies.13,25,28,29,33,34 Gender has not been shown to be an
dose gradient delivering differential dose to target and normal associated risk factor.13,25,28,34 Presentation and timing of treat-
tissues. This can be achieved with beam modulation, multiple ment were also examined, and they found that < 2 cranial nerve
isocenters, and micro-multileaf collimation. Compared with GK, deficits were associated with survival but not tumor control on
linac SRS is more homogeneous, but may be less conformal multivariate analysis. Symptom duration before SRS or interval
when irradiating irregularly shaped targets.24 Protons are also from diagnosis to SRS was not associated with longer tumor
being developed for delivery of SRS, although this is still in an control.
early stage and there are a limited number of facilities with Perhaps the most obvious variable in disease control outcome
proton therapy compared with the wider availability of photon is radiation dose delivered. The composite findings of several
SRS. studies suggest a margin dose ≥ 15 Gy is required for long-term
There is a wide range of local control rates reported control.13,25,28,35 Hasegawa et al reported that a margin dose ≥
(▶ Table 14.1).13,21,25–30 Reported overall survival at 5 years is 15 Gy and maximum dose ≥ 30 Gy approached statistical signifi-
generally higher than local control, consistent with local failure cance on univariate analysis, but not on multivariate analysis. 25
as the common site of disease progression more commonly In another series of 25 chordoma patients treated at the Mayo
than systemic disease. Few studies report long-term outcomes Clinic, a subset analysis demonstrated a 4-year actuarial control
of at least 10 years. A series from Japan treating 37 patients rate of 50% for patients treated to < 15 Gy and 100% for patients
with chordomas and chondrosarcomas of the skull base treated to ≥ 15 Gy.13 Koga et al found dose of treatment to be
reported 5- and 10-year actuarial local control of 76 and 67%, the only factor on multivariate analysis to affect long-term con-
and overall survival of 80 and 53%.25 trol, requiring doses > 16 Gy.33 Kondziolka et al demonstrated
A number of factors determine treatment outcomes such as no progression of small tumors < 30 cc when treated to a mar-
tumor size or volume at time of irradiation. Patients presenting gin dose > 20 Gy.35 Reports from smaller series have not found
with large tumor volume are especially challenging because margin dose to be a significant factor for tumor control. 26,34
the most effective first-line therapy, surgery, is unlikely to However, prescribed doses or tumor volume alone can
completely resect disease, even with aggressive skull base obscure the clinical picture. For example, patients with prior
approaches. Pamir et al reported a series of 26 skull base chor- radiotherapy were prescribed significantly lower doses and also
domas patients treated with surgery and radiation, with seven had larger tumor volumes than patients without prior radio-
patients receiving GK radiosurgery. Data from this study therapy, complicating the interpretation of these results.28
describe inevitable tumor progression after surgery in tumors Prior treatment with either surgery or radiotherapy may be
with initial volumes ≥ 20 cc.31 Based on this observation, their associated with long-term tumor control and survival. Kano
treatment algorithm recommends immediate adjuvant radio- et al found that not having prior radiotherapy was associated
surgery for residual tumors ≤ 30 cc. In the study by Hasegawa et with improved patient survival, but not tumor control on mul-
al, only tumor volume was significant for long-term control on tivariate analysis.28 Other series have not found prior resection,

122
Table 14.1 Studies and patient characteristics in published chordoma series treated with SRS
Study N Disease location Setting Interval SRS Median/mean Median/mean Normalization 5-y LC 5-y PFS 5-y OS Notes
between technique tumor volume margin dose
Dx and SRS (cm3) (Gy)
(mo)

9 clival Includes chondrosar-


Cho et al, 10 sphenopetroclival Adjuvant, comas. Only 6 patients
30 — GK 4.57 17 (15–20) 50–70% — 40% 80%
200830 2 petrous bone salvage with chordomas
9 not reported receiving SRS

10 clivus
Dassoulas et Adjuvant,
15 2 sphenopetroclival — GK 5.8 15.3 — 42.6% — — —
al, 200926 definitive
3 cavernous sinus

Hasegawa et Adjuvant, Includes


37 Skull base — GK 19.7 14 (9–20) — 72% 42% 80%
al, 200725 definitive chondrosarcomas.

Hauptman et 2 clivus 5 patients received


15 Adjuvant — Linac 12.3 15.5 90% — — —
al, 201136 3 cavernous sinus SRS

6 clivus
1 of upper clivus,
Henderson et cavernous sinus, or- Adjuvant, Includes extracranial
18 — CK 128 35 in 5 fractions 75% (60–84%) 59.1% — 74.3%
al, 200922 bits definitive chordomas
11 vertebral or sa-
cral

Adjuvant,
Ito et al,
19 19 clivus definitive, — GK 3.3 17.8 — — 47.9% — —
201027
salvage

10 clivus
2 clivus/sella 55% overall,
Jiang et al, Adjuvant, Includes extracranial
20 1 of cavernous sinus, 1–192 CK 16.1 32.5 (18–50) — 46% intracra- — 52.5%
201223 salvage chordomas
parasellar, and clival nial
7 vertebral or sacral

49 clivus
11 sphenopetroclival Adjuvant,
Kano et al, 14.6 (0.5– 28% underwent prior
71 1 craniocervical definitive, GK 7.1 15 — — — 80%
201128 297) RT
junction salvage
10 cavernous sinus

Koga et al, Includes


14 Skull base Adjuvant 8–96 GK 12.4 18 — — 43% —
201033 chondrosarcomas.

Krishnan et al, Adjuvant, Includes


34 — — GK 14.4 15 — 32% 67% —
200513 salvage chondrosarcomas.

Liu et al, Adjuvant, Includes


31 Skull base — GK 11.4 12.7 44% (35–60%) 21.4% — 75.8%
200821 salvage chondrosarcomas.
Stereotactic Radiosurgery for Chordomas

123
Stereotactic Radiosurgery for Chordomas

number of prior resections, or prior radiotherapy to be associ-

includes those who did

Abbreviations: CK, CyberKnife; Dx, diagnosis; GK, Gamma Knife; LC, local control; OS, overall survival; PFS, progression-free survival; Rx, prescription; linac, linear accelerator; SRS, stereotactic radiosurgery; RT,
ated with tumor control.13,34 A series from China found extent

Includes extracranial

Reported outcomes
of resection to correlate with progression-free survival (PFS)

Seven patients
not receive RT
and overall survival (OS), but not recurrence.29 They reported

received SRS
chordomas
dismal 5- and 8-year PFS of 45 and 18%, and OS of 71 and 46%. 29
These long-term results are consistent with the natural history
Notes

of the disease, with patients exhibiting multiply recurrent and


late failures.31
The limited data suggest mixed findings with regard to possi-
5-y OS

62.9%

70.9%
ble significance of histology with outcomes. Ouyang et al


reported PFS rates by chordoma subtype, reporting 3-year PFS
of 86, 100, and 40% for classic chordoma, chondroid type, and
5-y PFS

dedifferentiated type, respectively.29 The difference between


45%


classic and chondroid subtypes was statistically significant. Ito
et al found MIB-1 labeling index to correlate with progression-
free period on simple regression with an optimum cutoff point
of 3.44%, the only factor found to significantly affect recur-
5-y LC

62.9%

rence.27 Other studies have not found histopathology to be a



significant factor.25,33
A few SRS series characterized their failures as in-field versus
Normalization

out-of-field. Liu et al reported with mean imaging follow-up of


28 months overall tumor progression in 9 of 31 patients (29%),
57.1%

with the majority (78%) occurring out-of-field.21 Krishnan et al


50%

showed a more even split between in-field (43%) and out-of-


field (57%), with an overall rate of 7 of 25 (28%) patients with
Median/mean
margin dose

evidence of progression on MRI at a median follow-up of 4.5


years.13 Hasegawa et al reported a higher proportion of in-field
failures (9/16, 56%) versus out-of-field failures (44%), as well as
16.5

16.2
(Gy)

a higher overall failure rate of 43% at a mean follow-up of 59


months.25
tumor volume
Median/mean

The tumor size and median tumor margin dose among these
series are variable, with tumor sizes ranging from 11.4 to 19.7
< 3 cm
(cm3)

cc and margin dose from 12.7 to 15 Gy. Despite variable follow-


12.4
9.8

up and patient populations, the reported data suggest an asso-


ciation between tumor size and in-field failure, with larger
tumors associated with a higher proportion of in-field failures.
technique

The association with margin dose is less clear.


These outcomes underscore the importance of dose and vol-
SRS

GK

GK

GK

ume parameters on local control and raise questions regarding


Dx and SRS

the mechanisms of in-field versus out-of-field failures. Sug-


between
Interval

gested hypotheses for in-field failures include insufficient dose


0–133
(mo)

or inherent biological resistance, whereas geographic misses


are thought to relate to out-of-field failures.13


Plan heterogeneity is postulated to have a role in out-of-field
Definitive,

definitive,
Adjuvant,

Adjuvant,

failures.25 Use of multiple isocenters in treatment planning to


salvage

salvage

salvage
Setting

provide an aggregate dose necessitates overlap of high-dose


spheres and ultimately nonuniform target dose. This is common
in Gamma Knife radiosurgery, which is typically prescribed
such that the 50% isodose line encompasses the target. Gamma
12 middle cranial
Disease location

3 craniocervical
5 multiple sites

Knife radiosurgery results in a heterogeneous dose distribution


with a decreasing dose gradient from the target center to the
Skull base

Skull base
21 sellar
36 clivus

margins, which may lead to an increasing risk of tumor recur-


fossa

rence outside the treatment volume due to underdosing at the


margin. This can be magnified in cases where the dose at the
Table 14.1 continued

margin is reduced to spare nearby critical structures.


28

77

26

radiation therapy.
N

Chordoma growth patterns, surgery, and postoperative imag-


Ouyang et al,

ing may also affect the rate of out-of-field failures.23,25,33 Chor-


Martin et al,

Pamir et al,

domas are invasive and destructive of both bone and adjacent


200734

201329

200431
Study

soft tissue, leading to indistinct margins even with combined


use of CT and MRI for treatment planning. Postoperative

124
Stereotactic Radiosurgery for Chordomas

changes and specific surgical approach, which may entail hypofractionated radiosurgery over two or more fractions.21 A
extraction of tumor piecemeal, can contribute to greater uncer- number of studies detail instances of adverse radiation events
tainty of the target volume. For patients with out-of-field fail- in chordoma patients treated with SRS (▶ Table 14.2).
ures, repeat radiosurgery remains an option for further In a series of 25 patients with skull base chordomas treated
control.25,26 with SRS, 34% experienced adverse radiation events, with the
Patients treated with other SRS modalities, including Cyber- most common complication being cranial nerve dysfunction. 13
Knife and linac, exhibit outcomes within the reported range of Per Hauptman et al, one patient who underwent SRS experi-
Gamma Knife series. Henderson et al reported a series of 18 enced lower cranial nerve dysfunction after receiving 7.78 Gy to
patients who underwent CyberKnife radiosurgery (CKRS) for 21.11% of the brainstem at the 50% isodose line, which was sig-
either clival or extracranial chordomas. Unlike other series, nificantly greater brainstem exposure as compared with other
Henderson et al delivered treatment in five fractions. Overall patients within the series.36 To minimize brainstem complica-
quality-of-life measures of patients improved and remained tions, it is recommended that exposure dose should be held to
stable.22 Jiang et al presented a series of 20 patients receiving less than 12 Gy. Cranial nerve deficits resulting from tumor exit
CKSRS as either primary adjuvant therapy or for tumor radiation dose may include, but is not limited to, ocular paraly-
recurrence (▶ Table 14.1). Within this study, postoperative sis, diplopia, dysarthria, and dysphagia.13,21,33
clinical improvement was achieved despite a larger volume As with conventional radiotherapy, endocrinopathy has been
than many of the other series.23 Hauptman et al reported reported as a potential complication of SRS as well. Krishnan et
their linac experience treating 15 patients with skull base al reported anterior pituitary dysfunction in 10% of patients. 13
chordomas and chondrosarcomas.36 Thirteen of these In addition, in a series of 31 patients, Liu et al reported one pa-
patients had chordoma, and five of them were treated with tient treated with GK who experienced pituitary dysfunction
SRS. Three patients remained stable, one recurred, and one 18 months after radiosurgery.21 However, in a series of 15
was lost to follow-up. No differences in radiation dosage or patients with skull base chordomas treated with either linac
volume were noted between those with control and the pa- stereotactic radiotherapy or SRS, no patients treated with SRS
tient that failed. displayed endocrinopathy, whereas patients who underwent
When SRS is used in the recurrent adjuvant or salvage set- fractionated radiotherapy experienced pituitary endocrinopa-
ting, outcomes are suboptimal compared with its use in the pri- thies at cumulative doses > 30 Gy.36
mary adjuvant setting. Jiang and colleagues reported that only Radiation-induced brain necrosis and temporal lobe toxicity
29% of patients treated for recurrence had stable or improved has been observed in both photon and proton-based radiother-
outcomes in the recurrent setting compared with 82% in the apy. In 62 patients with skull base tumors treated with proton
primary adjuvant setting.23 Nevertheless, repeat SRS can offer irradiation from 1998 to 2005, 7 patients demonstrated tempo-
improved local control. Dassoulas et al reported that 5-year ral lobe damage, ranging from asymptomatic grade 1 to symp-
actuarial tumor control after one Gamma Knife radiosurgery tomatic grade 3 toxicity, associated with a fractionated relative
(GKRS) was 43% and increased slightly to 50% when including biological effective dose > 60 Gy.37,38 In patients treated with
patients who underwent repeat GKRS, albeit this was among a SRS, temporal lobe toxicity has been demonstrated to be a
small series of 15 patients.26 Stereotactic radiosurgery in com- rarely encountered complication except in the case of reirradia-
bination with fractionated radiotherapy may increase the risk tion. Hauptman et al described one patient who developed
of adverse events; the timing of SRS should be carefully consid- temporal lobe radiation-related changes after reirradiation for
ered as well.33 recurrent chordoma, including feelings of déjà vu and general-
ized anxiety. These symptoms subsequently resolved and may
be attributed to recurrent radiation treatments.36 Krishnan et al
reported that 17% of patients receiving SRS experienced radi-
14.6 Complications ation-induced brain necrosis and temporal lobe toxicity. 13 Of
Given the often challenging location and relative radioresist- note, all of these patients had received external beam radio-
ance of skull base chordomas, injury secondary to radiation is a therapy prior to radiosurgery. For patients previously with-
significant risk. The likelihood of radiation-related complica- out irradiation, radiosurgery alone may reduce the risk of
tions in the management of these tumors can be influenced by complications.
characteristics of treatment plans, including dose, duration of Reirradiation due to recurrent chordoma can increase the
treatment, fractionation, type of radiation delivered, and the likelihood of adverse radiation events, although the association
ability to conform the dose to tumor contours. Although several between repeat radiation and treatment-related complications
series demonstrate adverse radiation events associated with is not well documented. Fractionated photon or proton-based
conventional radiotherapy, including endocrinopathies, cranial radiotherapy for skull base chordomas prior to future single-
nerve deficits, and temporal lobe toxicity, the association with fraction SRS confounds analysis of potential radiation-associ-
single-fraction radiosurgery and radiation-related complica- ated complications. For example, the high percentage of adverse
tions has been poorly characterized.36 Fractionated radiation, radiation events in patients who underwent CKRS as reported
particularly with more conformal techniques such as the use of by Krishnan et al can be mitigated by the fact that no patients
proton therapy, may lower the risk of radiation-related compli- who underwent SRS alone experienced treatment complica-
cations in adjacent critical structures due to reduced tumor exit tions.13 In patients who underwent CKRS alone, Jiang et al
dose, but, in general, SRS is also associated with low morbidity reported no postradiosurgery radiation-induced complications
due to its high dose conformality.26,28 An alternative to decrease in patients with clival chordomas.23 With respect to repeat SRS,
radiation injury risks of single-fraction radiosurgery is in a series of 37 patients with skull base chordomas treated

125
Stereotactic Radiosurgery for Chordomas

Table 14.2 Complication rates in published chordoma series treated with SRS
Study Improved symptoms (%) Overall complication rate (%) Complications

Hasegawa et al, 200725 44.8 3 Facial numbness

Pituitary dysfunction, lower cranial nerve palsies,


Hauptman et al, 201136 — 13.3
transient temporal lobe radiation injury

Henderson et al, 200922 — 50 Diplopia, lower cranial nerve palsies

PBRT-induced visual deterioration and gait dis-


Jiang et al, 201223 30.7 15 turbance, leg weakness, no SRS-related complica-
tions

Kano et al, 201128 — 9 CN V–VII neuropathy, anterior pituitary dysfunction

Koga et al, 201033 — 10 Diplopia

Diplopia, ocular neuromyotonia, hearing loss, dys-


Krishnan et al, 200513 — 34.5 arthria, dysphagia, radiation necrosis, anterior
pituitary dysfunction

Liu et al, 200821 75 13 Pituitary dysfunction, ocular paralysis

Martin et al, 200734 — 5.5 Dizziness, nausea

Abbreviations: CN, cranial nerve; PBRT, proton beam radiotherapy; SRS, stereotactic radiosurgery.

with multiple GK treatments, reportedly no patients experi- [4] Gangadhar K, Santhosh D. Radiopathological evaluation of primary malignant
skull tumors: a review. Clin Neurol Neurosurg 2012; 114: 833–839
enced complications.25 However, Henderson et al reported
[5] Kano H, Lunsford LD. Stereotactic radiosurgery of intracranial chordomas,
three patients who had previously received conventional radio- chondrosarcomas, and glomus tumors. Neurosurg Clin N Am 2013; 24:
therapy and/or GK who developed diplopia and lower nerve 553–560
cranial palsies after CKRS.22 [6] Chambers KJ, Lin DT, Meier J, Remenschneider A, Herr M, Gray ST. Incidence
and survival patterns of cranial chordoma in the United States. Laryngoscope
[7] Vujovic S, Henderson S, Presneau N, et al. Brachyury, a crucial regulator of no-
14.7 Summary tochordal development, is a novel biomarker for chordomas. J Pathol 2006;
209: 157–165
Although maximum safe resection remains the primary treat- [8] Yang XR, Ng D, Alcorta DA, et al. T (brachyury) gene duplication confers major
ment, SRS has an important role in the multimodality manage- susceptibility to familial chordoma. Nat Genet 2009; 41: 1176–1178
[9] Soo MY. Chordoma: review of clinicoradiological features and factors affect-
ment of chordomas. It has been shown to be a safe and effective
ing survival. Australas Radiol 2001; 45: 427–434
treatment option for patients with residual or recurrent dis- [10] Tzortzidis F, Elahi F, Wright D, Natarajan SK, Sekhar LN. Patient outcome at
ease, and in some cases as definitive treatment for patients with long-term follow-up after aggressive microsurgical resection of cranial base
small-volume tumors. There is a wide range of outcomes, chordomas. Neurosurgery 2006; 59: 230–237, discussion 230–237
including local control, overall survival, and complication rates, [11] Yasuda M, Bresson D, Chibbaro S, et al. Chordomas of the skull base and cer-
vical spine: clinical outcomes associated with a multimodal surgical resection
reflecting the variability of patient populations and treatment
combined with proton-beam radiation in 40 patients. Neurosurg Rev 2012;
settings. Retrospective studies have examined the association of 35: 171–182, discussion 182–183
a number of prognostic factors, with high margin dose and [12] Catton C, O’Sullivan B, Bell R, et al. Chordoma: long-term follow-up after radi-
smaller tumor volume as most consistently associated with cal photon irradiation. Radiother Oncol 1996; 41: 67–72
[13] Krishnan S, Foote RL, Brown PD, Pollock BE, Link MJ, Garces YI. Radiosurgery
improved patient outcomes. Delivery of an appropriately high
for cranial base chordomas and chondrosarcomas. Neurosurgery 2005; 56:
margin dose can be challenging given anatomical constraints 777–784, discussion 777–784
and dose-limiting critical structures. However, dosimetric [14] Amichetti M, Amelio D, Minniti G. Radiosurgery with photons or protons for
advantages of SRS allow for favorable trade-offs between tumor benign and malignant tumours of the skull base: a review. Radiat Oncol
control and adverse radiation events. Stereotactic radiosurgery 2012; 7: 210
[15] Bugoci DM, Girvigian MR, Chen JC, Miller MM, Rahimian J. Photon-based frac-
may be considered as primary adjuvant therapy for residual
tionated stereotactic radiotherapy for postoperative treatment of skull base
small-volume chordomas. chordomas. Am J Clin Oncol 2013; 36: 404–410
[16] Noël G, Feuvret L, Calugaru V, et al. Chordomas of the base of the skull
and upper cervical spine. One hundred patients irradiated by a 3D confor-
References mal technique combining photon and proton beams. Acta Oncol 2005; 44:
700–708
[1] Chugh R, Tawbi H, Lucas DR, Biermann JS, Schuetze SM, Baker LH. Chordoma:
[17] Noël G, Feuvret L, Ferrand R, Boisserie G, Mazeron JJ, Habrand JL. Radio-
the nonsarcoma primary bone tumor. Oncologist 2007; 12: 1344–1350
therapeutic factors in the management of cervical-basal chordomas and
[2] Walcott BP, Nahed BV, Mohyeldin A, Coumans JV, Kahle KT, Ferreira MJ. Chor-
chondrosarcomas. Neurosurgery 2004; 55: 1252–1260, discussion 1260–
doma: current concepts, management, and future directions. Lancet Oncol
1262
2012; 13: e69–e76
[18] Schulz-Ertner D, Nikoghosyan A, Thilmann C, et al. Carbon ion radiotherapy
[3] McMaster ML, Goldstein AM, Bromley CM, Ishibe N, Parry DM. Chordoma: in-
for chordomas and low-grade chondrosarcomas of the skull base. Results in
cidence and survival patterns in the United States, 1973–1995. Cancer Causes
67 patients. Strahlenther Onkol 2003; 179: 598–605
Control 2001; 12: 1–11

126
Stereotactic Radiosurgery for Chordomas

[19] Schulz-Ertner D, Karger CP, Feuerhake A, et al. Effectiveness of carbon ion ra- [29] Ouyang T, Zhang N, Zhang Y, et al. Clinical characteristics, immunohisto-
diotherapy in the treatment of skull-base chordomas. Int J Radiat Oncol Biol chemistry, and outcomes of 77 patients with skull base chordomas. World
Phys 2007; 68: 449–457 Neurosurg 2014; 81: 790–797
[20] Schulz-Ertner D, Haberer T, Jäkel O, et al. Radiotherapy for chordomas and [30] Cho YH, Kim JH, Khang SK, Lee JK, Kim CJ. Chordomas and chondrosarcomas
low-grade chondrosarcomas of the skull base with carbon ions. Int J Radiat of the skull base: comparative analysis of clinical results in 30 patients. Neu-
Oncol Biol Phys 2002; 53: 36–42 rosurg Rev 2008; 31: 35–43, discussion 43
[21] Liu AL, Wang ZC, Sun SB, Wang MH, Luo B, Liu P. Gamma knife radiosurgery [31] Pamir MN, Kiliç T, Türe U, Ozek MM. Multimodality management of 26 skull-
for residual skull base chordomas. Neurol Res 2008; 30: 557–561 base chordomas with 4-year mean follow-up: experience at a single institu-
[22] Henderson FC, McCool K, Seigle J, Jean W, Harter W, Gagnon GJ. Treatment of tion. Acta Neurochir (Wien) 2004; 146: 343–354, 354
chordomas with CyberKnife: Georgetown University experience and treat- [32] Hug EB, Loredo LN, Slater JD, et al. Proton radiation therapy for chordomas
ment recommendations. Neurosurgery 2009; 64 (Suppl): A44–A53 and chondrosarcomas of the skull base. J Neurosurg 1999; 91: 432–439
[23] Jiang B, Veeravagu A, Lee M, et al. Management of intracranial and extracrani- [33] Koga T, Shin M, Saito N. Treatment with high marginal dose is mandatory
al chordomas with CyberKnife stereotactic radiosurgery. J Clin Neurosci to achieve long-term control of skull base chordomas and chondrosarco-
2012; 19: 1101–1106 mas by means of stereotactic radiosurgery. J Neurooncol 2010; 98: 233–
[24] Calcerrada Díaz-Santos N, Blasco Amaro JA, Cardiel GA, Andradas Aragonés E. 238
The safety and efficacy of robotic image-guided radiosurgery system treat- [34] Martin JJ, Niranjan A, Kondziolka D, Flickinger JC, Lozanne KA, Lunsford LD.
ment for intra- and extracranial lesions: a systematic review of the literature. Radiosurgery for chordomas and chondrosarcomas of the skull base. J Neuro-
Radiother Oncol 2008; 89: 245–253 surg 2007; 107: 758–764
[25] Hasegawa T, Ishii D, Kida Y, Yoshimoto M, Koike J, Iizuka H. Gamma Knife sur- [35] Kondziolka D, Lunsford LD, Flickinger JC. The role of radiosurgery in the ma-
gery for skull base chordomas and chondrosarcomas. J Neurosurg 2007; 107: nagement of chordoma and chondrosarcoma of the cranial base. Neurosur-
752–757 gery 1991; 29: 38–45, discussion 45–46
[26] Dassoulas K, Schlesinger D, Yen CP, Sheehan J. The role of Gamma Knife [36] Hauptman JS, Barkhoudarian G, Safaee M, et al. Challenges in linear accelera-
surgery in the treatment of skull base chordomas. J Neurooncol 2009; 94: tor radiotherapy for chordomas and chondrosarcomas of the skull base: focus
243–248 on complications. Int J Radiat Oncol Biol Phys 2012; 83: 542–551
[27] Ito E, Saito K, Okada T, Nagatani T, Nagasaka T. Long-term control of clival [37] Pehlivan B, Ares C, Lomax AJ, et al. Temporal lobe toxicity analysis after pro-
chordoma with initial aggressive surgical resection and gamma knife radio- ton radiation therapy for skull base tumors. Int J Radiat Oncol Biol Phys 2012;
surgery for recurrence. Acta Neurochir (Wien) 2010; 152: 57–67, discussion 83: 1432–1440
67 [38] Santoni R, Liebsch N, Finkelstein DM, et al. Temporal lobe (TL) damage follow-
[28] Kano H, Iqbal FO, Sheehan J, et al. Stereotactic radiosurgery for chordoma: a ing surgery and high-dose photon and proton irradiation in 96 patients af-
report from the North American Gamma Knife Consortium. Neurosurgery fected by chordomas and chondrosarcomas of the base of the skull. Int J Radi-
2011; 68: 379–389 at Oncol Biol Phys 1998; 41: 59–68

127
Stereotactic Radiosurgery for Chondrosarcomas

15 Stereotactic Radiosurgery for Chondrosarcomas


Hideyuki Kano, Aditya Iyer, and L. Dade Lunsford

must be considered for residual tumors.14,15 Crockard et al


Key Points reported a 93% 5-year survival rate for 17 patients who under-
went surgery alone.16 Wanebo et al17 reported that 23 patients
● Chondrosarcomas of the skull base are relatively slow grow- with chondrosarcoma of the skull base underwent 43 surgical
ing, locally invasive tumors that usually do not metastasize resections with a mean follow-up of 97 months. Ten patients
until very late in their natural history. underwent adjuvant radiotherapy (RT). The 5- and 10-year
● Stereotactic radiosurgery for skull base chondrosarcomas is overall survival rates were 93 and 71%, respectively. Bloch et al
an important adjuvant option for these rare tumors, as part found a recurrence rate of 44% in patients who underwent sur-
of a team approach that includes initial surgical removal of gical resection alone, compared with 9% in patients who had
symptomatic larger tumors. surgery followed by RT.12 A recent review of the literature
● Stereotactic radiosurgery is associated with a high rate of lo- described the management of 560 patients with intracranial
cal tumor control and a low risk of neurologic complications. chondrosarcomas; the 5-year mortality was 11.5% and the
median survival was only 24 months. There was also no associ-
ation between the rate of recurrence and the histological grade
of the tumor.12
15.1 Introduction
Chondrosarcoma is an osteocartilaginous tumor that is usually
dural based.1 Cranial chondrosarcomas originate from primitive
15.3 Radiotherapy for
mesenchymal cells within the cartilaginous matrix of the skull Chondrosarcomas
base.2 The typical sites of origin are synchondroses of the clivus,
Reports of the use of fractionated proton beam RT have
the sphenoid-ethmoid region, the parasellar area, and the tem-
increased in the past decade. Over the past 30 years, depend-
poral-occipital junction.3,4 The differential diagnosis of chon-
able dose constraints for normal tissues at the skull base for
drosarcoma includes chordoma, chondromyxoid fibroma,
protons were studied, and doses have been escalated to
plasmacytoma, nasopharyngeal carcinoma, and other meta-
improve biological response while maintaining a reasonable
static tumors.5 The ability to distinguish chondrosarcomas from
toxicity profile.18,19,20,21 Most proton facilities rely on the use of
chordomas by imaging alone is often difficult but important
the Bragg peak effect, which facilitates energy deposition at the
because the prognosis is generally considered better for chon-
target with a sharp exit dose falloff medially, thereby reducing
drosarcomas.6,7 Chordomas have a tendency to cause brainstem
the potential for brainstem injury. Although most studies are
compression because they arise from the clivus, but chondro-
small patient series, the reported tumor progression-free sur-
sarcomas tend to affect the lower cranial nerves, as they
vival (PFS) at 5 years after surgical resection with or without
frequently originate from the occipitotemporal bone synchond-
proton RT varies from 85 to 95%.22,23,24 Advances in photon-
rosis.8 The most common presenting symptom is diplopia sec-
based radiotherapy (PBRT) such as intensity-modulated radio-
ondary to an abducens nerve palsy, closely followed by
therapy (IMRT) have facilitated improved dose conformality to
headache.9 Chondrosarcomas are relatively slow growing,
a defined target volume and increased sparing of adjacent nor-
locally invasive tumors that usually do not metastasize until
mal tissue. Combs et al25 reported on a series of patients who
very late in the natural history.10 The main subtypes of chon-
received IMRT with dose ranging from 66 to 76 Gy. They noted a
drosarcomas include a conventional/classic type that consti-
5-year PFS and survival of 100% without cranial nerve deficit or
tutes 80% of cases and clear cell, mesenchymal, and
brainstem damage.
dedifferentiated variants that constitute the remaining 20%. 11
The conventional/classic type is further divided into World
Health Organization (WHO) grade I to III tumors based on the 15.4 Stereotactic Radiosurgery for
degree of cellularity and atypia. The mesenchymal type exhibits
the most aggressive growth behavior and poorest prognosis.12 Chondrosarcomas
Stereotactic radiosurgery (SRS) has been used as a minimally
invasive primary, adjuvant, or salvage option for chondrosarco-
15.2 Surgical Resection for mas. Stereotactic radiosurgery has been shown to result in less
Chondrosarcomas toxicity to surrounding structures and have fewer complica-
tions than fractionated RT in the management of chondrosarco-
Maximal safe microsurgical resection is considered the first- mas. Relatively little data exist to define the use of SRS in the
line management for intracranial chondrosarcomas to confirm multimodality management of chondrosarcomas (▶ Table 15.1).
the histological analysis and pursue the first-line option of The tumor margin after surgical resection is often unclear on
maximal safe resection.13,14 The ability to obtain a gross total magnetic resonance imaging (MRI) because of bone destruction
resection has improved as microsurgical skull base access tech- and surrounding tissue infiltration. In such cases, the coregis-
niques have matured. However, chondrosarcomas are rarely tration of MR and computed tomography (CT) images during
completely resectable and additional management options dose planning is useful. Koga et al reported the results of four

128
Stereotactic Radiosurgery for Chondrosarcomas

Table 15.1 Studies and patient characteristics in published series of chondrosarcoma treated with stereotactic radiosurgery
Study N Radiation Median age Median Tumor Local control Survival Median fol-
margin dose volume low-up (mo)

Krishnan et al, 4 SRS 45 15 Gy 14.4 cc 5 y: 100 NA 58


200527

Hasegawa et al, 7 SRS 47 14 Gy 19.7 cc 5 y: 76% 5 y: 90% 59


200728 10 y: 67% 10 y: 53%

Cho et al, 200831 11 SRS ± RT 38 12.7 ± 58.2 Gy 3.7 cc 5 y: 89% 5 y: 100% 56


10 y: 80% 10 y: 100%

Koga et al, 201026 4 SRS 43 15.6 Gy NA 5 y: 100% NA 65

Iyer et al, 201229 22 SRS ± RT 42 15 Gy 8.0 cc 5 y: 72% 5 y: 70% 60


10 y: 54% 10 y: 56%

Abbreviations: NA, not applicable; RT, fractionated radiotherapy; SRS, stereotactic radiosurgery.

patients who had surgical resection followed by SRS at a died. The overall survival after SRS was 89% at 3 years, 86% at 5
median follow-up of 99 months.26 Three of the four patients years, and 76% at 10 years. The overall survival after SRS in
(margin doses: 15, 16, 20 Gy) had no change in tumor size dur- patients without prior RT (N = 41) was 94% at 3 years, 91% at 5
ing follow-up. One patient, who received a lower tumor margin years, and 80% at 10 years (▶ Fig. 15.1). In the group of patients
dose of 12 Gy, had tumor recurrence 100 months after SRS. without prior RT, larger tumor volume (continuous data) was
Krishnan et al27 reported 25 patients with chordomas and four significantly associated with worse overall survival (p = 0.049,
patients with chondrosarcomas who were treated by SRS. All hazard ratio [HR] = 1.07, 95% confidence interval [CI] = 1.00–
four patients with chondrosarcomas had tumor control at 5 1.14). In the group of patients without prior RT, the 5-year
years. No prognostic factors were identified in these early stud- overall survival was 100% when the tumor volume was < 5 cc
ies. Hasegawa et al28 reported 30 patients with chordomas and compared with 84% for patients whose tumor volumes ≥ 5 cc
seven patients with chondrosarcomas who underwent SRS. The (p = 0.049). Local tumor progression occurred in 10 patients.
actuarial 5-year PFS rate in patients with low-grade chondro- The PFS after SRS was 88% at 3 years, 85% at 5 years, and 70% at
sarcomas was 80%. A tumor volume of > 20 mL significantly 10 years. Prior RT was significantly associated with shorter PFS.
improved PFS. Iyer et al studied 22 patients who underwent The PFS in patients without prior RT was 92% at 3 years, 88% at
Gamma Knife SRS for residual or recurrent intracranial chon- 5 years, and 81% at 10 years (▶ Fig. 15.1). In patients without
drosarcomas.29 The overall patient survival after SRS was 95, prior RT, larger tumor volume (continuous: p = 0.035, HR =
70, and 56% at 1, 5, and 10 years, respectively. Factors associated 1.074, 95% CI = 1.005–1.147; ≥ 5 cc: p = 0.043) was significantly
with longer survival after SRS included a shorter interval (< 6 associated with shorter PFS. The PFS in patients without prior
months) between diagnosis and SRS, age > 40 years, and either RT and with tumor volume < 5 cc was 100% at 10 years, whereas
a single or no prior resection. Treated tumor control rates were the PFS in patients without prior RT and with tumor volume ≥ 5
91% at 1 year, 72% at 5 years, and 54% at 10 years. Factors asso- cc was 87% at 3 years, 81% at 5 years, and 69% at 10 years. The
ciated with longer PFS after SRS included patient age > 40 years PFS in patients with residual tumor and no prior RT (N = 33)
and no prior RT. Jiang et al30 reported 16 intracranial and spinal was 94% at 3 years, 90% at 5 years, and 82% at 10 years after
chondrosarcoma patients who underwent CyberKnife SRS at SRS. The PFS in patients with recurrent tumor and no prior RT
the median follow-up of 33 months. The mean tumor volume (N = 8) was 86% at 3, 5, and 10 years after SRS. Recurrent tumors
was 35.1 cc. The margin dose varied from 22 Gy with a single were not associated with PFS (p = 0.977). Eight patients required
fraction to 30 Gy with five fractions. The overall patient survival a salvage surgical resection and three patients (7%) developed
was 81% at 1 year, 67% at 3 years, and 55% at 5 years. The tumor adverse radiation effects. Twenty-two (56%) of 39 patients who
control was 80% in primary, 50% in recurrent, and 0% in meta- had cranial nerve deficits before SRS improved. Clinical
static tumor. Adverse radiation effects were seen in one patient. improvement after SRS was noted in patients with abducens
paralysis (61%), oculomotor paralysis (50%), lower cranial dys-
function (50%), optic neuropathy (43%), facial neuropathy (38%),
15.5 Report of the North Amer- trochlear paralysis (33%), trigeminal neuropathy (15%), and
hearing loss (10%).
ican Gamma Knife Consortium
Seven participating centers of the North American Gamma
Knife Consortium (NAGKC) identified 46 patients who under-
15.6 Summary
went Gamma Knife SRS for skull base chondrosarcomas. Thirty- Stereotactic radiosurgery provides a reasonable benefit-to-risk
six patients had prior surgical resections and six had prior frac- profile for patients with residual or newly diagnosed small-vol-
tionated RT. The median target volume was 7.8 cc (0.9–41 cc) ume skull base chrondrosarcomas. The ability to achieve tumor
and the median margin dose was 15 Gy (10.5–20 Gy). At a control of chondrosarcomas is likely enhanced by earlier recog-
median follow-up of 66 months after SRS, eight patients had nition and the application of multimodal treatment in

129
Stereotactic Radiosurgery for Chondrosarcomas

Fig. 15.1 (a) A T1-weighted contrast-enhanced axial magnetic resonance image (MRI) of a 32-year-old woman shows a chondrosarcoma at the time of
stereotactic radiosurgery (SRS). She underwent surgical resection followed by SRS for a residual tumor. Tumor volume was 1.2 cc and margin dose was
14 Gy. (b) An axial computed tomography image (CT) of the same patient at the time of SRS. (c) A T1-weighted contrast-enhanced reconstructed
coronal MRI. (d) A reconstructed coronal CT. (e) A T1-weighted contrast-enhanced reconstructed sagittal MRI. (f) A reconstructed sagittal CT.

appropriate patients. Maximal safe resection should be the pri- [7] Munzenrider JE, Liebsch NJ. Proton therapy for tumors of the skull base.
Strahlenther Onkol 1999; 175 (Suppl 2): 57–63
mary initial management of chondrosarcomas. Stereotactic
[8] Volpe NJ, Liebsch NJ, Munzenrider JE, Lessell S. Neuro-ophthalmologic find-
radiosurgery is a potent treatment option for small- to me- ings in chordoma and chondrosarcoma of the skull base. Am J Ophthalmol
dium-sized chondrosarcomas and is associated with improve- 1993; 115: 97–104
ment of cranial nerve function in selected cases, especially [9] Bloch OG, Jian BJ, Yang I, et al. Cranial chondrosarcoma and recurrence. Skull
patients who present with diplopia related to an abducens Base 2010; 20: 149–156
[10] Gay E, Sekhar LN, Rubinstein E, et al. Chordomas and chondrosarcomas of the
palsy. The improvement of neuroimaging and radiosurgical
cranial base: results and follow-up of 60 patients. Neurosurgery 1995; 36:
techniques has likely led to earlier detection of residual or 887–896, discussion 896–897
recurrent chondrosarcomas following resection and facilitated [11] Amichetti M, Amelio D, Cianchetti M, Enrici RM, Minniti G. A systematic re-
improvements in radiosurgical delivery. view of proton therapy in the treatment of chondrosarcoma of the skull base.
Neurosurg Rev 2010; 33: 155–165
[12] Bloch OG, Jian BJ, Yang I, et al. A systematic review of intracranial chondrosar-
References coma and survival. J Clin Neurosci 2009; 16: 1547–1551
[13] Lanzino G, Dumont AS, Lopes MB, Laws ER Jr. Skull base chordomas: over-
[1] Cosar M, Iplikcioglu AC, Bek S, Gokduman CA. Intracranial falcine and convex- view of disease, management options, and outcome. Neurosurg Focus 2001;
ity chondromas: two case reports. Br J Neurosurg 2005; 19: 241–243 10: E12
[2] Heffelfinger MJ, Dahlin DC, MacCarty CS, Beabout JW. Chordomas and cartila- [14] Sekhar LN, Pranatartiharan R, Chanda A, Wright DC. Chordomas and chon-
ginous tumors at the skull base. Cancer 1973; 32: 410–420 drosarcomas of the skull base: results and complications of surgical manage-
[3] Rosenberg AE, Nielsen GP, Keel SB, et al. Chondrosarcoma of the base of the ment. Neurosurg Focus 2001; 10: E2
skull: a clinicopathologic study of 200 cases with emphasis on its distinction [15] Sen CN, Sekhar LN. The subtemporal and preauricular infratemporal ap-
from chordoma. Am J Surg Pathol 1999; 23: 1370–1378 proach to intradural structures ventral to the brain stem. J Neurosurg 1990;
[4] Lee SY, Lim YC, Song MH, Seok JY, Lee WS, Choi EC. Chondrosarcoma of the 73: 345–354
head and neck. Yonsei Med J 2005; 46: 228–232 [16] Crockard HA, Cheeseman A, Steel T, et al. A multidisciplinary team approach
[5] Neff B, Sataloff RT, Storey L, Hawkshaw M, Spiegel JR. Chondrosarcoma of the to skull base chondrosarcomas. J Neurosurg 2001; 95: 184–189
skull base. Laryngoscope 2002; 112: 134–139 [17] Wanebo JE, Bristol RE, Porter RR, Coons SW, Spetzler RF. Management of cra-
[6] Goel A. Chordoma and chondrosarcoma: relationship to the internal carotid nial base chondrosarcomas. Neurosurgery 2006; 58: 249–255, discussion
artery. Acta Neurochir (Wien) 1995; 133: 30–35 249–255

130
Stereotactic Radiosurgery for Chondrosarcomas

[18] Austin-Seymour M, Munzenrider J, Goitein M, et al. Fractionated proton radi- [25] Combs SE, Laperriere N, Brada M. Clinical controversies: proton radiation
ation therapy of chordoma and low-grade chondrosarcoma of the base of the therapy for brain and skull base tumors. Semin Radiat Oncol 2013; 23: 120–
skull. J Neurosurg 1989; 70: 13–17 126
[19] Debus J, Hug EB, Liebsch NJ, et al. Brainstem tolerance to conformal radiother- [26] Koga T, Shin M, Saito N. Treatment with high marginal dose is mandatory to
apy of skull base tumors. Int J Radiat Oncol Biol Phys 1997; 39: 967–975 achieve long-term control of skull base chordomas and chondrosarcomas by
[20] Fuji H, Nakasu Y, Ishida Y, et al. Feasibility of proton beam therapy for chordo- means of stereotactic radiosurgery. J Neurooncol 2010; 98: 233–238
ma and chondrosarcoma of the skull base. Skull Base 2011; 21: 201–206 [27] Krishnan S, Foote RL, Brown PD, Pollock BE, Link MJ, Garces YI. Radiosurgery
[21] Hug EB, Slater JD. Proton radiation therapy for chordomas and chondrosarco- for cranial base chordomas and chondrosarcomas. Neurosurgery 2005; 56:
mas of the skull base. Neurosurg Clin N Am 2000; 11: 627–638 777–784, discussion 777–784
[22] Ares C, Hug EB, Lomax AJ, et al. Effectiveness and safety of spot scanning pro- [28] Hasegawa T, Ishii D, Kida Y, Yoshimoto M, Koike J, Iizuka H. Gamma Knife sur-
ton radiation therapy for chordomas and chondrosarcomas of the skull base: gery for skull base chordomas and chondrosarcomas. J Neurosurg 2007; 107:
first long-term report. Int J Radiat Oncol Biol Phys 2009; 75: 1111–1118 752–757
[23] Santoni R, Liebsch N, Finkelstein DM, et al. Temporal lobe (TL) damage follow- [29] Iyer A, Kano H, Kondziolka D, et al. Stereotactic radiosurgery for intracranial
ing surgery and high-dose photon and proton irradiation in 96 patients af- chondrosarcoma. J Neurooncol 2012; 108: 535–542
fected by chordomas and chondrosarcomas of the base of the skull. Int J Radi- [30] Jiang B, Veeravagu A, Feroze AH, et al. CyberKnife radiosurgery for the ma-
at Oncol Biol Phys 1998; 41: 59–68 nagement of skull base and spinal chondrosarcomas. J Neurooncol 2013; 114:
[24] Weber DC, Rutz HP, Pedroni ES, et al. Results of spot-scanning proton radia- 209–218
tion therapy for chordoma and chondrosarcoma of the skull base: the Paul [31] Cho YH, Kim JH, Khang SK, Lee JK, Kim CJ. Chordomas and chondrosarcomas
Scherrer Institut experience. Int J Radiat Oncol Biol Phys 2005; 63: 401–409 of the skull base: comparative analysis of clinical results in 30 patients. Neu-
rosurg Rev 2008; 31: 35–43, discussion 43

131
Stereotactic Radiosurgery for Glomus Tumors

16 Stereotactic Radiosurgery for Glomus Tumors


Ajay Niranjan, Edward A. Monaco III, Hideyuki Kano, John C. Flickinger, and L. Dade Lunsford

sometimes results in severe adverse neurologic events, ranging


Key Points from deficits in hearing, facial weakness, and symptoms of
vagal and glossopharyngeal dysfunction. Stereotactic radiosur-
● Stereotactic radiosurgery is a minimally invasive option for gery is a primary option for these patients: It minimizes the
management of glomus tumors. risks of open surgical techniques, preserves existing cranial
● It can be used as a primary management or as an adjunct nerve (CN) function in most patients, and achieves tumor
treatment for recurrent or residual glomus jugulare tumors growth arrest. Adjuvant Gamma Knife stereotactic radiosurgery
after surgical resection. (GKSRS) is used for larger tumors after their initial partial
● It is associated with a high rate of local tumor control and a resection, providing long-term tumor growth control of a sig-
low risk of neurologic complications for glomus tumors. nificantly reduced tumor volume.

16.2 University of Pittsburgh


16.1 Introduction
Experience
Glomus jugulare tumors are benign, but locally invasive tumors
that arise from the paraganglionic chemoreceptors and may Between 1987 and 2010, 20 patients (9 men and 13 women)
occur in intracranial and extracranial locations. There are two with glomus tumors underwent GKSRS at our institution.
types of paragangliomas: sympathetic and parasympathetic. Median patient age was 54 years (32–88 years). Seven patients
Most head and neck paragangliomas are derived from parasym- had SRS for residual tumor after prior resection, whereas
pathetic paraganglionic tissue that travels along the glossophar- 13 had SRS as the primary management. Histological confirma-
yngeal and vagus nerves. Head and neck glomus tumors tion was available for 8 patients; 12 were diagnosed based on
include carotid body tumors, jugulotympanic tumors, and glo- imaging characteristics. The tumors were categorized as Glass-
mus vagale. Histologically, these are benign tumors with rare cock-Jackson grade 1 (n = 5), grade 2 (n = 2), grade 3 (n = 5), and
malignant forms. Paragangliomas contain two cell types: the grade 4 (n = 8). The tumors were also categorized as Fisch stage
chief cells and the supporting sustentacular cells. The chief cells A (n = 1), stage B (n = 2), stage C (n = 4), and stage D (n = 13).
belong to the diffuse neuroendocrine system (DNES) and con-
tain neurosecretory granules. Physiologically, these are chemo-
receptors sensitive to changes in serum pH, pCO2, and pO2.
16.2.1 Stereotactic Imaging
These tumors typically occur in older patients, but can be Radiosurgery was performed using the GK (models U, B, C, 4C,
found in young adults, especially in those with familial inheri- or Perfexion; Elekta Instruments AB, Stockholm, Sweden). Gam-
tance. Glomus tumors are highly vascular; they are usually soli- ma Knife SRS is a single-day outpatient surgical procedure. It
tary. Generally, symptoms are caused by local invasion of begins with rigid fixation of an imaging- (computed tomogra-
adjacent structures or the effect of the tumor mass. The symp- phy [CT], magnetic resonance imaging [MRI]) compatible Lek-
toms of these tumors include tinnitus, hearing loss, disequili- sell stereotactic frame (model G; Elekta Instruments) to the
brium, otalgia, dysphagia, hoarseness, and shoulder drooping patient’s head. Local anesthetic scalp infiltration (5% Marcaine
related to progressive cranial symptoms as the tumors pro- and 1% Xylocaine) is supplemented by mild intravenous seda-
gresses. In rare cases, these tumors may produce the hormone tion with fentanyl and midazolam. A three-dimensional (3D)
norepinephrine, which may cause headaches, anxiety, hyper- volume acquisition MRI using a gradient pulse sequence (div-
tension, and tachycardia. Because of the highly vascular nature ided into 1- or 1.5-mm-thick 28–36 axial slices) was performed
and relatively inaccessible anatomical location of glomus jugu- to cover the entire lesion and surrounding critical structures.
lare tumors, surgical resection is often challenging. The ideal Stereotactic images were transferred via a fiberoptic Ethernet
treatment for patients with a glomus tumor remains controver- to the GammaPlan (Elekta Instruments) dose-planning com-
sial. Treatment options include surgical resection, endovascular puter where images are first checked for any distortion or inac-
embolization, fractionated radiation therapy, and stereotactic curacy. Planning was performed on thin-slice axial MRI with
radiosurgery (SRS), alone or in combination. Skull base tumors coronal and sagittal reconstructions. Computed tomography is
may extend intra- as well as extracranially, as is typical in the also valuable when bony involvement is defined in advance.
glomus jugulare variant. When located entirely in the middle Using newer-generation dose-planning systems, both MRI and
ear, glomus tympanicum tumors can be seen via otoscopic CT data can be fused to best define the tumor margins and rela-
examination. tionship to skull base bony structures (▶ Fig. 16.1).
Stereotactic radiosurgery has become an integral part of con-
ventional and advanced skull base surgery. Despite the advan-
ces in skull base techniques, the goal of total resection of such
16.2.2 Dose Planning
tumors is often problematic and associated with significant Dose planning is one of the most critical aspects of radiosur-
risk to critical structures of the skull base, including those with- gery. A 3D conformal radiosurgery plan is necessary to limit the
in the jugular bulb. Aggressive resection of such tumors radiation dose to adjacent critical structures (selectivity).

132
Stereotactic Radiosurgery for Glomus Tumors

Fig. 16.1 (a) A 67-year-old man presented with 8-month history of pulsatile tinnitus and headache. Magnetic resonance imaging (MRI) showed a
vascular mass in the region of left jugular foramen. He underwent Gamma Knife radiosurgery as primary management of this tumor. At radiosurgery
the target volume was 3.5 cc. A conformal radiosurgery dose plan was designed using T1-weighted contrast-enhanced MRI, T2-weighted MRI,
computed tomography (CT), and angiography. A margin dose of 18 Gy was prescribed to 50% isodose line. (b) One-year follow-up MRI shows
regressed tumor.

Highlights of GK meningioma radiosurgery planning include 16.2.3 Dose Prescription


accurate 3D delineation of tumor volume, use of multiple iso-
centers, beam weighting, and use of beam-blocking patterns, After optimizing the plan, a maximum dose within the target
when appropriate, to achieve selectivity. Precise 3D conformal- and minimal dose at the target edge was determined. The treat-
ity between planned isodose volume and tumor volumes is ment isodose, maximum dose, and dose to the margin were
needed to avoid radiation-related complications. This degree of jointly decided by a multidisciplinary team including a neuro-
conformality can be achieved through complex multi-isocenter surgeon, a radiation oncologist, and a medical physicist, after
planning. Glomus tumor dose planning at our center was typi- considering the goal of radiosurgery in an individual patient
cally performed using a combination of small-diameter (4- and and the estimated tolerance of the surrounding structures. A
8-mm) collimators. For large tumors, 14-, 16- (Perfexion mod- mean margin dose of 13 Gy (12–18 Gy) was prescribed to the
el), or 18-mm collimators may be used. The median target vol- tumor margins. For the last decade, we have prescribed a mar-
ume in this cohort was 12.5 cc (1.1–26.7 cc). An average of 10 gin dose of 12–15 Gy to the edge of a glomus tumor, depending
isocenters (3–14) was used to yield conformal and selective on volume, anatomical location, and CN function. This radiosur-
radiosurgery. gical dose is associated with both a low complication rate and a

133
Stereotactic Radiosurgery for Glomus Tumors

high rate of tumor control. After prescribing the margin dose, the management of glomus tumor using fractionated radiation
the falloff to surrounding critical structures (selectivity) was therapy suggested a high tumor control rate, with minimal
assessed so that adjacent critical structures received a tolerance morbidity and mortality.3 In a later study, Mukherji et al
dose. Dose delivery was accomplished in a single session by reported a tumor control rate of 70% or higher using fractio-
positioning the head serially for each subsequent isocenter until nated radiation therapy.4 The rate of complications is relatively
a fully conformal radiosurgical dose was delivered. low (0–10%), and complications include osteonecrosis of the
temporal bone, mastoiditis, and alopecia. Traditional external
beam radiation, however, is limited by the relative imprecision
16.2.4 Postoperative Care and of radiation delivery and needs a prolonged treatment course
Evaluations often requiring several weeks. Advances in delivery of fractio-
nated radiation therapy using intensity-modulated or image-
Patients received an intravenous dose of 40 mg of methylpredni- guided techniques likely will further improve tumor control
solone at the conclusion of the radiosurgical procedure. The rates and reduce adverse radiation effects.
stereotactic frame was removed and patients were observed for
1 to 2 hours prior to discharge. After radiosurgery, all patients
were evaluated with serial gadolinium-enhanced MRI scans and 16.4 Clinical Outcomes of
clinical examination. Studies were generally requested following
a schedule of 6 months, 12 months, and 2, 4, 8, and 16 years. Radiosurgery for Glomus Jugulare
Tumors
16.2.5 Results Several single-institution small retrospective studies reporting
the use of SRS for glomus tumors have established the role of
At a median follow-up of 29.5 months (range 6–112 months),
radiosurgery in the management of this rare tumor
17 patients were alive and 3 had died from unrelated causes.
(▶ Table 16.1). Liscák et al reported 52 patients with glomus
Overall survival was 85%. Imaging follow-up was available in 17
jugulare tumors treated by GKSRS.5 Twenty-four patients had
patients. Tumors were stable or regressed in 88.25% of patients.
prior surgical resection, 14 had prior embolization, and 5 had
Five patients reported improvement in tinnitus, and one noted
prior fractionated radiation therapy (RT). The median tumor
worsening.
volume was 5.7 cc (range 0.5–27 cc) and median margin dose
was 16.5 Gy (range 10–30 Gy). All patients had tumor control
16.2.6 Discussion after a median of 24 months. The neurologic symptom control
rate was 96%. Sheehan et al reported results for glomus tumor
Glomus jugulare tumors are indolent, locally destructive lesions
radiosurgery from their institution.6 Eight patients were treated
arising from the paraganglionic tissue surrounding the jugular
using 15 Gy margin dose. At a median follow-up of 28 months,
bulb. Current treatment options for glomus jugulare tumors
all seven patients with radiological follow-up showed tumor
include surgical resection, endovascular embolization, convention-
growth control. In a recent analysis from the University of Vir-
ally fractionated external beam radiation therapy, SRS, or a combi-
ginia, a margin dose of 13 Gy or greater appeared to confer a
nation of these modalities. Surgery preceded by embolization is
higher rate of tumor control with minimal morbidity in a series
often considered a mainstay of treatment, especially in young
of 15 patients with glomus tumors.7
patients. Postoperative cranial neuropathies are common and can
Although the majority of published radiosurgical series for
serve to limit functional capacity after surgery. This postoperative
glomus tumor cases have been from GK centers, sporadic
morbidity has led to an increase in the use of fractionated radio-
reports of linear accelerator (linac) radiosurgery have also been
therapy for the treatment of glomus jugulare tumors, especially in
published (▶ Table 16.1). In a 2009 report on a series of 34
elderly patients or those with very large tumors.
patients with large (defined as > 15 cm3) skull base tumors,
The goal of surgery is complete tumor removal, whereas the
including some patients harboring glomus tumors, Tuniz et al 8
goal of conventionally fractionated external beam radiation
demonstrated local tumor control in all patients and 21% clini-
therapy and SRS is long-term tumor control. Both complete
cal improvement after multisession CyberKnife treatment. In
tumor resection and long-term tumor control should prevent
another series of nine patients (eight with glomus jugulare
tumor growth and regional extension that could lead to pro-
tumors), CyberKnife treatment achieved local tumor control in
gressive symptoms, neurologic deficits, and ultimately death.
all patients.9
Irrespective of the mode or outcome of treatment, few patients
die of residual or recurrent tumor, unless there is intracranial
extension.1 Therefore, glomus jugulare treatment should focus
primarily on decreasing morbidity rather than on prolonging
16.5 Meta-analysis
survival. Ivan et al reported a meta-analysis of tumor control and mor-
bidity for patients with glomus jugulare tumors. 10 They iden-
tified 869 patients with from 46 publications. Patients who
16.3 Role of Fractionated Radia- underwent gross total resection alone had a tumor control
rate of 86% at a mean follow-up of 88 months. Patients who
tion Therapy underwent subtotal resection followed by SRS had a tumor
Fractionated radiation therapy has been used to treat residual control rate of 71% at a mean follow-up of 96 months.
tumor after resection.2 A meta-analysis of published reports on Patients who underwent SRS alone had a tumor control rate

134
Stereotactic Radiosurgery for Glomus Tumors

Table 16.1 Published series of glomus jugulare tumor radiosurgery


Study N Radiosurgery device Median margin Tumor volume (cc) Local control (%) Median follow-up
dose (Gy) (mo)

Liscák et al, 19995 66 GK 16.5 5.7 100 24

Pollock et al,
42 GK 14.9 19.7 97 44
200413

Gerosa et al,
20 GK 17.5 7 100 51
200614

Foote et al, 200215 25 GK 15 10.4 100 35

Maarouf et al,
12 Linac 15 NR 100 48
200316

Saringer et al,
13 GK 12 9 100 50
200117

Eustacchio et al,
19 GK 14 5.22 94.7 86
200218

Jordan et al, 200019 8 GK 16.3 9.81 100 35

Verma et al, 200620 17 GK 15 6.95 76 48

Wegner et al,
18 CyberKnife 20 (3 fractions) — 100 22.5
201021

Genc et al, 201014 18 SRS 15.6 5.5 94 53

Sheehan et al,
134 GK 15 5.5 92.7 50.5
201212

Abbreviations: GK, Gamma Knife; Linac, linear accelerator; SRS, stereotactic radiosurgery.

of 95% at a mean follow-up of 71 months. Patients who microsurgical resection. The absence of trigeminal nerve dys-
underwent gross total resection suffered worse rates of CN function at the time of radiosurgery and higher number of
deficits involving CNs IX–XI than those who underwent SRS isocenters were significantly associated with progression-free
alone. survival. Trigeminal nerve dysfunction in patients with glo-
Guss et al reported a meta-analysis in patients with glomus mus tumors likely suggests a larger volume. Pulsatile tinnitus
jugulare tumors who underwent SRS.11 They identified 335 improved in 49% of patients who reported it at presentation.
patients from 19 studies. Across all studies, 97% of patients New or progressive CN deficits were noted in 15% of patients;
achieved tumor control, and 95% of patients achieved clinical improvement in preexisting CN deficits was observed in 11%
control. These meta-analyses of glomus tumor cases revealed of patients. Twenty-five percent of patients with new or
that radiosurgery is associated with a high local tumor control worsened CN deficits demonstrated tumor progression on
rate > 95%. In a comparison of gross total resection, subtotal follow-up neuroimaging studies. The development of a new
resection, subtotal resection plus radiosurgery, and radiosur- or worsening CN deficit was a good predictor of delayed
gery alone, radiosurgery alone had the lowest rate of CN IX–XII tumor growth. No patient died as a result of tumor
deficits and the lowest rate of tumor progression. progression.

16.6 Multi-institutional Studies 16.7 Patient Selection and


Sheehan et al performed a multicenter retrospective analysis
under the auspices of the North American Gamma Knife Con-
Radiosurgery Considerations
sortium (NAGKC). These authors reported results of 134 Several factors are considered in determining whether radio-
GKSRS procedures performed for 132 patients with a median surgery is the optimal initial treatment. These include the
follow-up of 50.5 months.12 Prior resection was performed in patient’s age, presenting signs and symptoms, associated
51 patients and prior RT was performed in 6 patients. The comorbidities, tumor size and invasiveness, as well as growth
median tumor volume was 5.5 cc (range 0.6–58.6 cc). The rate. Patients with small- to moderate-volume size (volume < 15
median margin dose was 15 Gy (range 10–18 Gy). In this ser- cc) and typical imaging features are considered good candidates
ies, tumor control was achieved in 93% of patients. The 5-year for up-front radiosurgery. Modern radiosurgery systems such
tumor control rate was 88% after SRS. The majority of the as Leksell Gamma Knife Perfexion can treat intra- as well as
patients in this series had Fisch type C and D glomus tumors. extracranial tumor components in a single session. Most of
Such tumors are typically considered more challenging for these tumors are slow-growing neoplasms that respond well to

135
Stereotactic Radiosurgery for Glomus Tumors

radiosurgery. Current recommended radiosurgical doses (12– [5] Liscák R, Vladyka V, Wowra B, et al. Gamma Knife radiosurgery of the glomus
jugulare tumour—early multicentre experience. Acta Neurochir (Wien) 1999;
15 Gy), are associated with a high rate of local tumor control for
141: 1141–1146
patients with glomus tumors. In addition, a dose plan that con- [6] Sheehan J, Kondziolka D, Flickinger J, Lunsford LD. Gamma knife surgery for
forms to the shape of tumor and respects dose constraints of glomus jugulare tumors: an intermediate report on efficacy and safety. J Neu-
adjacent neural and vascular structures at risk is necessary. rosurg 2005; 102 (Suppl): 241–246
After radiosurgery, it is important for patients with glomus [7] Chen PG, Nguyen JH, Payne SC, Sheehan JP, Hashisaki GT. Treatment of glo-
mus jugulare tumors with gamma knife radiosurgery. Laryngoscope 2010;
tumors to undergo serial long-term neuroimaging clinical
120: 1856–1862
evaluations. [8] Tuniz F, Soltys SG, Choi CY, et al. Multisession cyberknife stereotactic radio-
surgery of large, benign cranial base tumors: preliminary study. Neurosur-
gery 2009; 65: 898–907, discussion 907
16.8 Summary [9] Bianchi LC, Marchetti M, Brait L, et al. Paragangliomas of head and neck: a
treatment option with CyberKnife radiosurgery. Neurol Sci 2009; 30:
Glomus jugulare tumors are rare, highly vascular tumors that 479–485
arise from the paraganglionic structures of the glossopharyng- [10] Ivan ME, Sughrue ME, Clark AJ, et al. A meta-analysis of tumor control rates
and treatment-related morbidity for patients with glomus jugulare tumors. J
eal and vagal nerves. Because of the highly vascular nature and
Neurosurg 2011; 114: 1299–1305
surgically formidable anatomical location of glomus tumors, [11] Guss ZD, Batra S, Limb CJ, et al. Radiosurgery of glomus jugulare tumors: a
curative resection often proves challenging. Treatment options meta-analysis. Int J Radiat Oncol Biol Phys 2011; 81: e497–e502
include surgical resection, endovascular embolization, fractio- [12] Sheehan JP, Tanaka S, Link MJ, et al. Gamma Knife surgery for the manage-
ment of glomus tumors: a multicenter study. J Neurosurg 2012; 117:
nated radiation therapy, and SRS, alone or in combination.
246–254
Although radiation therapy confers a reasonable rate of tumor
[13] Pollock BE. Stereotactic radiosurgery in patients with glomus jugulare tu-
control, radiosurgery appears to offer at least a comparable rate, mors. Neurosurg Focus 2004; 17: E10
but without the risks inherent with fractionated approaches. [14] Gerosa M, Visca A, Rizzo P, Foroni R, Nicolato A, Bricolo A. Glomus jugulare
Stereotactic radiosurgery can be used as an up-front treatment tumors: the option of gamma knife radiosurgery. Neurosurgery 2006; 59:
561–569, discussion 561–569
or as an adjuvant therapy for patients with recurrent or residual
[15] Foote RL, Pollock BE, Gorman DA, et al. Glomus jugulare tumor: tumor control
glomus jugulare tumor after surgical resection. Stereotactic and complications after stereotactic radiosurgery. Head Neck 2002; 24: 332–
radiosurgery is associated with a high rate of local tumor con- 338, discussion 338–339
trol and a low risk of neurologic complications for patients with [16] Maarouf M, Voges J, Landwehr P, et al. Stereotactic linear accelerater-based
glomus jugulare tumor. radiosurgery for the treatment of patients with glomus jugulare tumors. Can-
cer 2003; 97: 1093–1098
[17] Saringer W, Khayal H, Ertl A, Schoeggl A, Kitz K. Efficiency of gamma knife
radiosurgery in the treatment of glomus jugulare tumors. Minim Invasive
References Neurosurg 2001; 44: 141–146
[18] Eustacchio S, Trummer M, Unger F, Schröttner O, Sutter B, Pendl G. The role of
[1] van der Mey AG, Frijns JH, Cornelisse CJ, et al. Does intervention improve the
Gamma Knife radiosurgery in the management of glomus jugular tumours.
natural course of glomus tumors? A series of 108 patients seen in a 32-year
Acta Neurochir Suppl (Wien) 2002; 84: 91–97
period. Ann Otol Rhinol Laryngol 1992; 101: 635–642
[19] Jordan JA, Roland PS, McManus C, Weiner RL, Giller CA. Stereotastic radiosur-
[2] Williams HL, Childs DS Jr, Parkhill EM, Pugh DG. Chemodectomas of the glo-
gery for glomus jugulare tumors. Laryngoscope 2000; 110: 35–38
mus jugulare (nonchromaffin paragangliomas) with especial reference to their
[20] Varma A, Nathoo N, Neyman G, et al. Gamma knife radiosurgery for glomus
response to roentgen therapy. Ann Otol Rhinol Laryngol 1955; 64: 546–566
jugulare tumors: volumetric analysis in 17 patients. Neurosurgery 2006; 59:
[3] Springate SC, Weichselbaum RR. Radiation or surgery for chemodectoma of
1030–1036, discussion 1036
the temporal bone: a review of local control and complications. Head Neck
[21] Wegner RE, Rodriguez KD, Heron DE, Hirsch BE, Ferris RL, Burton SA. Linac-
1990; 12: 303–307
based stereotactic body radiation therapy for treatment of glomus jugulare
[4] Mukherji SK, Kasper ME, Tart RP, Mancuso AA. Irradiated paragangliomas of
tumors. Radiother Oncol 2010; 97: 395–398
the head and neck: CT and MR appearance. AJNR Am J Neuroradiol 1994; 15:
357–363

136
Stereotactic Radiosurgery for Nonvestibular Schwannomas

17 Stereotactic Radiosurgery for Nonvestibular


Schwannomas
Edward A. Monaco III, Gurpreet Surinder Gandhoke, Ajay Niranjan, Hideyuki Kano, and L. Dade Lunsford

most affected.2 It is believed that the growth patterns and bio-


Key Points logical behavior of CN schwannomas are similar regardless of
the nerve of origin.3,4,5,6 Nedzelski and Tator coined the terms
● Nonvestibular schwannomas are relatively rare intracranial “nonacoustic neuroma” and “nonvestibular schwannoma” in
tumors that can arise from any cranial nerve and are thought 1982 in a review of eight cases involving patients harboring
to show similar growth patterns and biological behavior as such tumors.7 Nonvestibular cranial nerve schwannomas
vestibular schwannomas. account for less than 10% of intracranial schwannomas and less
● In a fashion similar to that for vestibular schwannomas, treat- than 0.5% of all intracranial neoplasms.5,8 Symptoms from these
ment of nonvestibular schwannomas has evolved with the tumors are typically referable to dysfunction of the nerve from
advent of modern microsurgical skull base approaches and which the tumor arises, but can also be related to mass effect
stereotactic radiosurgery. on surrounding structures or even nonspecific such as head-
● The results after stereotactic radiosurgery for nonvestibular aches. The most common presenting complaint of patients
schwannomas compare favorably with those after surgery, found to have trigeminal schwannomas is facial sensory dis-
while avoiding surgically related complications. turbance or pain.9
● The paradigm shift in management of skull base tumors Prior to the advent or stereotactic radiosurgery (SRS), sur-
extends to nonvestibular schwannoma treatment, with ster- gical resection was the mainstay of treatment of NVCNS.
eotactic radiosurgery becoming the dominant modality and Complete resection of NVCNS, like that for vestibular
surgical resection being reserved for large tumors or those schwannomas, can be curative. Advancements in microsur-
causing symptoms from mass effect. gical techniques have contributed to improved outcomes for
patients harboring such tumors, but the morbidity associ-
ated with surgical resection is not trivial, especially for
tumors arising from the lower CNs. 10,11,12 Stereotactic radio-
17.1 Introduction surgery has an established role in adjuvant and primary
Cranial nerve (CN) schwannomas are typically benign tumors management of skull base tumors and has the benefit of
arising from the Schwann cells making up the nerve’s myelin avoiding surgery-related morbidity. 3,5,8,13–18 The first pub-
sheath. Cranial nerve VIII is the most frequent site of origin for lished radiosurgical series was reported by Pollock et al in
these tumors. However, they can arise from any of the other 1993, and the work described the results of Gamma Knife
CNs, with the trigeminal nerve being the second-most common (GK) SRS in a small number of patients with trigeminal or
site of origin (▶ Fig. 17.1). In a series of 62 patients with nonves- jugular foramen schwannomas. 16 Regardless of the treat-
tibular cranial nerve schwannomas (NVCNS), Safavi-Abbasi et al ment modality, the goals of management for these tumors
reported that 76% of these tumors originated from either the should include excellent tumor control, improvement or
trigeminal nerve or the jugular foramen.1 In patients with type preservation of neurologic function, and limitation of treat-
2 neurofibromatosis, the oculomotor and trigeminal nerves are ment-related morbidity.

Fig. 17.1 (a) Axial, (b) sagittal, and (c) coronal contrasted T1-weighted magnetic resonance images of a 19-year-old woman who presented with
complaints of headache, nausea, and vomiting. Physical examination identified hypoesthesia of the right face. Imaging revealed a large dumbbell-
shaped tumor involving Meckel’s cave and the posterior fossa consistent with a trigeminal schwannoma. The patient went on to receive multimodality
therapy including surgical resection, followed by stereotactic radiosurgery. Pathology revealed a benign schwannoma.

137
Stereotactic Radiosurgery for Nonvestibular Schwannomas

Headache, the third-most common presenting symptom,


17.2 Treatment Options resolved in 93.3% of affected patients postoperatively.
17.2.1 Microsurgery Microsurgical resection of NVCNS utilizing the operative
microscope and modern skull base approaches has greatly
The rationale for surgical resection of NVCNS is that it can be improved the effectiveness of surgery. When performed by
curative. Resection is valuable in instances where there is diag- experienced surgeons, in carefully selected patients, micro-
nostic uncertainty, mass effect from the tumor with associated surgery can be curative and result in meaningful improve-
symptoms, or when the tumor has a large cystic component ment in symptoms. It is not surprising that all evidence is
that may be unresponsive to radiosurgery. Resection can also class 3, given the extremely limited numbers of patients pos-
be undertaken in patients who show tumor progression after sessing these tumors. Results of microsurgical management
radiosurgery. For patients with larger tumors in whom the risk of schwannomas arising from the other CNs must be extrapo-
of complete tumor removal without significant morbidity is lated. ▶ Table 17.1 shows results of select recent surgical
judged to be high, a staged approach where surgical resection is series for NVCNS.
combined with another treatment modality, such as radiosur-
gery, can be undertaken.1
The published experience with microneurosurgery for
17.2.2 Stereotactic Radiosurgery
NVCNS is meaningfully smaller than that for vestibular schwan- Stereotactic radiosurgery has become an established primary
nomas, in line with the epidemiology of these tumors. The most and adjuvant treatment modality for skull base neoplasms
data involve patients harboring trigeminal schwannomas. Sur- (▶ Fig. 17.2). Stereotactic radiosurgery has been reported to
gical resection of all types of skull base lesions has evolved dra- achieve tumor control for NVCNS at rates similar to those of the
matically over the last several decades, owing to the best surgical series.8 One of the earliest series on SRS manage-
development of modern skull base approaches. The same is true ment of NVCNS was by Pollock et al.16 In this series, six patients
for NVCNS. Prior to modern techniques, total resection of with trigeminal schwannomas and five with jugular foramen
NVCNS occurred, at best, only 50% of the time.19 Contrast those schwannomas underwent GKSRS. Doses ranged from 12 to
results with those of Samii et al.20 In a 27-patient retrospective 20 Gy. The tumor control rate for the trigeminal tumors was
series involving trigeminal schwannomas, the tumor recur- 100% at a median follow-up of 21 months and 75% for the jugu-
rence rate was only 7.4%, with recurrent tumors having lar foramen tumors at a mean follow-up of 10 months. Two
involved the cavernous sinus. Details are scant in this study patients showed improvement in CN symptoms. This early
about the effects of surgery on patients’ symptoms. Day and experience paralleled results for vestibular schwannomas; since
Fukushima, in their report on 37 patients, achieved gross total then, numerous publications have appeared in the literature
resection in 73% of patients with trigeminal schwannomas. 21 describing the results of SRS for NVCNS.
Symptomatically, the number of patients with facial pain was Kano et al reported on 33 consecutive patients with trigemi-
reduced from 13 to 2 after surgery. Hypesthesia was rela- nal schwannomas treated by GK.14 A third of patients had pre-
tively unaffected by surgery in this cohort, as 29 of 32 viously undergone microsurgical resection. A median margin
patients had persistent hypesthesia after surgery. Sharma et dose of 15 Gy was administered to a median target volume of
al reported a total tumor excision rate for trigeminal schwan- 4.2 cc. The rates of progression-free survival in this cohort at 1,
nomas of 76% with a mean follow-up period of over 5 years. 22 5, and 10 years after SRS were 97.0, 82.0, and 82.0%, respec-
Zhang et al surgically treated 42 patients harboring trigemi- tively (▶ Fig. 17.3). Over 90% of patients had stable or improved
nal schwannomas and observed an 81% total resection rate neurologic signs and symptoms following SRS. Factors associ-
when the tumor did not involve the cavernous sinus, and ated with improved tumor control included smaller tumor size
only a 40% rate when it did. 23 Fukaya et al reported a retro- and root or ganglion-type tumors. Not unexpectedly, patients
spective surgical series of 57 patients with trigeminal who had primary SRS management also had improved out-
schwannomas.9 This study is notable, as it included patients comes when compared with patients who had previously
treated by conventional approaches and those who under- undergone craniotomy. Hasegawa et al administered a mean
went skull base approaches. In this cohort, only 38% of the dose of 14.2 Gy to a mean tumor volume of 10 cc in 42 patients
patients treated before 1990 with conventional approaches harboring trigeminal schwannomas.25 Actuarial 5- and 10-year
achieved complete tumor removal. Using skull base tumor control rates were both 84.0%. Forty percent of patients
approaches, 90% of the patients treated after 1990 had com- showed improvement in clinical symptoms. Larger tumors in
plete tumor removal. During the mean follow-up period of this series were more likely to progress despite SRS treatment.
4.9 years, these authors reported no tumor recurrences. Pan et al evaluated 56 patients treated with GK for trigeminal
Wanibuchi et al describe their larger surgical experience in schwannomas, 42 of whom had primary SRS management. 4
105 patients with trigeminal schwannomas. 24 Total removal The mean target volume was 8.7 cc and a mean tumor margin
was achieved in 75.2% of patients. The most common preope- dose of 13.3 Gy was utilized. After a mean follow-up period of
rative symptom in this series was facial hypesthesia, which 68 months, the tumor growth control rate was 93.0%. Neuro-
was only improved in 15.9% of patients. Facial pain, the sec- logic deficits improved in 44.6% of patients. Phi et al reviewed a
ond-most common presenting symptoms in the cohort, sub- cohort of 22 consecutive patients with trigeminal schwanno-
sided in 91.7% of patients. Trigeminal motor dysfunction mas.26 Having administered a mean marginal dose of 13.3 Gy to
improved in only one of three patients. Diplopia related to a median volume of 4.1 cc, they observed tumor growth control
abducens nerve dysfunction resolved in 13 of 18 patients, but in 95% of patients after a median follow-up of 46 months.
only in 1 of 3 patients with oculomotor nerve dysfunction. Two thirds of patients presenting with facial pain showed

138
Stereotactic Radiosurgery for Nonvestibular Schwannomas

Table 17.1 Recent selected microsurgical series


Study N Cranial Tumor Follow-up Tumor Improvement in Complications (%) Tumor recur- Mortality
nerve volume (mo) control clinical signs and rence rate (%) (%)
(cc) rate (%) symptoms (%)

Sedney et al, 81 JF NR NR 91 NR New cranial neuro- 8.9 0


201342 GTR, 54 pathy, 4.5
NTR, 23 Worsened cranial
STR, 1 neuropathy, 22.6
CSF leak, 4.5
ICH, 3

McRackan et al, 53 VII 27.9 NR 100 NR HB IV or worse, 51.8 0 0


201243 GTR, 45
STR, 8

Fukaya et al, 57 V NR 58.8 98 NR New cranial neuro- 0 0


20109 GTR, 43 pathy, 58.5
NTR, 3 CSF leak, 4.2
PR, 1

Safavi-Abbasi et 43 V, 24 14.8 44.5 97 Improved KPS New cranial neuro- 3.2 0


al, 20101 JF, 7 pathy, 18.4
VII and XII, CSF leak, 15.8
3 each Hydrocephalus, 2.6
IV and X, 2 Meningitis, 2.6
each
III and VI,
1 each

Fukuda et al, 15 JF NR 84.3 70 NR IX, X deficit, 40 60 0


200946 NTR, 10
STR, 5

Bulsara et al, 53 JF NR 100.8 94.3 NR Worsened IX/X neu- 5.7 0


200839 CTR, 48 ropathy, 30.0
Other, 4 CSF leak, 5.7

McMonagle et 33 VII NR NR NR NR VII nerve sacrificed, 9.1 0


al, 200844 CTR, 21 63
STR, 4 CSF leak, 2.8
Other, XII deficit, 2.8
11 Headache, 2.8
Intractable vertigo,
2.8

Sanna et al, 22 JF 36.6 NR 100 NR Lower cranial nerve 0 0


200641 CTR, 21 deficit, 50
STR, 1 CSF leak, 13.5
Meningitis, 4.5

Abbreviations: CTR, complete tumor removal; GTR, gross total resection; HB, House-Brackman; ICH, intracranial hemorrhage; JF, jugular foramen; KPS,
Karnofsky Performance Score; NTR, near-total resection; NR, not reported; PR, partial resection; STR, subtotal resection.

meaningful improvement in this symptom. A third of patients neuropathies were observed in preoperatively intact nerves.
with facial hypesthesia also improved. Peker et al retrospectively identified 17 patients who had been
Results after SRS for jugular foramen tumors have been nota- treated with GKSRS for jugular foramen schwannomas.28 They
bly similar (▶ Fig. 17.4). Martin et al reported the results of GK administered a mean marginal dose of 13 Gy to a mean tumor
treatment for 35 tumors in 34 patients.27 The median margin volume of 5.9 cc. After a mean follow-up period of 64 months,
tumor dose was 14 Gy to a median tumor volume of 4.2 cc. The they observed a tumor control rate of 100%. Neurologic symp-
reported actuarial tumor control rates were 97 ± 2.7% at 5 years toms improved in 35.3% of patients and no patients died during
and 94 ± 5.9% at 10 years. Over 48% of tumors showed radio- follow-up. Zhang et al treated 27 patients with jugular foramen
graphic reduction in size, although a small number of patients tumors using the GK.29 Nearly 45% of these patients had under-
(four) experienced a brief period of slight tumor expansion with gone previous microsurgery. The mean tumor volume was 13.5
associated loss of central contrast enhancement. All four of cc and the mean margin dose was 14.6 Gy. Ninety-six percent of
these patients eventually demonstrated tumor regression. patients who were followed over a mean of 38.6 months
Improvement in CN function occurred for 20% of affected showed shrinkage or stability of their tumors. Neurologic
nerves, whereas 77% remained stable. No new cranial improvement was noted in 64% of patients.

139
Stereotactic Radiosurgery for Nonvestibular Schwannomas

Fig. 17.2 GammaPlan (Elekta Instruments AB, Stockholm, Sweden) poster shows a conformal radiosurgery dose plan for a 20-year-old woman with
residual trigeminal schwannoma left after a two-stage surgical resection. Axial, sagittal, and coronal contrast-enhanced magnetic resonance images
were used for planning. A margin dose of 12.5 Gy was prescribed to the 50% isodose line.

Fig. 17.3 Axial contrast-enhanced magnetic res-


onance images of a young woman treated by
stereotactic radiosurgery for residual trigeminal
schwannoma after surgery, at the time of (a) her
Gamma Knife procedure in 2009, and (b) 4 years
later at follow-up. In response to radiosurgery,
the residual area of tumor has decreased in size.

Data on schwannomas arising from the remaining CNs are three hypoglossal schwannomas, two abducens tumors, and
quite limited (▶ Fig. 17.5). For example, in a series combining one each from the facial nerve, trochlear nerve, oculomotor
all types of NVCNS, Elsharkawy et al included treatment of nerve, and glossopharyngeal nerve.30 Two- and 5-year

140
Stereotactic Radiosurgery for Nonvestibular Schwannomas

Fig. 17.4 Axial contrast-enhanced magnetic res-


onance images of a 54-year-old woman who
initially presented with hoarseness and hearing
loss. She initially underwent a craniotomy that
resulted in an incomplete resection of a jugular
foramen schwannoma. The tumor rapidly grew
during subsequent surveillance and she was
referred for a Gamma Knife procedure. (a) A
stereotactic image obtained on the day of
radiosurgery showing the residual tumor. (b) Four
years later the tumor has responded to radio-
surgery with a dramatic decrease in size.

Fig. 17.5 Axial contrast-enhanced magnetic res-


onance images of a 51-year-old woman who
presented with vertical diplopia. The patient
initially elected for observation, and the tumor
grew over the course of several years. She elected
to undergo a Gamma Knife procedure. (a) A
stereotactic image obtained on the day of
radiosurgery revealing a tumor at the edge of the
tentorium on the left side consistent with a
trochlear schwannoma. (b) At follow-up 4 years
later, the tumor has meaningfully reduced in size
in response to radiosurgery.

actuarial progression-free survival rates were 91 and 78% for Taken together, the abundance of evidence suggests that SRS
the entire cohort, which constituted 69% trigeminal schwan- is an effective primary and adjunctive treatment for NVCNS.
nomas. The results were not stratified on the basis of tumor The largest experiences are with trigeminal and jugular fora-
location. Not surprisingly, higher maximum tumor dose was men tumors, but it is likely that the results can be extrapolated
significantly associated with better tumor control. Safavi- to tumor arising from other nerves. A meaningful proportion of
Abbasi et al reviewed a combined series of microsurgical and patients show improvement in their symptoms following SRS.
radiosurgical patients with NVCNS from multiple nerves. 1 In As expected, features such as smaller tumor volume and higher
this series of 62 tumors, only 24.2% of tumors arose from marginal doses are associated with improved tumor control. All
areas other than the trigeminal nerve or jugular foramen. available evidence is class 3. ▶ Table 17.2 provides an overview
Only four tumors outside of the trigeminal nerve and jugular of selected recent studies in which SRS was used to treat
foramen were treated by radiosurgery. There are limited NVCNS.
reports on facial schwannomas treated by radiosurgery. Litre
et al reported a series of 11 patients with facial schwanno-
17.2.3 Fractionated Radiotherapy
mas treated with GK.31 In 10 of the 11 patients, tumors
shrank or remained stable during a median follow-up of 39 Although the rates of new cranial neuropathy after single-ses-
months. Two patients had resolution of hemifacial spasm sion SRS compare very favorably with those observed after
after treatment, four had improvement in facial nerve func- open resection, some have argued that conventional fractio-
tion, and one patient had improvement in vestibular and nated radiotherapy or fractionated SRS, by sparing normal tis-
hearing function. Kida et al reported on 14 patients who sues and allowing for repair of sublethal damage between
received SRS for facial schwannomas. 13 The mean tumor vol- fractions, may improve upon this risk to benefit ratio. Wallner
ume was 5.5 cc and the mean tumor margin dose was et al in an early report identified eight patients treated with
12.9 Gy. The tumor control rate after a mean follow-up of conventional fractionated therapy to NVCNS with doses of 45 to
31.4 months was 100%. Facial nerve function improved in five 54 Gy.33 Half of these patients failed this conventional therapy.
of the cases. Finally, Madhok et al reported results in six These authors could make no firm conclusions on the utility of
patients with facial schwannomas. 32 Tumor control after a fractionated therapy in this setting. Zabel et al performed frac-
median follow-up of 61.5 months was 100% and facial nerve tionated stereotactic radiotherapy using a median dose of
function was preserved at preradiosurgery status in all 57.6 Gy, with 1.8 Gy per fraction in 13 patients with NVCNS. 34
patients. Local control was 100% after a median follow-up of 33 months.

141
Stereotactic Radiosurgery for Nonvestibular Schwannomas

Table 17.2 Overview of recent selected radiosurgical series


Study N Cranial Tumor Follow- up Tumor Improvement in Complications (%) Marginal Mortality
nerve volume (mo) control clinical signs and dose (Gy) (%)
(cc) rate (%) symptoms (%)

Elsharkawy et al, 36 III, 1 2.9 54 91, 78 (2, 64 Facial paresthesia, 2 13.5 0


201230 IV, 1 5 y) Slurred speech,
V, 25 tongue deviation,
VI, 2 and drooling, 1
VII, 6
IX, 1
XII, 3
JF, 2

Peker et al, 17 JF 5.9 64 100 35 Transient hoarse- 13 0


201228 ness, 5.9

Yianni et al, 74 V 5.3 48.2 93, 79 (5, 14.9 Facial paresthesia 16.4 0
201247 10 y) and numbness, 8.6
Diplopia, 1.3

Kimball et al, 49 III, 2 5.3 37 97, 83 (1, 39 Facial palsy, 2 12.5 0


201148 V, 25 5 y) Anesthesia dolorosa,
VII, 2 2
XII, 1 Facial numbness, 4.1
JF, 18 Diplopia, 2
RS, 1

Kano et al, 33 V 4.2 72 82 33.3 Facial pain, 3 15 0


200914 (5, 10 y) Facial sensory loss, 3

Hasegawa et al, 37 V 10.3 54 84 40 Facial numbness, 8.1 14.2 0


200725 (5, 10 y) Facial pain, 8.1
Corneal ulcer, 2.7

Kida et al, 200713 14 VII 5.5 31.4 100 42 HB III facial palsy, 7.1 12.9 0

Martin et al, 34 JF 4.2 83 97, 94 (5, 20 Worsened lower CN 14 0


200727 10 y) function, 2

Litre et al, 200731 11 VII 0.9 39 91 38 None 13 0

Abbreviations: CN, cranial nerve; HB, House-Brackman; JF, jugular foramen.

Four patients experienced improvement in their presenting No prospective randomized trials comparing single versus
symptoms. Showalter et al compared 39 patients harboring multisession SRS or fractionated stereotactic radiotherapy for
NVCNS treated by either single-session SRS or multisession NVCNS have been conducted, nor are they likely to be con-
stereotactic radiotherapy.35 The single-session group was ducted. To date, there does not appear to be a meaningful
treated to a median marginal dose of 12 Gy, whereas the frac- improvement in the risk-to-benefit ratio by performing fractio-
tionated cohort received a median dose of 50.4 Gy in 1.8- to nated SRS or stereotactic radiotherapy. The inconvenience of a
2.0-Gy fractions. They observed no statistically significant dif- protracted treatment course, sometimes requiring 25 to 30 dai-
ference in tumor control or CN function. Nishioka et al ly radiation sessions, and the limited repeatability of conven-
reviewed their experience with NVCNS and fractionated stereo- tional techniques makes a fractionated treatment option less
tactic radiotherapy.36 Almost 60% of patients were treated pri- preferred to single-session approaches.
marily. Doses ranged from 40 to 54 Gy in 20 to 26 fractions.
Sixteen of 17 patients experienced stable or regressed tumors
over the median follow-up of 59.5 months. Forty-seven percent
of patients had improvement in their presenting symptoms. No
17.3 Complications
patient experienced worsened or new CN deficits. Choi et al
17.3.1 Microsurgery
report treating 42 NVCNS located adjacent to the optic appara-
tus, brainstem, and other CNs with multi- and single-session Not only has the efficacy of microsurgery improved with the
linear accelerator (linac)-based SRS (one to three sessions). 37 advent of modern skull base techniques, so has the safety. One-
Although the patients treated with single-session SRS had two year mortality following attempted surgical resection of NVCNS
new cranial neuropathies compared with none in the multises- prior to the advent of modern microsurgery and skull base
sion cohort, this difference was not statistically different. Tumor approaches was 41% as reported by Schisano and Olivecrona in
control was equivalent. 1956.38 In a selected group of patients, Samii et al reduced this

142
Stereotactic Radiosurgery for Nonvestibular Schwannomas

number to 0% with the use of modern techniques to resect tri- Hydrocephalus,1,21 quadriparesis, sinus thrombosis requiring
geminal schwannomas.20 In the hands of very experienced skull anticoagulation,20,42 meningitis,1 and intracranial hemorrhage42
base surgeons and for selected patients, greatly improved rates have also been reported.
of mortality have become the norm.1,9,21,24
Cranial neuropathy is one of the primary complications
related to microsurgical resection of NVCNS. Day and Fukushi- 17.3.2 Stereotactic Radiosurgery
ma, in their report on trigeminal schwannomas, observed a
Stereotactic radiosurgery is a minimally invasive approach that
recurrence rate of 3%, and the development of new trigeminal
eliminates certain surgical complications such as CSF leaks and
deficits in 11 patients.21 Specifically, one patient developed
sinus thromboses. However, SRS is not without complications.
new facial pain, six developed facial hypesthesia, another five
Minor transient complications such as headache and nausea
facial anesthesia, and 20 experienced new trigeminal motor
have been reported in NVCNS treatment. 1 But as with microsur-
deficits. In the trigeminal schwannoma series by Samii et al,
gery, the most common concern following SRS is the develop-
75% of patients had new or worsened trigeminal, facial, or coch-
ment of new or worsened cranial neuropathy. Kano et al found
lear nerve function postoperatively.20 Wanibuchi et al surgically
that 2 of 33 patients treated with GK for trigeminal schwanno-
treated 105 patients and observed worsened or new facial hyp-
mas developed new trigeminal deficits, although both patients
esthesia in 42.1% and new facial pain in 2.5% of patients. 24 Trige-
also demonstrated tumor progression.14 Adverse radiation
minal motor weakness was observed in 3.9% of patients. One
effects (AREs) in the form of increased peritumoral T2 signal
patient suffered worsened oculomotor diplopia. Fukaya et al
change were only noted in 6.1% of patients, but these were
noted an 18.8% rate in transient and a 58.5% rate of permanent
asymptomatic. Pan et al noted a 9% rate of slightly worsened CN
cranial nerve palsies after microsurgery for trigeminal
symptoms after GKSRS for trigeminal tumors. 4 Hasegawa et al
schwannomas.9
observed a 14% rate of new or worsened symptoms with SRS to
Neurologic deficits involving the lower CNs are of particular
trigeminal schwannomas, including facial numbness, facial
concern because they can lead to breathing difficulty, aspiration
pain, corneal ulcer, and abducent palsy.25 Sheehan et al
pneumonias, and dysphagia, all of which can be life threatening.
reported that 12% of patients suffered worsening facial pain
Bulsara et al observed no new lower CN deficits in 53 patients
after trigeminal tumor treatment and ultimately required surgi-
after microsurgical resection of jugular foramen tumors, but
cal resection (n = 2) or repeat SRS (n = 1).45 No patients devel-
did report frequent worsening.39 For example, there was a 26%
oped symptoms related to adverse radiation effects. In the
rate of permanent glossopharyngeal and vagal dysfunction
report by Phi et al, 27% of patients experienced new or wors-
postoperatively. Samii et al observed a nearly 40% rate of CN
ened cranial neuropathies after treatment of trigeminal
dysfunction following resection.40 Sanna et al reported a 95%
schwannomas.26 However, only half of the cranial neuropathies
rate of CN morbidity in 23 resected patients.41 Specifically, they
were permanent. Interestingly, loss of central contrast enhance-
noted that 50% of patients suffered worsened CN IX and X func-
ment, tumor expansion, and tumor location in the cavernous
tion postoperatively. Sedney et al reported that a more conser-
sinus were factors significantly related to the development of
vative surgical approach with subtotal resection decreased, but
these cranial neuropathies.
did not eliminate, postoperative CN IX and X deficits. 42 Using
Martin et al reported on their results treating jugular fora-
the more conservative surgical approach, the number of perma-
men tumors using GKSRS and noted only one patient (2%)
nently lower CN deficits decreased nearly sevenfold, a finding
experienced CN dysfunction.27 In the 17 patients treated for
that was statistically significant. Despite this, gastrostomy tube
jugular foramen tumors by Peker et al, only one (5.8%) devel-
placement was required in 5 of 28 patients.
oped increased hoarseness in the setting of slight tumor expan-
Facial nerve dysfunction can be extremely disabling and can
sion with loss of central contrast enhancement. 28 Hasegawa et
meaningfully diminish a patient’s quality of life. There is nota-
al reported on the treatment of jugular foramen schwannomas
ble facial nerve morbidity with resection of facial nerve
with SRS in a series where no patient developed lower CN defi-
schwannomas. McRackan et al sacrificed the facial nerve in 36
cits after SRS.15
of 53 patients at the time of microsurgical resection. 43 At one
In the treatment of facial nerve schwannomas, Kida et al
year, only seven patients possessed House-Brackmann (HB)
reported that one of 14 patients treated developed persistent
grade 1 function. The rate of HB grade 4 function was 14.3%
facial palsy of HB grade 218, whereas Litre et al did not observe
preoperatively and 51.8% postoperatively. McMonagle et al also
new or worsened CN deficit in their 11 treated patients. 31 Mad-
reported requiring 21 facial reconstructions after resection in
hok et al also did not detect new cranial neuropathies or
33 patients with facial schwannomas.44 Fifty-five percent of
changes in hearing after SRS for facial schwannomas.32 These
patients had total removal of their tumors with loss of the facial
series, though small, confirm the experience with vestibular
nerve. Only one patient had complete resection with preserva-
schwannomas in that facial nerve injury is meaningfully less
tion of the nerve. Notably, they report that facial outcomes were
common with SRS than with microsurgical resection.
better in patients in whom “no removal, decompression or sub-
total removal was undertaken.” Thus, these authors concluded
that surgical intervention for schwannomas should occur after
facial nerve function deteriorates to at least a HB grade 4.
17.4 Summary
Other noteworthy complications have been observed follow- Nonvestibular cranial nerve schwannomas are rare skull base
ing microsurgery. Spinal fluid leaks occur at variable rates. lesions and can arise from any of the CNs. They most frequently
Samii et al noted cerebrospinal fluid (CSF) leaks in 16.7% arise from the trigeminal nerve and the jugular foramen. Evolu-
of patients after trigeminal schwannoma resection.20 tion in the management of NVCNS has paralleled that of the

143
Stereotactic Radiosurgery for Nonvestibular Schwannomas

Fig. 17.6 A simplified algorithm for the ma-


nagement of nonvestibular schwannomas is
shown. At the time of initial diagnosis, an
asymptomatic lesion could either be observed or
treated by stereotactic radiosurgery (SRS). In
younger individuals there is a meaningful like-
lihood of growth; thus, SRS may be elected. If
during observation a tumor grows, SRS is the
preferred approach, especially in the setting of a
continued lack of symptoms. For symptomatic
tumors without mass effect, our group recom-
mends SRS due to its minimal invasiveness,
safety, and efficacy. It is the primary treatment
modality in individuals who are not candidates for
surgery. In individuals with tumors producing
symptomatic mass effect, microsurgery is the
preferred approach. In the event of residual
tumor or tumor recurrence after microsurgery,
SRS is the recommended treatment.

much more common vestibular schwannoma. Surgical resec- [8] Mabanta SR, Buatti JM, Friedman WA, Meeks SL, Mendenhall WM, Bova FJ.
Linear accelerator radiosurgery for nonacoustic schwannomas. Int J Radiat
tion has long been a primary treatment option. However, only
Oncol Biol Phys 1999; 43: 545–548
after the advent of modern skull base techniques have the mor- [9] Fukaya R, Yoshida K, Ohira T, Kawase T. Trigeminal schwannomas: experience
bidity and mortality been reduced to reasonable levels. with 57 cases and a review of the literature. Neurosurg Rev 2010; 34: 159–
Stereotactic radiosurgery has become an established primary 171
and adjuvant treatment modality for skull base neoplasms, [10] Goldenberg RA, Gardner G. Tumors of the jugular foramen: surgical preserva-
tion of neural function. Otolaryngol Head Neck Surg 1991; 104: 129
including NVCNS. Radiosurgery avoids most surgically related
[11] Ojemann RG. Skull-base surgery: a perspective. J Neurosurg 1992; 76: 569–
complications. The rates of new or worsened cranial neuropa- 570
thy after SRS compare favorably with the results of microsur- [12] Netterville JL, Civantos FJ. Rehabilitation of cranial nerve deficits after neuro-
gery. Direct comparisons of surgery and SRS are difficult tologic skull base surgery. Laryngoscope 1993; 103 (Suppl 60): 45–54
[13] Kida Y, Yoshimoto M, Hasegawa T. Radiosurgery for facial schwannoma. J
because surgery is often reserved for larger symptomatic
Neurosurg 2007; 106: 24–29
lesions, and SRS cohorts include a meaningful number of post-
[14] Kano H, Niranjan A, Kondziolka D, Flickinger JC, Dade Lunsford L. Stereotactic
surgical recurrences. It is not clear that the theoretical benefits radiosurgery for trigeminal schwannoma: tumor control and functional pres-
of fractionated approaches have been established in the current ervation Clinical article. J Neurosurg 2009; 110: 553–558
literature to convincingly justify its use versus single-session [15] Hasegawa T. Stereotactic radiosurgery for nonvestibular schwannomas. Neu-
rosurg Clin N Am 2013; 24: 531–542
SRS. There will not be a one-size-fits-all therapy for NVCNS.
[16] Pollock BE, Kondziolka D, Flickinger JC, Maitz A, Lunsford LD. Preservation of
Instead, therapy needs to be individualized. Large tumors or cranial nerve function after radiosurgery for nonacoustic schwannomas. Neu-
tumors causing symptoms from mass effect are candidates for rosurgery 1993; 33: 597–601
surgical resection. Smaller tumors and tumors in patients con- [17] Muthukumar N, Kondziolka D, Lunsford LD, Flickinger JC. Stereotactic radio-
sidered a high risk for surgery can be addressed safely by SRS. surgery for jugular foramen schwannomas. Surg Neurol 1999; 52: 172–179
[18] Kida Y, Kobayashi T, Tanaka T, Oyama H, Niwa M. [A new strategy for the
▶ Fig. 17.6 shows a treatment algorithm that can be applied to
treatment of jugular foramen tumors using radiosurgery]. No Shinkei Geka
patients harboring NVCNS. 1995; 23: 671–675
[19] Yasui T, Hakuba A, Kim SH, Nishimura S. Trigeminal neurinomas: operative
approach in eight cases. J Neurosurg 1989; 71: 506–511
References [20] Samii M, Migliori MM, Tatagiba M, Babu R. Surgical treatment of trigeminal
schwannomas. J Neurosurg 1995; 82: 711–718
[1] Safavi-Abbasi S, Bambakidis NC, Zabramski JM, et al. Nonvestibular schwan- [21] Day JD, Fukushima T. The surgical management of trigeminal neuromas. Neu-
nomas: an evaluation of functional outcome after radiosurgical and micro- rosurgery 1998; 42: 233–240, discussion 240–241
surgical management. Acta Neurochir (Wien) 2010; 152: 35–46 [22] Sharma BS, Ahmad FU, Chandra PS, Mahapatra AK. Trigeminal schwannomas:
[2] Fisher LM, Doherty JK, Lev MH, Slattery WH III. Distribution of nonvestibular experience with 68 cases. J Clin Neurosci 2008; 15: 738–743
cranial nerve schwannomas in neurofibromatosis 2. Otol Neurotol 2007; 28: [23] Zhang L, Yang Y, Xu S, Wang J, Liu Y, Zhu S. Trigeminal schwannomas: a re-
1083–1090 port of 42 cases and review of the relevant surgical approaches. Clin Neurol
[3] Huang CF, Kondziolka D, Flickinger JC, Lunsford LD. Stereotactic radiosur- Neurosurg 2009; 111: 261–269
gery for trigeminal schwannomas. Neurosurgery 1999; 45: 11–16, discus- [24] Wanibuchi M, Fukushima T, Zomordi AR, Nonaka Y, Friedman AH. Trigeminal
sion 16 schwannomas: skull base approaches and operative results in 105 patients.
[4] Pan L, Wang EM, Zhang N, et al. Long-term results of Leksell gamma knife sur- Neurosurgery 2012; 70 (Suppl Operative): 132–143, discussion 143–144
gery for trigeminal schwannomas. J Neurosurg 2005; 102 (Suppl): 220–224 [25] Hasegawa T, Kida Y, Yoshimoto M, Koike J. Trigeminal schwannomas: results
[5] Pollock BE, Foote RL, Stafford SL. Stereotactic radiosurgery: the preferred ma- of gamma knife surgery in 37 cases. J Neurosurg 2007; 106: 18–23
nagement for patients with nonvestibular schwannomas? Int J Radiat Oncol [26] Phi JH, Paek SH, Chung HT, et al. Gamma Knife surgery and trigeminal
Biol Phys 2002; 52: 1002–1007 schwannoma: is it possible to preserve cranial nerve function? J Neurosurg
[6] Sun S, Liu A, Wang C, Luo B, Wang M. Clinical analysis of Gamma Knife sur- 2007; 107: 727–732
gery for trigeminal schwannomas. J Neurosurg 2006; 105 (Suppl): 144–148 [27] Martin JJ, Kondziolka D, Flickinger JC, Mathieu D, Niranjan A, Lunsford LD.
[7] Nedzelski J, Tator C. Other cerebellopontine angle (non-acoustic neuroma) tu- Cranial nerve preservation and outcomes after stereotactic radiosurgery for
mors. J Otolaryngol 1982; 11: 248–252 jugular foramen schwannomas. Neurosurgery 2007; 61: 76–81, discussion 81

144
Stereotactic Radiosurgery for Nonvestibular Schwannomas

[28] Peker S, Sengöz M, Kılıç T, Pamir MN. Gamma knife radiosurgery for jugular [38] Schisano G, Olivecrona H. Neurinomas of the Gasserian ganglion and trigemi-
foramen schwannomas. Neurosurg Rev 2012; 35: 549–553, discussion 553 nal root. J Neurosurg 1960; 17: 306–322
[29] Zhang N, Pan L, Dai JZ, Wang BJ, Wang EM, Cai PW. Gamma knife radiosurgery [39] Bulsara KR, Sameshima T, Friedman AH, Fukushima T. Microsurgical manage-
for jugular foramen schwannomas. J Neurosurg 2002; 97 (Suppl): 456–458 ment of 53 jugular foramen schwannomas: lessons learned incorporated into
[30] Elsharkawy M, Xu Z, Schlesinger D, Sheehan JP. Gamma Knife surgery for a modified grading system. J Neurosurg 2008; 109: 794–803
nonvestibular schwannomas: radiological and clinical outcomes. J Neurosurg [40] Samii M, Babu RP, Tatagiba M, Sepehrnia A. Surgical treatment of jugular
2012; 116: 66–72 foramen schwannomas. J Neurosurg 1995; 82: 924–932
[31] Litre CF, Gourg GP, Tamura M, et al. Gamma knife surgery for facial nerve [41] Sanna M, Bacciu A, Falcioni M, Taibah A. Surgical management of jugular fora-
schwannomas. Neurosurgery 2007; 60: 853–859, discussion 853–859 men schwannomas with hearing and facial nerve function preservation: a
[32] Madhok R, Kondziolka D, Flickinger JC, Lunsford LD. Gamma knife radiosurgery series of 23 cases and review of the literature. Laryngoscope 2006; 116:
for facial schwannomas. Neurosurgery 2009; 64: 1102–1105, discussion 1105 2191–2204
[33] Wallner KE, Pitts LH, Davis RL, Sheline GE. Radiation therapy for the treat- [42] Sedney CL, Nonaka Y, Bulsara KR, Fukushima T. Microsurgical management of
ment of non-eight nerve intracranial neurilemmoma. Int J Radiat Oncol Biol jugular foramen schwannomas. Neurosurgery 2013; 72: 42–46, discussion 46
Phys 1988; 14: 287–290 [43] McRackan TR, Rivas A, Wanna GB, et al. Facial nerve outcomes in facial nerve
[34] Zabel A, Debus J, Thilmann C, Schlegel W, Wannenmacher M. Management of schwannomas. Otol Neurotol 2012; 33: 78–82
benign cranial nonacoustic schwannomas by fractionated stereotactic radio- [44] McMonagle B, Al-Sanosi A, Croxson G, Fagan P. Facial schwannoma: results of
therapy. Int J Cancer 2001; 96: 356–362 a large case series and review. J Laryngol Otol 2008; 122: 1139–1150
[35] Showalter TN, Werner-Wasik M, Curran WJ Jr, Friedman DP, Xu X, Andrews [45] Sheehan J, Yen CP, Arkha Y, Schlesinger D, Steiner L. Gamma knife surgery for
DW. Stereotactic radiosurgery and fractionated stereotactic radiotherapy for trigeminal schwannoma. J Neurosurg 2007; 106: 839–845
the treatment of nonacoustic cranial nerve schwannomas. Neurosurgery [46] Fukuda M, Oishi M, Saito A, Fujii Y. Long-term outcomes after surgical treat-
2008; 63: 734–740, discussion 740 ment of jugular foramen schwannoma. Skull Base 2009; 19: 401–408
[36] Nishioka K, Abo D, Aoyama H, et al. Stereotactic radiotherapy for intracranial [47] Yianni J, Dinca EB, Rowe J, Radatz M, Kemeny AA. Stereotactic radiosur-
nonacoustic schwannomas including facial nerve schwannoma. Int J Radiat gery for trigeminal schwannomas. Acta Neurochir (Wien) 2012; 154:
Oncol Biol Phys 2009; 75: 1415–1419 277–283
[37] Choi CY, Soltys SG, Gibbs IC, et al. Stereotactic radiosurgery of cranial nonves- [48] Kimball MM, Foote KD, Bova FJ, Chi YY, Friedman WA. Linear accelerator ra-
tibular schwannomas: results of single- and multisession radiosurgery. Neu- diosurgery for nonvestibular schwannomas. Neurosurgery 2011; 68: 974–
rosurgery 2011; 68: 1200–1208, discussion 1208 984, discussion 984

145
Stereotactic Radiosurgery for Hemangioblastomas

18 Stereotactic Radiosurgery for Hemangioblastomas


Hideyuki Kano and L. Dade Lunsford

interventions against the slowly progressive natural history of


Key Points the disease. Surgical resection is the usual treatment of choice
for most symptomatic hemangioblastomas.11 However, vascular-
● Hemangioblastomas of the brain present as sporadic lesions ity or critical location often makes complete resection impossi-
or as manifestations of von Hippel-Lindau disease. ble. In patients with multiple hemangioblastomas, complete
● Surgical resection is the usual treatment of choice for most resection of all tumors is not feasible. Furthermore, cystic lesions
symptomatic hemangioblastomas, but vascularity or critical tend to have a more favorable surgical outcome compared with
location often makes complete resection impossible. the more vascular solid lesions. Wang et al12 reported 39
● Stereotactic radiosurgery is an important tool in selected hemangioblastoma patients who underwent radical surgical
patients with hemangioblastomas; it is associated with a resection. Postoperatively, 11 patients deteriorated after surgery
satisfactory tumor control rate as well as relatively low risk of and two patients died. Jagannathan et al13 reported 80 heman-
adverse radiation effects. gioblastoma patients (who underwent 126 surgical resections
for 164 cerebellar hemangioblastomas) and noted no tumor
recurrences at an average of 5 years after surgery. In contrast,
subtotal resection is associated with a high risk of tumor recur-
18.1 Introduction rence. Conway et al3 reported 40 hemangioblastoma patients
who underwent partial resection and noted progression in 8
Hemangioblastomas are rare, highly vascular, but often well-
patients (20%) and additional morbidity in 6 patients (15%).
circumscribed tumors with cystic components of the central
nervous system (CNS) and are most often detected in the poste-
rior fossa.1,2 Hemangioblastomas of the brain present both as
sporadic lesions or as manifestations of von Hippel-Lindau 18.4 Fractionated Radiation
(VHL) disease.3,4,5 In approximately 75 to 80% of patients, the Therapy
hemangioblastoma is a sporadic, single lesion involving the cer-
ebellum, brainstem, or upper cervical spinal cord. 6 Approxi- Conventional radiation therapy (RT) has been used for residual,
mately 20 to 25% of hemangioblastomas are associated with recurrent, or inaccessible tumors.11,14 Smalley et al14 treated 27
VHL disease.6 Although they occur throughout the cerebrospi- hemangioblastoma patients with fractionated RT after incom-
nal axis, multifocal hemangioblastomas associated with VHL plete surgical removal. The recurrence-free survival rates at 5,
disease predominate in the posterior fossa.3,7 10, and 15 years were 76, 52, and 42%, respectively. Local con-
trol was significantly better in patients receiving ≥ 50 Gy com-
pared to those treated with lower doses (57% vs. 33%).
18.2 Von Hippel-Lindau Disease Radiation therapy may be administered following resection. In
contrast, the progression-free survival after SRS for all heman-
Von Hippel-Lindau (VHL) disease is an autosomal dominant
gioblastomas was 89.9 and 85.9% at 5 and 10 years, respec-
neoplasia syndrome resulting from a germline mutation of the
tively.15 These data suggest that local control rate after SRS may
VHL tumor suppressor gene located on chromosome 3. 8
be enhanced compared to RT. However, no prospective trial has
Patients with VHL disease are predisposed to develop CNS and
been performed.
visceral lesions including renal cell carcinoma, pancreatic neu-
roendocrine tumors, and pheochromocytomas.9 Hemangioblas-
tomas in the CNS are a major cause of morbidity and mortality.
Ammerman et al10 reported the long-term natural history of
18.5 Stereotactic Radiosurgery
VHL-associated hemangioblastomas. Hemangioblastomas tend During the past 25 years stereotactic radiosurgery (SRS) has
to exhibit a saltatory growth pattern characterized by periods been used as a minimally invasive primary, adjuvant, or salvage
of growth over an average of 13 ± 15 months followed by peri- option for hemangioblastomas (▶ Table 18.1).1,5–24 Although
ods of dormancy for an average of 25 ± 19 months. Despite surgical resection and radiation therapy both have a role in the
measurable growth in almost all hemangioblastomas (97% of treatment of hemangioblastomas, the role of SRS has not been
patients), only 41% of patients became symptomatic. Forty-five well defined. Stereotactic radiosurgery is applied most com-
percent of the hemangioblastomas that eventually produced monly for residual tumors that were not completely removed
symptoms were present at the time of the initial magnetic reso- or for patients with deep-seated tumors that are considered to
nance imaging (MRI) study. have excessive risks for microsurgery (▶ Fig. 18.1 and
▶ Fig. 18.2). Patrice et al21 reported an 86% actuarial rate of
tumor control at 2 years after SRS. Niemela et al25 reported 11
18.3 Surgical Resection of hemangioblastoma cases and showed a reduction in the treated
Hemangioblastomas volume in 6 tumors at a median 30 months after SRS. Chang et
al17 reported that in 13 patients (with 29 hemangioblastomas)
Management of hemangioblastomas frequently requires weigh- who underwent linear accelerator (linac) radiosurgery, a higher
ing the benefits and potential complications of therapeutic dose improved results.

146
Stereotactic Radiosurgery for Hemangioblastomas

Table 18.1 Summary of studies of stereotactic radiosurgery for hemangioblastomas


Study No. of patients/ Mean follow-up Mean tumor vol- Mean margin dose 5-y OAS 5-y PFS
tumors (mo) ume or diameter (Gy)

Patrice et al, 199621 22/38 24.5 0.97 cc 15.5 88% 86%


(2 y) (2 y)

Niemela et al, 10/11 43 13.5 mm 20 67.5% 100%


199625

Chang et al, 199817 13/29 43 1.6 cc 23.2 92.3% (overall) 96.6%


(overall)

Pan et al, 199819 13/20 29 20 mm 18.4 92.3% (overall) 69.2%


(overall)

Wang et al, 200524 35/93 66 13 mm 17.2 69.0% 82.9%


(overall) (overall)

Tago et al, 200523 13/38 36 0.23 cc 20 80.8% 96.2%

Kano et al, 200815 32/74 61.4 0.72 cc 16 94.4% 89.9%

Moss et al, 20097 31/94 69 1.8 cc 23.4 NA 82%

Sayer et al, 201122 14/26 55.4 1.65 cc 18 NA 74%

Abbreviations: NA, not available; OAS, overall survival; PFS, progression-free survival.

Fig. 18.1 (a) A T1-weighted contrast-enhanced


axial magnetic resonance image (MRI) of a 50-
year-old male shows a sporadic cystic heman-
gioblastoma before surgical resection. (b) A T1-
weighted contrast-enhanced axial MRI shows
residual tumor after surgical resection. (c) A T1-
weighted contrast-enhanced axial MRI shows
tumor recurrence at the time of stereotactic
radiosurgery (SRS). Tumor volume was 3.4 cc and
margin dose was 18 Gy. (d) A T1-weighted
contrast-enhanced axial MRI shows tumor re-
gression 10 years after SRS.

Wang et al24 reported 35 patients with 93 hemangioblasto- et al22 reported a 5-year PFS of 74% in 14 patients with 26
mas treated with Gamma Knife SRS (GKSRS). The 5-year pro- hemangioblastomas. Tago et al23 reported actuarial 5- and 10-
gression-free survival was 71% and the actuarial survival rate year control rates of 96.2%. Kano et al15 reported that tumor
was 83%. Matsunaga et al18 reported 22 patients with 67 volume played a role in progression-free survival. Margin doses
hemangioblastomas treated with GKSRS. The progression-free of ≥ 15, 16, and 18 Gy in patients with VHL-related hemangio-
survival after SRS was 88% at 5 years and 78% at 10 years. Sayer blastoma were associated with improved progression-free

147
Stereotactic Radiosurgery for Hemangioblastomas

Fig. 18.2 A T1-weighted contrast-enhanced axial


magnetic resonance image of a 40-year-old
woman shows cystic and solid von Hippel-Lindau
disease–related hemangioblastomas in multiple
locations at the time of stereotactic radiosurgery
(SRS). She underwent prior surgical resections 16
and 2 years before SRS.

survival. Moss et al7 indicated that cessation of cyst fluid pro- in VHL patients and 15 Gy in patients with sporadic hemangio-
duction typically took over 1 year in their series of patients, and blastomas. At a median of 5 years (0.5–18 years), 20 patients had
in four patients, associated mass effects necessitated drainage died from intracranial disease progression and 9 patients had died
of the associated cyst or resection of the tumor nodule. They from other causes. The overall survival was 94% at 3 years, 90% at
concluded that SRS is effective in treating only the mural nod- 5 years, and 74% at 10 years. Factors associated with longer sur-
ule of hemangioblastomas, as opposed to an associated cystic vival included younger age, no neurologic symptoms, fewer
component; it is not the ideal treatment for quickly reducing tumors, and higher Karnofsky Performance Status. Thirty of 80
associated mass effect symptoms. However, in patients with (38%) VHL patients developed new tumors and 15 (14%) of 106
surgically inaccessible tumors, radiosurgery can be a useful sporadic hemangioblastoma patients had recurrences of residual
alternative, as tumor control usually leads to ultimate resolu- tumor from the original tumor. The 5-year rate of developing a
tion of the cystic component as well. Sayer et al26 treated 14 new tumor was 39% for VHL patients and a recurrence of residual
patients with 26 hemangioblastomas by GKSRS. In their analy- tumor from the original tumor was 21% for sporadic hemangio-
sis, tumor progression was more common in sporadic patients, blastoma patients. Factors associated with a reduced risk of devel-
women, and in patients with larger tumors. In patients with oping a new tumor or recurrence of residual tumor included
multiple hemangioblastomas as compared with those with only younger age, fewer tumors, and sporadic rather than VHL-associ-
a solitary hemangioblastoma, the radiosurgically treated lesion ated hemangioblastomas. The progression-free survival of treated
was 7.9 times more likely to progress after SRS. tumors was 92% at 3 years, 89% at 5 years, and 79% at 10 years.
Factors associated with an improved progression-free survival
included VHL-associated hemangioblastoma, solid tumor, smaller
18.6 Multicenter Retrospective tumor volume, and higher marginal dose. Twelve patients (6%)
Trial developed adverse radiation effects (AREs) after SRS and one pa-
tient died due to AREs.
Six participating centers of the North American Gamma Knife
Consortium and 13 Japanese GK centers identified 186 patients
with 517 hemangioblastomas who underwent SRS.27 Eighty
patients had 335 hemangioblastomas associated with VHL disease
18.7 Summary
and 106 patients had 182 sporadic hemangioblastomas. The Stereotactic radiosurgery is an important tool in selected
median tumor volume was 0.2 cc (median diameter: 7 mm) in patients with hemangioblastomas: It is associated with a satis-
VHL patients and 0.7 cc (median diameter: 11 mm) in sporadic factory tumor control rate as well as relatively low risk of
hemangioblastoma patients. The median margin dose was 18 Gy adverse radiation effects. Stereotactic radiosurgery can be

148
Stereotactic Radiosurgery for Hemangioblastomas

applied for growing residual tumors, for patients with progres- [13] Jagannathan J, Lonser RR, Smith R, DeVroom HL, Oldfield EH. Surgical ma-
nagement of cerebellar hemangioblastomas in patients with von Hippel-Lin-
sive deep-seated tumors who are high risk for microsurgery, for
dau disease. J Neurosurg 2008; 108: 210–222
VHL patients who develop new tumors, and for recurrences of [14] Smalley SR, Schomberg PJ, Earle JD, Laws ER Jr, Scheithauer BW, O’Fallon JR.
residual tumor from the original tumor in sporadic hemangio- Radiotherapeutic considerations in the treatment of hemangioblastomas of
blastoma patients who progress under serial observation. the central nervous system. Int J Radiat Oncol Biol Phys 1990; 18: 1165–1171
[15] Kano H, Niranjan A, Mongia S, Kondziolka D, Flickinger JC, Lunsford LD. The
role of stereotactic radiosurgery for intracranial hemangioblastomas. Neuro-

References surgery 2008; 63: 443–450, discussion 450–451


[16] Asthagiri AR, Mehta GU, Zach L, et al. Prospective evaluation of radiosurgery
[1] Chakraborti PR, Chakrabarti KB, Doughty D, Plowman PN. Stereotactic multi- for hemangioblastomas in von Hippel-Lindau disease. Neuro-oncol 2010; 12:
ple are radiotherapy. IV—haemangioblastoma. Br J Neurosurg 1997; 11: 110– 80–86
115 [17] Chang SD, Meisel JA, Hancock SL, Martin DP, McManus M, Adler JR Jr. Treat-
[2] Wanebo JE, Lonser RR, Glenn GM, Oldfield EH. The natural history of heman- ment of hemangioblastomas in von Hippel-Lindau disease with linear accel-
gioblastomas of the central nervous system in patients with von Hippel-Lin- erator-based radiosurgery. Neurosurgery 1998; 43: 28–34, discussion 34–35
dau disease. J Neurosurg 2003; 98: 82–94 [18] Matsunaga S, Shuto T, Inomori S, Fujino H, Yamamoto I. Gamma knife radio-
[3] Conway JE, Chou D, Clatterbuck RE, Brem H, Long DM, Rigamonti D. Heman- surgery for intracranial haemangioblastomas. Acta Neurochir (Wien) 2007;
gioblastomas of the central nervous system in von Hippel-Lindau syndrome 149: 1007–1013, discussion 1013
and sporadic disease. Neurosurgery 2001; 48: 55–62, discussion 62–63 [19] Pan L, Wang EM, Wang BJ, et al. Gamma knife radiosurgery for hemangioblas-
[4] Filling-Katz MR, Choyke PL, Oldfield E, et al. Central nervous system involve- tomas. Stereotact Funct Neurosurg 1998; 70 (Suppl 1): 179–186
ment in Von Hippel-Lindau disease. Neurology 1991; 41: 41–46 [20] Park YS, Chang JH, Chang JW, Chung SS, Park YG. Gamma knife surgery for
[5] Sora S, Ueki K, Saito N, Kawahara N, Shitara N, Kirino T. Incidence of von Hip- multiple hemangioblastomas. J Neurosurg 2005; 102 (Suppl): 97–101
pel-Lindau disease in hemangioblastoma patients: the University of Tokyo [21] Patrice SJ, Sneed PK, Flickinger JC, et al. Radiosurgery for hemangioblastoma:
Hospital experience from 1954–1998. Acta Neurochir (Wien) 2001; 143: results of a multiinstitutional experience. Int J Radiat Oncol Biol Phys 1996;
893–896 35: 493–499
[6] Neumann HP, Berger DP, Sigmund G, et al. Pheochromocytomas, multiple en- [22] Sayer FT, Nguyen J, Starke RM, Yen CP, Sheehan JP. Gamma knife radiosurgery
docrine neoplasia type 2, and von Hippel-Lindau disease. N Engl J Med 1993; for intracranial hemangioblastomas—outcome at 3 years. World Neurosurg
329: 1531–1538 2011; 75: 99–105, discussion 45–48
[7] Moss JM, Choi CY, Adler JR Jr, Soltys SG, Gibbs IC, Chang SD. Stereotactic ra- [23] Tago M, Terahara A, Shin M, et al. Gamma knife surgery for hemangioblasto-
diosurgical treatment of cranial and spinal hemangioblastomas. Neurosur- mas. J Neurosurg 2005; 102 (Suppl): 171–174
gery 2009; 65: 79–85, discussion 85 [24] Wang EM, Pan L, Wang BJ, et al. The long-term results of gamma knife radio-
[8] Seizinger BR, Rouleau GA, Ozelius LJ, et al. Von Hippel-Lindau disease maps to surgery for hemangioblastomas of the brain. J Neurosurg 2005; 102 (Suppl):
the region of chromosome 3 associated with renal cell carcinoma. Nature 225–229
1988; 332: 268–269 [25] Niemelä M, Lim YJ, Söderman M, Jääskeläinen J, Lindquist C. Gamma knife ra-
[9] Lonser RR, Glenn GM, Walther M, et al. von Hippel-Lindau disease. Lancet diosurgery in 11 hemangioblastomas. J Neurosurg 1996; 85: 591–596
2003; 361: 2059–2067 [26] Sayer FT, Nguyen J, Starke RM, Yen CP, Sheehan JP. Gamma knife radiosurgery
[10] Ammerman JM, Lonser RR, Dambrosia J, Butman JA, Oldfield EH. Long-term for intracranial hemangioblastomas—outcome at 3 years. World Neurosurg
natural history of hemangioblastomas in patients with von Hippel-Lindau 2011; 75: 99–105, discussion 45–48
disease: implications for treatment. J Neurosurg 2006; 105: 248–255 [27] Kano H, Shuto T, Iwai Y, et al. Stereotactic radiosurgery for intracranial he-
[11] Sung DI, Chang CH, Harisiadis L. Cerebellar hemangioblastomas. Cancer 1982; mangioblastomas: a retrospective international outcome study. J Neurosurg
49: 553–555 2015. In press
[12] Wang C, Zhang J, Liu A, Sun B. Surgical management of medullary hemangio-
blastoma. Report of 47 cases. Surg Neurol 2001; 56: 218–226, discussion
226–227

149
Stereotactic Radiosurgery for Vestibular Schwannomas

19 Stereotactic Radiosurgery for Vestibular Schwannomas


Jean Régis, Romain Carron, Christine Delsanti, Denis Porcheron, Jean-Marc Thomassin, Xavier Muracciole, and Pierre-Hugues Roche

from pneumoencephalography, rudimentary head fixation, and


Key Points very-high-dose prescription.5 Over time, the pioneers under-
stood that the therapeutic goal of long-term tumor control was
● The superior safety efficacy of stereotactic radiosurgery over achievable by using a lower regimen of dose while sparing the
microsurgery in small- to middle-sized vestibular schwanno- motor facial nerve, and without compromising hearing and tri-
mas is demonstrated in five comparative studies. Normal geminal nerve sensory function.6 This preliminary experience
motor facial nerve function and serviceable hearing are more stressed the need for 2 years of follow-up for proper assessment
likely to be preserved with radiosurgery as compared with of the clinical outcome, and at least 3 years for the evaluation of
microsurgery. tumor control.
● In very large Koos stage IV schwannomas, a combined Over the last three decades, microsurgery (MS) and stereo-
approach with a deliberate subtotal removal with functional tactic radiosurgery (SRS) have become well-established ma-
monitoring of the facial nerve followed by radiosurgery of nagement options for VSs. The evolution in the management of
the remnant dramatically reduces the risk of facial palsy as VSs can be separated into three periods: the microsurgical pio-
compared with radical removal. neers period; the demonstration of SRS as a first-line therapy
● Hypofractionation of radiosurgery (stereotactic radiotherapy for small- and medium-sized VSs; and at present, a period of
or multisession radiosurgery) has failed until now to SRS maturity based on a large worldwide accrual of patients.
demonstrate any advantage over single-dose, high-precision The tumor size is classified according to Koos et al,7 the extent
radiosurgery. of the tumor in the bottom of the internal auditory canal
according to Ohata et al,8 the functionality of hearing according
to Gardner and Robertson,9 and the facial motor nerve function
according to House.10 To date, five comparative studies (two
19.1 Introduction level II and three level III) published in peer-review journals
have demonstrated improved hearing preservation rate, a bet-
Acoustic neuromas are usually benign tumors of the cerebello-
ter motor facial function, and a similar long-term tumor control
pontine angle (CPA) arising from the Schwann cells of the
when SRS is performed as compared with MS for tumors of the
sheath of the vestibular nerve. At the beginning of the 20th cen-
same size.11–15 Additionally, the direct and indirect costs are
tury, pioneers such as Cushing, based on crude preoperative
much lower with SRS.15,16
work-up (X-ray of the internal auditory canal, audiometry, and
Given the long life expectancy of these patients and the
vestibulometry), removed huge tumors in patients presenting
severe potential disability related to the functional risks inher-
with neurologic symptoms.1 Surgery was performed through a
ent to the management of these tumors, VSs should be man-
suboccipital approach with the only goal to save the life of the
aged by an experienced multidisciplinary team that is able to
patient. Surgery performed through a suboccipital approach
integrate all micro- and radiosurgical approaches to provide the
was at that time attempted as a life-saving procedure and lim-
highest level of care and the highest probability of functional
ited concern was given to other neurologic function (e.g., crani-
preservation to ensure good quality of life.
al nerve [CN] VII or VIII preservation). Surgery was usually
incomplete, the mortality was high, and facial nerve function
and hearing were systematically sacrified. 1
Following the advent of ventriculography with Dandy in the
19.2 Therapeutic Options and/or
1920s, the preoperative diagnosis was improved and further Surgical Technique(s)
refined by angiography and cisternography. Complete removal
The main therapeutic options for VSs are a “wait-and-see”
became a realistic goal.2 In the 1960s, tumors were more fre-
strategy,17 radiosurgery, SRS (2–10 fractions), and microsurgical
quently detected at an earlier stage with only otologic symp-
resection (either through a retrosigmoid translabyrinthine or
toms. Otologists and neurosurgeons then developed new
suprapetrous approach).
surgical approaches such as the translabyrinthine and the
suprapetrous approaches.3 Technical advances—especially the
introduction of the operative microscope—allowed operation 19.2.1 Simple Follow-ups Based on
on these smaller tumors with the aim to preserve anatomically
and sometimes even functionally the motor function of the
Clinical Interviews and Serial MRI Scans
facial nerve. Simple follow-ups (i.e., observation) based on clinical inter-
In the 1970s and 1980s, with the advent of computed tomog- views and serial MRI scans have been advocated for some
raphy (CT) and magnetic resonance imaging (MRI), even early time.18 For several years, this has been the standard in our
diagnosis of smaller tumors became commonplace. In 1968, group for small intracanalicular VSs. Under conditions of suffi-
Leksell proposed the introduction of Gamma Knife radiosurgery ciently long periods between follow-ups, combined with strict
(GKRS) for the management of vestibular schwannomas (VSs).4 criterion for defining growth, the vast majority of the modern
In 1968, the first cases of VSs were treated using the Gamma observational series demonstrates that the subtotality of
Knife. The procedure was based on crude localization derived tumors will grow and that the majority of patients, whose

150
Stereotactic Radiosurgery for Vestibular Schwannomas

hearing was still functional at the commencement of the wait- patients. This rate reached 95% of patients who had no prior
and-see strategy, will progressively lose their hearing over the history of sudden hearing loss. Other predictors of functional
course of a few years.17,19,20 hearing preservation were age (specifically younger), small size
Upon their very comprehensive review of the literature (34 of the tumor, and a dose to the modiolus of the cochlea < 4 Gy.
series), Sughrue and colleagues17 found that 982 patients, who We observed no radioinduced tumor. Only large Koos stage IV
had follow-ups over a period of 26 and 52 months, had a mean VSs were contraindicated for up-front radiosurgery. For
growth of VSs of 2.9 mm/y. Fifty percent of the patients lost patients harboring large-sized Koos stage IV VSs, we used a
their hearing, despite the short duration of the maximum fol- combined approach of deliberate partial microsurgical removal
low-up relative to their life expectancy. The authors concluded followed by radiosurgery of the residue. This cohort is unique
that the speed of the tumor’s growth is a much more reliable because of the size of its population and length of the follow-
predictor of hearing loss than the initial size of the VS. Bakkouri up; it demonstrates the efficacy of radiosurgery as well as its
et al21 published a series detailing 325 patients in which they safety with regard to its high rate of hearing preservation.
emphasized the difficulties faced in maintaining rigorous pa- At our center, patients were preoperatively evaluated and
tient follow-up of conservatively managed patients. In fact, they prospectively followed up with a clinical evaluation comprising
report a tracking loss of 24% of the 325 patients. This loss the following: pure tone audiometry (PTA) and speech discrim-
occurred despite their efforts to retain and maintain contact ination score (SDS), vestibulometry, the Schirmer test, and seri-
and follow-ups. We have also recently done a comparison of al MRI scans (at 6 months and 1, 2, 3, 5, 7, 10, and 15 years).
this wait-and-see attitude with an early proactive radiosurgical One hundred forty-eight patients with NF2 harbored VSs. A fol-
management strategy.20 We found (p = 0.0009) that at 3 years, low-up of more than 3 years is available for 2,336 patients (NF2
patients who were operated on by Gamma Knife had a 73.3% excluded). The mean age at the time of radiosurgery is 66.3
chance of preserving hearing as opposed to 35% with the mere years. The Koos stage was stage I in 17.6% of patients, stage II in
wait-and-see strategy.20 Hence, radiosurgery is the preferred 51.8% of patents, stage III in 27% patients, and stage IV in 3.6% of
treatment option for these patients to maintain their residual patients. Initial symptoms were as follows: hypoacousia in
serviceable hearing. 49.5% of patients, tinnitus in 19.4%, vertigo in 13.2%, and 5.1%
In another study, we placed our focus on patients who had experienced instability. A past history of sudden hearing loss
several years of serial pure tonal audiogram before radiosur- was reported in 21.5% of patients. It is important to note that
gery.22 Among 72 consecutive patients, we observed an annual microsurgical resection of the VS had been performed prior to
hearing loss of 3.72 dB/y before radiosurgery, 4.06 dB/y the first radiosurgery in 7% of these cases. The day before intervention,
year after GKRS, and 1.24 dB/y in the following years. This in 87% of cases patients reported hypoacousia, 65% reported
study22 supports the findings from the previous study, and it tinnitus, 52% reported imbalance, 31% reported veritgo, 8.2%
suggests a marginal additional risk of hearing loss correspond- reported facial palsy, 7.5% reported a hemifacial spasm, 5.3%
ing to radiosurgical toxicity (difference not significant).23 We reported trigeminal neuralgia, and 4.8% of the patients
have successfully demonstrated that in younger patients the reported hypoesthesia. Our methodology has been previously
probability of preserving functional hearing is high.24 Our find- described.26
ings have subsequently been confirmed by Lobato-Polo et al, 25 Defining the limits of the tumor is first done on the stereotac-
who provides additional support in favor of earlier proactive tic three-dimensional T1-weighted gadolinium-enhanced MRI
radiosurgical management of these patients. sequence (magnetization prepared rapid acquisition gradient
echo [MP-RAGE] Siemens or comparable sequences). The
absence of distortion between the MRI and stereotactic CT is
19.2.2 Radiosurgery systematically checked with scrutiny and a shift of the dose
Radiosurgery in the management of VSs is hereafter analyzed plan is performed consequently if and when necessary. The lim-
based on a review of our own prospective cohort as well as cur- its of the internal auditory canal, vestibule, semicircular canals,
rent literature resources. Of 13,341 GK procedures performed and cochlea are defined on CT. Dose planning corresponding to
by the staff at the Stereotactic & Functional Neurosurgery the cisternal portion of the tumor, adjacent cochlear and facial
Department of Timone University Hospital (Marseilles, France), nerves, respectively, is corrected according to high-resolution
3,717 patients had VSs, 2,991 of whom were operated on T2-weighted (constructive interference in steady state [CISS]
between July 14, 1992, and June 1, 2011. All 2,991 patients have Siemens) with and without contrast. The dose to the margin is
been prospectively evaluated; 2,336 patients were nonneurofi- 12 Gy (Gardner-Robertson ≥ 3) and 11 Gy if hearing is still serv-
bromatosis type II (NF2; 164 treatments) with follow-ups lon- iceable.9 Our dose-selection policy has not changed since 1992
ger than 3 years. for VS, which confers a very good homogeneity to our series.
In 7% of these patients, the VSs were previously resected. Patients were treated using a Gamma Knife model B, 4, or 4C, or
According to the Koos’ classification system, the VSs were of Perfexion. Additionally, between 2005 and 2010 an extensive
stage I in 17.6%; stage II in 51.8%; stage III in 27%; and stage IV search and cross-referencing exercise was conducted on
in 3.6%. The mean tumor volume was 2.63 cc. In 46% of patients, PubMed, which produced 213 articles. Of these, 55 reported on
hearing was still serviceable at the time of the radiosurgery a series of more than 30 patients, providing us with safety effi-
according to the Garner-Robertson scale, although it was sub- cacy data. A second filtering enabled us to rule out any dupli-
normal in 20.9% of the patients. Long-term tumor control was cated series and general review papers (15 articles). The
achieved in 97.5% of the patients. A transient facial palsy was majority are retrospective studies. However, three studies were
observed in 0.5% of the cases. The rate of trigeminal injury was prospective, reporting on 69, 78, and 111 patients, respec-
0.5%. Serviceable hearing was preserved after 3 years in 78% of tively.27,28,29

151
Stereotactic Radiosurgery for Vestibular Schwannomas

19.2.3 The Prospective Cohort of The results of the 15 series of GKRS are fairly homogene-
ous.24,27,28,29,31–40 Only one linear accelerator (linac) series has
Timone Hospital been found. In this linac series, hearing was not evaluated. 41 In
The prospective cohort of Timone Hospital demonstrates tumor the GKRS series, tumor control between 3 and 10 years of fol-
control in 97.5% of the patients (▶ Fig. 19.1) in the last of neuro- low-up varied from 92 to 98% (97.5% for our series) and the
imaging (MRI) follow-ups. With 2.5% of patients, as a result of a tumor control rate was 90% in the one linac-based series. Trige-
continuous growth of the schwannoma at the 3-year mark, we minal injury rates ranged from 0 to 9% (0.5% in our series and
have consequently been led to propose new radiosurgery in 20 with the one linac series exhibiting a rate of 3.6%).
patients or a resection in 39 patients. The MRI follow-ups during The rate of facial palsy varied between 0 and 7% (0.5% in our
the first year revealed an average of 20% in schwannoma growth series and 4.4% with the linac series). The rate of serviceable
rate, followed thereafter by stabilization at 3 years after radio- hearing preservation ranged from 56.6 to 78.6% (63% in our ser-
surgery. In these patients with transient enlargement, a ies). Whether other linac-based radiosurgery devices (adapted
decrease in tumor volume was observed after the third year fol- linac, Novalis Radiosurgery [Brainlab, Munich, Germany], or
lowing SRS. At 7 years, the volume corresponded to 60% of the CyberKnife) actually allow or not to reach the same level of
volume at the time of radiosurgery (−40% decrease). The rate of results published in the GKRS literature is still a matter of con-
transient facial palsy was < 0.5%. This rate was 3% during the first troversy. A recent interesting paper from Gevaert et al working
period corresponding to our learning curve (with the first 100 with a Novalis compares VS dose planing from Novalis with
patients). The rate was subsequently reduced to 1.4% during the those of a CyberKnife and a Gamma Knife. These authors con-
period preceding the introduction of the dose-planning com- clude that “only GK Perfexion is complying with all SRS con-
puter workstations (involving 212 patients), and dropped fur- straints of high conformity while minimizing low-dose spread”
ther to 0.5% after the introduction of the workstations, and their and that “non-isocentric beams (CyberKnife) or intensity-
integration in anatomical imaging (360 patients). Since the modulated radiotherapy (IMRT) beams (Novalis-Tx-DMLC-
introduction of robotic radiosurgery (2,319 patients), this transi- IMRT) are spreading more low dose than multiple isocenters
ent facial palsy rate has virtually disappeared in unilateral VS.30 (Gamma Knife).”

19.3 Radiosurgery Main Series of 19.3.1 Stereotactic Radiotherapy


Stereotactic radiotherapy (SRT) was reported in seven series
Literature (▶ Table 19.1), and the experience was very heterogeneous in
The main series of radiosurgery literature and their results are terms of technique.42–48 Maire et al report treating much larger
summarized in ▶ Table 19.1. These series are very heterogene- tumors than the others.45 The Chang series was hypofractio-
ous in terms of devices, size of tumors, prescribed doses, inclu- nated (three fractions, 18–21 Gy).43 The others were hyperfrac-
sion or exclusion of NF2 patients, the rate of patients who had tionated from 25 to 32 fractions, delivering 45 to 57.6 Gy by
been operated on previously, methods of measurement, defini- fractions of 1.8 to 2 Gy. In Andrews et al’s series, the follow-up
tion of tumor control, and overall duration of follow-up time, was short, which makes it difficult to draw substantial conclu-
from when the patient is released postsurgery to the final out- sions.48 In the remaining series, tumor control varied from 93
patient visit. to 98%. The rate of injury of the trigeminal nerve varied

Fig. 19.1 Example of result of radiosurgery in a unilateral vestibular schwannoma patient with deafness at the time of radiosurgery. The long-term
magnetic resonance imaging follow-up demonstrates favorable efficacy with a dramatic regression of the lesion at 10 years.

152
Stereotactic Radiosurgery for Vestibular Schwannomas

Table 19.1 Radiosurgery and stereotactic radiotherapy for vestibular schwannomas: Analysis of the literature from 2005 to 2010 (MEDLINE) and com-
parison with our own series
Study Population Volume Technique Dose Follow-up (mo) Tumor control CN CN VII CN VIII Cancer
(cm3) (Gy) Lost to follow- (%) V (%) (%) (%)
up (%) (Audio)

Chung et al, 195 4.1 (0.04– GK 13 (11–18.2) 31 (1–110) 10 y: 96.8% 1.1 1.5 60% 0
2005 R31 39% 23.1) 2 lost

Lunsford et 829 2.5 GK 13 (10–20) NR 10 y: 98% 3.1 <1 78.6% 0


al, 2005 R33 20% > 10 y 252 pa-
tients

Wowra et al, 111 1.6 (0.08– GK 13 (10–16) 7 y (5–9.6) 6 y: 95% 2.7 2.7 NR 0
2005 R28 33.3% 8.7)

Van Eck et al, 78 2.28 (0.1– GK 13–65% 22 97.4% 3.8 1.2 69.2% 0
2005 P27 NR 11.7)

Hasegawa et 317 5.6 (0.2– GK 13.2 (10–18) 93 94.4% (10 y: 2 2? 67.5% 0


al, 2005 R32 22.7% 36.7) 29 lost 92%)
GR 1–2:
30.6%

Hempel et al, 123 1.6 (0.1– GK 13 (10–14.5) 98 (63–129) NR 5.8 0 NR 0


2006 R66 NR 9.9)

Liu et al, 74 10.8 GK 12.3 (12–14) 68.3 (30–122) 5 & 10 y: 7 5 72.3% 0


2006 R35 25.6% (0.11– 95.9%
27.8)

Hudgins et 159 3.3 cm3 GK 14 (8 at 20) 12 y 96.4% 0 0 NR 0


al, NR
2006 R34

Chopra et al, 216 1.3 (0.08– GK 13 (12–13) 68 (max: 143) 10 y: 98.3% 3.7 0 56.6% 0
2007 R36 0% 37.5)

Niranjan et 96 0.112 mm3 GK 13 (10–18) 28 (12–144) 99% 0 0 63.3% 0


al, NR (0.1–0.5)
2008 R37 Koos I

Régis et al, 184 NR GK 12 7 y (3–13) 98% 0.6 0.7 3 y: 60% 0


2008 P38 0% GR 1 and 2

Timmer et al, 69 2.28 GK 11 (9.3–12.5) 14 (3–56) NR 9 7 75% 0


2009 P29 NR (0.02– 16 lost
10.2)

Kano et al, 77 0.75 GK 12,5 (12–13) 20 mo (6–40) 97.4% NR 0 1 y: 0


2009 R40 0% (0.07–7,7) 89.3%
GR 1: 46 2 y:
GR 2: 31 66.8%

Tamura et al, 74 1.35 (0.06 GK 12 (9–13) 55.6 (3–11 y) 93% NR 0 3 y: 0


2009 P24 0% à 4.6) 78.4%
GR 1

Franzin et al, 50 0.73 GK 13 (12–16) 36 (6–96) 96% NR 0 68% 0


2009 R39 0% (0.03–6.6)
GR 1 or 2

Régis et al, 2087 2.63 GK 12.3 Minimum 3 y 97.5% 0.5 0.5 63% 0
2010 P20 7% GR 1 and 2:
46%

Friedman et 390 NR Linac 12.5 (10– 40 5 y: 90% 3.6 4.4 NR 0


al, 20% SRS 22.5) 42 lost
2006 R41

Combs et al, 106 3.9 (2.7– SRT 57.6 Gy 32 fr 48.5 (3–172) 3 y: 94.3% 4.7 2.3 NR 0
2005 R44 16% 30.7) Linac 1.8 Gy 6 wk 5 y: 93% (25%)
94%

153
Stereotactic Radiosurgery for Vestibular Schwannomas

Table 19.1 continued


Study Population Volume Technique Dose Follow-up (mo) Tumor control CN CN VII CN VIII Cancer
(cm3) (Gy) Lost to follow- (%) V (%) (%) (%)
up (%) (Audio)

Chan et al, 70 2.,4 (0.05– SRT 54 Gy 30 fr 45 3 y: 100% 4 13 NRa 0


2005 R42 21% 21.1) Linac 1.8 Gy 6 wk 5 y: 98%

Chang et al, 61 Diameter SRT 18–21 Gy, 3 fr 48 (36–62) 98.4% 0 3.3 74.3% 0
2005 R43 13% mean: Linac 6–7 Gy 3 d 14 dB
18.5 mm Cyber Knife loss
(5–32)

Koh et al, 60 4.9 (0.3– SRT 50 Gy 25 fr 5 31.9 (6.1– 5 y: 96.2% 1.6 1.6 NRa 1,7
2007 R46 NR 49) Linac wk 107.4) (33%)
X-Knife 77.3%

Andrews et 89 1.6 SRT 48.5 fr of 1.8 14.7 97.8% ? 0? 2.2 ? 73.3% ? 0?


al, Linac (50.4/46.8)
2009 R48

Thomas et al, 34 1.1 (0.3– SRT 45 in 25 fr of 2 36.5 (12–85) 95.7% 0 6 56% 0


2007 P47 9.6) Linac

Maire et al, 45 NR RT 50.4 in fr of 80 (4–227) 95% (2 dead) 0 0 NR 2,2


2006 R45 Linac 1.8

Abbreviations: (Audio), % of patients with audiometry before and after; CN, cranial nerve; dB, decibels; dcd, patient dead because of tumor growth;
dose, marginal dose; fr, fraction; GK, Gamma Knife; Linac, linear accelerator; GR, Gardner and Robertson Scale; NR, not reported; P, prospective; pt,
patients; R, retrospective; RT, radiotherapy; SRS, stereotactic radiosurgery; SRT, stereotactic radiotherapy; ?, no sufficient follow-up for a serious
evaluation.
aNo audiometry before and after (telephone evaluation).

between 0 and 4.7%; with motor facial nerve impairment, the 19.3.2 Large Koos IV Management
range was 0 to 13%. In three instances of these SRT series, there
was no systematic objective evaluation based on pre- and post- Microsurgical resection is systematically discussed in large Koos
operative PTA and SDS enabling the evaluation of the rate of stage IV VS. We have stopped advocating radical resection for
serviceable hearing preservation.42,44,46 For Combs et al, PTA VS patients. Our team systematically proposes a combined
was performed in only 25% of the patients44 and for Koh et al approach.65 The first step is a cautious deliberate partial remov-
only 33%.46 In Chan et al, the evaluation of serviceable hearing al under neuromonitoring followed some months after by SRS
preservation was based on results from telephone interviews of the residual tumor. This evolution has led our team to per-
conducted on the patients.42 Maire et al provides no informa- form fewer translabyrinthine approaches, which had been our
tion regarding hearing outcome.45 Series 2 only gives an objec- preferred approach for radical removal. Presently, we favor ret-
tive assessment on functional hearing, where it was preserved rosigmoid approaches.
in 56 and 74.3% of the patients.43,47 Chan’s results show the
average loss in conversational frequencies (PTA) is of 15 dB as
opposed to 10 dB in our series of GKRS.42 The rate of functional 19.4 Complications and Concerns
hearing preservation therefore does not appear to be superior
in the fractionated series. However, carcinogenesis is reported If there is ever a condition in which the efficacy of radiosurgery
in two of these fractionated series, with an incidence of 1.7 and compares favorably with microsurgical resection, it is definitely
2.2% in Koh et al and Maire et al.45,46 with small- to middle-sized VSs (Koos stages I–III) in young
Microsurgical resection in small- or middle-sized VS is nowa- patients. All five of the existing comparative studies have given
days less relevant in terms of efficacy. There appears to be little consistent results and demonstrated a much lower rate of facial
benefit (from the perspective of the patient) to favor MS over motor nerve impairment and a much higher rate of serviceable
SRS.49 However, in Koos stage III with a very cystic tumor pre- hearing preservation with GKRS as compared with microsurgi-
sentation and no remaining functional hearing, resection can cal resection.11–15
be proposed based on the argument that cystic tumors have The linac-based series is limited, but the Friedman series
been described to present substantial volume increase after reports similar results, at least in terms of CN V and CN VII inju-
radiosurgery. Some teams put forth the argument that patients ries.41 A series that would allow a proper evaluation of the qual-
with disabling imbalance may be more likely to improve with a ity of hearing outcome following linac-based radiosurgery is
resection associated with vestibular neurotomy. In our experi- still lacking. Several centers using linacs have stated their fail-
ence, the probability of improvement of vestibular symptoms is ure to obtain satisfactory safety results from single doses. The
not higher after MS as compared with SRS. use of hypofractionation or hyperfractionation was attempted
to improve their results.

154
Stereotactic Radiosurgery for Vestibular Schwannomas

A rare series of SRT with a rigorous methodology (i.e., more not deter a patient from the benefits gained as well as the dra-
than 3 years follow-up, minimum number of patients, pre- and matic reduction of risk from radiosurgery as compared with
postoperative PTA and SDS) show rates of CN V and CN VII pres- MS. Using the UK Registry of Causes of Death, Rowe et al com-
ervation and rate of functional hearing preservation that are piled a comparison of the risks of those having had radiosur-
fairly comparable to those of GKRS.42–48 We have found no ser- gery with those with no history of radiosurgery. 55 In their
ies, relying on audiometric criteria to show the capacity of SRT findings, they clearly demonstrate the absence of increased
to provide patients with rates of functional hearing preserva- incidences of cancer in patients who had received radiosur-
tion, comparable to those achievable with GKRS. Severe acute gery.55 According to Cahan et al, the criteria for carcinogenesis
sudden hearing losses following SRT have been reported.50 can be discussed only if the nature of the tumor is different to
Big tumors, Koos stage IV, with a significant mass effect on the nature of the treated tumor.57
the brainstem, require microsurgical decompression. However, The malignant evolution of benign schwannomas, which is
the radical removal of these very big tumors, even in the best extremely rare, is a different phenomenon to carcinogenesis
hands, can be associated with a major risk of facial palsy to the proper (i.e., radio-induced tumors). Within the literature, seven
extent of 50 to 60%. There are various degrees of severity, but cases of VSs, which were regarded as benign at the time of the
permanent cases are not uncommon. In our experience, in just first surgery, were found to be malignant at the point of second
55% of the patients involved, the radical removal of large Koos surgery. A key fact is that all of these patients had had a surgical
stage IV lesions is compatible with the long-term preservation resection, but only five patients had radiosurgical exposure. 58,
of normal facial motor nerve functions (House-Brackman grade 59,60 Comey et al put forward the hypothesis “that the tumours

1 or 2). Thus, 7 years ago we gave up this approach and initially regarded as benign, may already contain a minority of
switched to a combined approach as standard management for malignant components, which have subsequently taken over
big Koos stage IV cases. The first step of this combined approach the majority of the tumour, and led to the recurrence and
is the deliberate, partial removal of the tumor under neuromo- malignant behaviour.” This explanation is for patients receiving
nitoring of the motor facial nerve. several stand-alone MS procedures and in rare cases may
The retrosigmoid approach is used in the majority of cases. be valid for patients receiving several MS procedures and
The second step is carried out 3 to 6 months following the MS radiosurgery.61
procedure, with radiosurgery being used to treat the remnants. Statistics on the occurrence of facial palsy following radiosur-
This paradigm shift from radical removal to the combined gery show that in all the modern series of high-precision sin-
approach has increased our preservation rate of the motor gle-dose radiosurgeries, risks of facial palsy are less than 1%.
facial function, from 55% to 85%. Functional hearing, however, is This is far lower than that of the most favorable results from
rarely retained. MS, when performed in the very best hands.11–15
Stage IV tumors of a reasonable size (generally in patients Communicating hydrocephalus can classically be observed in
with small posterior fossa)51 with a modest mass effect on the patients with a vestibular schwannoma. Hydrocephalus is par-
brainstem can benefit from cautious radiosurgery, especially ticularly seen in older patients and in those with larger tumors.
when the patient’s hearing is still functional.52 The classical Roche et al62,63 have nicely demonstrated that there is no
phenomenon of transient increase of VS after radiosurgery53,54 increased risk of developing any radiological and clinical signs
might bring about a degradation of the patient’s neurologic of hydrocephalus through radiosurgery.
condition, thus making a resection mandatory.53 However, in In cases of failure, if the lesion size is still compatible with
our experience with 95 patients who had moderate Koos stage radiosurgery, a second radiosurgery is proposed. This, in our
IV at the time of radiosurgery, after years of follow-up, none experience, has a high probability of success.64 However, in
presented such a complication. The tumor control rate statistics these cases the risk of functional hearing loss is higher than in
remained at 95%, and the functional hearing preservation rate the first round of radiosurgery. When the tumor is too large for
was at 65%. No patients had facial palsy. a second radiosurgery procedure, we always propose subtotal
Young age is undoubtedly related to better clinical results, resection followed by repeat radiosurgery. We no longer pro-
especially in terms of hearing preservation.24,25 Preserving neu- pose the radical removal of a VS due to the significantly higher
rologic function and particularly motor facial function is of rate of facial palsy.65
utmost importance in young patients with a long life expect-
ancy, making radiosurgery the preferred surgical method for
this population group (▶ Fig. 19.2).
However, the risk of malignancy must be factored and dis-
19.5 Summary
cussed whenever younger patients are being treated with ioniz- Our series of 2,336 patients who underwent GKRS and were
ing radiation therapy. The most recent literature available to us, subsequently followed up and monitored for more than 3
directly pertaining to this issue, covers studies set to exception- years demonstrates a high rate of tumor control and the rarity
ally high standards.55,56 All these studies have been performed of toxicity with 0.5% trigeminal nerve injury and 0.5% facial
using evidence-based methodology. The consensus is that there palsy. Serviceable hearing preservation stands at 65% after 3
is no appreciable evidence of an increased risk of carcinogenesis years, but is highly dependent on the initial quality of hearing,
in patients who have received radiosurgery. past history of hearing loss, age, and dose to the cochlea. In
This means that if this risk actually exists, then it must be favorable cases, the rate of hearing preservation can be as high
very low, and should be weighed against the risk of periopera- as 90%. These results, when compared with the natural history
tive mortality from MS—statistics hover around 1 to 2%. These of these tumors, favor a protective effect of radiosurgery on
studies clearly demonstrate that the risk of neoplasia should hearing.

155
Stereotactic Radiosurgery for Vestibular Schwannomas

Fig. 19.2 Example of Gamma Knife (GK) radiosurgery in a young patient presenting with Koos class I vestibular schwannoma and normal hearing. At
more than 3 years after radiosurgery, the patient has retained serviceable hearing. This case illustrates the benefit of a proactive radiosurgical
approach in young patients with small lesions and high-quality hearing.

With more than 20 years’ experience in the field of clinical [5] Norén G. Gamma knife radiosurgery of acoustic neurinomas. A historic per-
spective. Neurochirurgie 2004; 50: 253–256
experiments in fractionated radiotherapy in acoustics, advan-
[6] Flickinger JC, Kondziolka D, Lunsford LD. Dose and diameter relationships for
tages over high-precision single-dose radiosurgery remain facial, trigeminal, and acoustic neuropathies following acoustic neuroma ra-
ambiguous. diosurgery. Radiother Oncol 1996; 41: 215–219
[7] Koos WT, Day JD, Matula C, Levy DI. Neurotopographic considerations in the
microsurgical treatment of small acoustic neurinomas. J Neurosurg 1998; 88:
References 506–512
[8] Ohata K, Nunta-aree S, Morino M, et al. Aetiology of delayed facial palsy after
[1] Cushing H. Intracranial Tumours: Notes upon a Series of 2000 Verified Cases vestibular schwannoma surgery: clinical data and hypothesis. Acta Neurochir
with Surgical Mortality Pertaining Thereto. Springfield, IL: C.C. Thomas; 1932 (Wien) 1998; 140: 913–917
[2] Dandy WE. An operation for the total extirpation of tumors in the cerebello- [9] Gardner G, Robertson JH. Hearing preservation in unilateral acoustic neuro-
pontine angle. A preliminary report. Johns Hopkins Hospital Bull 1922; 33: ma surgery. Ann Otol Rhinol Laryngol 1988; 97: 55–66
344–345 [10] House JW. Facial nerve grading systems. Laryngoscope 1983; 93: 1056–1069
[3] House WF. Transtemporal bone microsurgical removal of acoustic neuromas. [11] Pollock BE, Driscoll CL, Foote RL, et al. Patient outcomes after vestibular
Report of cases. Arch Otolaryngol 1964; 80: 617–667 schwannoma management: a prospective comparison of microsurgical resec-
[4] Leksell L. A note on the treatment of acoustic tumours. Acta Chir Scand 1971; tion and stereotactic radiosurgery. Neurosurgery 2006; 59: 77–85, discussion
137: 763–765 77–85

156
Stereotactic Radiosurgery for Vestibular Schwannomas

[12] Pollock BE, Lunsford LD, Kondziolka D, et al. Outcome analysis of acoustic [36] Chopra R, Kondziolka D, Niranjan A, Lunsford LD, Flickinger JC. Long-term fol-
neuroma management: a comparison of microsurgery and stereotactic radio- low-up of acoustic schwannoma radiosurgery with marginal tumor doses of
surgery. Neurosurgery 1995; 36: 215–224, discussion 224–229 12 to 13 Gy. Int J Radiat Oncol Biol Phys 2007; 68: 845–851
[13] Myrseth E, Møller P, Pedersen PH, Lund-Johansen M. Vestibular schwannoma: [37] Niranjan A, Mathieu D, Flickinger JC, Kondziolka D, Lunsford LD. Hearing
surgery or gamma knife radiosurgery? A prospective, nonrandomized study. preservation after intracanalicular vestibular schwannoma radiosurgery.
Neurosurgery 2009; 64: 654–661, discussion 661–663 Neurosurgery 2008; 63: 1054–1062, discussion 1062–1063
[14] Myrseth E, Møller P, Pedersen PH, Vassbotn FS, Wentzel-Larsen T, Lund-Jo- [38] Régis J, Tamura M, Delsanti C, Roche PH, Pellet W, Thomassin JM. Hearing
hansen M. Vestibular schwannomas: clinical results and quality of life after preservation in patients with unilateral vestibular schwannoma after gamma
microsurgery or gamma knife radiosurgery. Neurosurgery 2005; 56: 927– knife surgery. Prog Neurol Surg 2008; 21: 142–151
935, discussion 927–935 [39] Franzin A, Spatola G, Serra C, et al. Evaluation of hearing function after Gam-
[15] Régis J, Pellet W, Delsanti C, et al. Functional outcome after gamma knife sur- ma Knife surgery of vestibular schwannomas. Neurosurg Focus 2009; 27: E3
gery or microsurgery for vestibular schwannomas. J Neurosurg 2002; 97: [40] Kano H, Kondziolka D, Khan A, Flickinger JC, Lunsford LD. Predictors of hear-
1091–1100 ing preservation after stereotactic radiosurgery for acoustic neuroma. J Neu-
[16] Pollock BE. Vestibular schwannoma management: an evidence-based com- rosurg 2009; 111: 863–873
parison of stereotactic radiosurgery and microsurgical resection. Prog Neurol [41] Friedman WA, Bradshaw P, Myers A, Bova FJ. Linear accelerator radiosurgery
Surg 2008; 21: 222–227 for vestibular schwannomas. J Neurosurg 2006; 105: 657–661
[17] Sughrue ME, Yang I, Aranda D, et al. The natural history of untreated sporadic [42] Chan AW, Black P, Ojemann RG, et al. Stereotactic radiotherapy for vestibular
vestibular schwannomas: a comprehensive review of hearing outcomes. J schwannomas: favorable outcome with minimal toxicity. Neurosurgery
Neurosurg 2010; 112: 163–167 2005; 57: 60–70, discussion 60–70
[18] Stangerup SE, Caye-Thomasen P, Tos M, Thomsen J. The natural history of ves- [43] Chang SD, Gibbs IC, Sakamoto GT, Lee E, Oyelese A, Adler JR Jr. Staged stereo-
tibular schwannoma. Otol Neurotol 2006; 27: 547–552 tactic irradiation for acoustic neuroma. Neurosurgery 2005; 56: 1254–1261,
[19] Sughrue ME, Kane AJ, Kaur R, et al. A prospective study of hearing preserva- discussion 1261–1263
tion in untreated vestibular schwannomas. J Neurosurg 2011; 114: 381–385 [44] Combs SE, Volk S, Schulz-Ertner D, Huber PE, Thilmann C, Debus J. Manage-
[20] Régis J, Carron R, Park MC, et al. Wait-and-see strategy compared with proac- ment of acoustic neuromas with fractionated stereotactic radiotherapy
tive Gamma Knife surgery in patients with intracanalicular vestibular (FSRT): long-term results in 106 patients treated in a single institution. Int J
schwannomas. J Neurosurg 2010; 113 (Suppl): 105–111 Radiat Oncol Biol Phys 2005; 63: 75–81
[21] Bakkouri WE, Kania RE, Guichard JP, Lot G, Herman P, Huy PT. Conserva- [45] Maire JP, Huchet A, Milbeo Y, et al. Twenty years’ experience in the treatment
tive management of 386 cases of unilateral vestibular schwannoma: tu- of acoustic neuromas with fractionated radiotherapy: a review of 45 cases.
mor growth and consequences for treatment. J Neurosurg 2009; 110: Int J Radiat Oncol Biol Phys 2006; 66: 170–178
662–669 [46] Koh E-S, Millar BA, Ménard C, et al. Fractionated stereotactic radiotherapy for
[22] Yomo S, Carron R, Thomassin JM, Roche PH, Régis J. Longitudinal analysis of acoustic neuroma: single-institution experience at The Princess Margaret
hearing before and after radiosurgery for vestibular schwannoma. J Neuro- Hospital. Cancer 2007; 109: 1203–1210
surg 2012; 117: 877–885 [47] Thomas C, Di Maio S, Ma R, et al. Hearing preservation following fractionated
[23] Kondziolka D. Hearing after Gamma Knife surgery. J Neurosurg 2012; 117: stereotactic radiotherapy for vestibular schwannomas: prognostic implica-
874–875, discussion 875–876 tions of cochlear dose. J Neurosurg 2007; 107: 917–926
[24] Tamura M, Carron R, Yomo S, et al. Hearing preservation after gamma knife [48] Andrews DW, Werner-Wasik M, Den RB, et al. Toward dose optimization for
radiosurgery for vestibular schwannomas presenting with high-level hearing. fractionated stereotactic radiotherapy for acoustic neuromas: comparison of
Neurosurgery 2009; 64: 289–296, discussion 296 two dose cohorts. Int J Radiat Oncol Biol Phys 2009; 74: 419–426
[25] Lobato-Polo J, Kondziolka D, Zorro O, Kano H, Flickinger JC, Lunsford LD. Gam- [49] Hayashi M, Chernov MF, Lipski SM, et al. Do we really still need an open sur-
ma knife radiosurgery in younger patients with vestibular schwannomas. gery for treatment of patients with vestibular schwannomas? Acta Neurochir
Neurosurgery 2009; 65: 294–300, discussion 300–301 Suppl (Wien) 2013; 116: 25–36
[26] Régis J, Tamura M, Wikler D, Porcheron D, Levrier O. Radiosurgery: operative [50] Chang SD, Poen J, Hancock SL, Martin DP, Adler JR Jr. Acute hearing loss fol-
technique, pitfalls and tips. Prog Neurol Surg 2008; 21: 54–64 lowing fractionated stereotactic radiosurgery for acoustic neuroma. Report of
[27] van Eck AT, Horstmann GA. Increased preservation of functional hearing after two cases. J Neurosurg 1998; 89: 321–325
gamma knife surgery for vestibular schwannoma. J Neurosurg 2005; 102 [51] Roche PH, Robitail S, Régis J. Two- and three dimensional measures of vestib-
(Suppl): 204–206 ular schwannomas and posterior fossa—implications for the treatment. Acta
[28] Wowra B, Muacevic A, Jess-Hempen A, Hempel JM, Müller-Schunk S, Tonn JC. Neurochir (Wien) 2007; 149: 267–273, discussion 273
Outpatient gamma knife surgery for vestibular schwannoma: definition of [52] Roche PH, Robitail S, Pellet W, Devèze A, Thomassin JM, Régis J. [Results and
the therapeutic profile based on a 10-year experience. J Neurosurg 2005; 102 indications of gamma knife radiosurgery for large vestibular schwannomas].
(Suppl): 114–118 Neurochirurgie 2004; 50: 377–382
[29] Timmer FC, Hanssens PE, van Haren AE, et al. Gamma knife radiosurgery for [53] Nagano O, Serizawa T, Higuchi Y, et al. Tumor shrinkage of vestibular schwan-
vestibular schwannomas: results of hearing preservation in relation to the nomas after Gamma Knife surgery: results after more than 5 years of follow-
cochlear radiation dose. Laryngoscope 2009; 119: 1076–1081 up. J Neurosurg 2010; 113 (Suppl): 122–127
[30] Régis J, Delsanti C, Roche PH, Thomassin JM, Pellet W. [Functional outcomes [54] Delsanti C, Roche PH, Thomassin JM, Régis J. Morphological changes of vestib-
of radiosurgical treatment of vestibular schwannomas: 1000 successive cases ular schwannomas after radiosurgical treatment: pitfalls and diagnosis of
and review of the literature]. Neurochirurgie 2004; 50: 301–311 failure. Prog Neurol Surg 2008; 21: 93–97
[31] Chung WY, Liu KD, Shiau CY, et al. Gamma knife surgery for vestibular [55] Rowe J, Grainger A, Walton L, Silcocks P, Radatz M, Kemeny A. Risk of malig-
schwannoma: 10-year experience of 195 cases. J Neurosurg 2005; 102 nancy after gamma knife stereotactic radiosurgery. Neurosurgery 2007; 60:
(Suppl): 87–96 60–65, discussion 65–66
[32] Hasegawa T, Fujitani S, Katsumata S, Kida Y, Yoshimoto M, Koike J. Stereotac- [56] Ganz JC. Gamma knife radiosurgery and its possible relationship to malig-
tic radiosurgery for vestibular schwannomas: analysis of 317 patients fol- nancy: a review. J Neurosurg 2002; 97 (Suppl): 644–652
lowed more than 5 years. Neurosurgery 2005; 57: 257–265, discussion 257– [57] Cahan WG, Woodard HQ, et al. Sarcoma arising in irradiated bone; report of
265 11 cases. Cancer 1948; 1: 3–29
[33] Lunsford LD, Niranjan A, Flickinger JC, Maitz A, Kondziolka D. Radiosurgery of [58] Akamatsu Y, Murakami K, Watanabe M, Jokura H, Tominaga T. Malignant pe-
vestibular schwannomas: summary of experience in 829 cases. J Neurosurg ripheral nerve sheath tumor arising from benign vestibular schwannoma
2005; 102 (Suppl): 195–199 treated by gamma knife radiosurgery after two previous surgeries: a case re-
[34] Hudgins WR, Antes KJ, Herbert MA, et al. Control of growth of vestibular port with surgical and pathological observations. World Neurosurg 2010; 73:
schwannomas with low-dose Gamma Knife surgery. J Neurosurg 2006; 105 751–754
(Suppl): 154–160 [59] Murakami K, Jokura H, Kawagishi J, Watanabe M, Tominaga T. Development
[35] Liu D, Xu D, Zhang Z, Zhang Y, Zheng L. Long-term outcomes after Gamma of intratumoral cyst or extratumoral arachnoid cyst in intracranial schwan-
Knife surgery for vestibular schwannomas: a 10-year experience. J Neurosurg nomas following gamma knife radiosurgery. Acta Neurochir (Wien) 2011;
2006; 105 (Suppl): 149–153 153: 1201–1209

157
Stereotactic Radiosurgery for Vestibular Schwannomas

[60] Régis J, Muracciole X. Associated is not induced! World Neurosurg 2010; 73: [64] Yomo S, et al. Repeat gamma knife surgery for regrowth of vestibular
642–643 schwannomas. Neurosurgery 2009; 64: 48–54, discussion 54–55
[61] Comey CH, McLaughlin MR, Jho HD, Martinez AJ, Lunsford LD. Death from a [65] Fuentes S, Arkha Y, Pech-Gourg G, Grisoli F, Dufour H, Régis J. Management of
malignant cerebellopontine angle triton tumor despite stereotactic radiosur- large vestibular schwannomas by combined surgical resection and gamma
gery. Case report.[see comments] J Neurosurg 1998; 89: 653–658 knife radiosurgery. Prog Neurol Surg 2008; 21: 79–82
[62] Roche PH, Ribeiro T, Soumare O, Robitail S, Pellet W, Régis J. [Hydrocephalus [66] Hempel LU, Rathke C, Raja SJ, Renkawitz-Pohl R. In Drosophila, don juan and
and vestibular schwannomas treated by Gamma Knife radiosurgery]. Neuro- don juan like encode proteins of the spermatid nucleus and the flagellum and
chirurgie 2004; 50: 345–349 both are regulated at the transcriptional level by the TAF II80 cannonball
[63] Roche PH, Khalil M, Soumare O, Régis J. Hydrocephalus and vestibular while translational repression is achieved by distinct elements. Dev Dyn
schwannomas: considerations about the impact of gamma knife radiosur- 2006; 235: 1053–1064
gery. Prog Neurol Surg 2008; 21: 200–206

158
20 Stereotactic Radiosurgery for Trigeminal
Part V Neuralgia 160

Functional Indications 21 Stereotactic Radiosurgery in Movement


Disorders 168

22 Gamma Knife Radiosurgery for


Obsessive–Compulsive Disorder 177

23 Stereotactic Radiosurgery for


Hypothalamic Hamartomas 182

24 Stereotactic Radiosurgery for Epilepsy 193

V
Stereotactic Radiosurgery for Trigeminal Neuralgia

20 Stereotactic Radiosurgery for Trigeminal Neuralgia


Michael D. Chan and Stephen B. Tatter

Table 20.1 Burchiel classification scheme for facial pain


Key Points Burchiel pain type Clinical history

● Microvascular decompression is the surgical treatment Type I trigeminal neuralgia > 50% episodic pain
of choice for younger patients in good health with trige- Type II trigeminal neuralgia < 50% episodic pain
minal neuralgia because of the greater durability of pain
Trigeminal neuropathic pain Caused by unintentional trauma (e.
relief.
g., tooth extraction)
● Radiosurgery represents an excellent minimally invasive al-
ternative to microvascular decompression for patients who Trigeminal deafferentation pain Caused by intentional trauma (e.g.,
are older or have significant medical comorbidities. rhizotomy)
● A median pain response rate of 85% with a median durability Symptomatic trigeminal neuralgia Multiple sclerosis
of pain relief of 5 years can be expected from patients with
trigeminal neuralgia who undergo radiosurgery. Postherpetic neuralgia Herpes zoster outbreak in trigemi-
nal distribution
● A single isocenter delivering 70 to 90 Gy to the 100% isodose
line without any collimation or plugging would be considered Atypical facial pain Somatoform pain
a standard radiosurgical technique.
● Radiosurgery may be safely repeated at least once if neces-
sary for recurrent trigeminal neuralgia pain. Table 20.2 Barrow Neurologic Institute (BNI) Pain Intensity Scale
Pain score Degree of pain

BNI I Complete pain relief without


20.1 Introduction medications

BNI II Some pain, but not requiring


Trigeminal neuralgia is a severe paroxysmal facial pain
medications
located within the trigeminal distribution on the face. Also
known as tic douloureux, this condition has been described BNI III Some pain, but adequately con-
as a “suicide disease” because of the severity of the pain. The trolled with medications
age-adjusted incidence of trigeminal neuralgia has been BNI IV Some pain, but inadequately con-
reported to be greater in women (5.9 per 100,000) than men trolled with medications
(3.4 per 100,000).1
BNI V Continued severe pain or no pain
relief

20.2 Classification of Trigeminal


Neuralgia 20.3 Pathophysiology of
There are several facial pain etiologies that can closely
Trigeminal Neuralgia
resemble trigeminal neuralgia. As such, it is vital to obtain a The pathophysiology of idiopathic trigeminal neuralgia has
detailed clinical history as well as imaging to rule out etiol- been explained by what has become known as the vascular
ogies such as tumors or multiple sclerosis (MS). Classifica- hypothesis. This hypothesis explains the episodic pain syndro-
tion of facial pain based on clinical presentation can help to me of trigeminal neuralgia by the compression of the trigeminal
properly select patients for the appropriate interventions. nerve by a blood vessel,3 with the most common offending ves-
The most commonly accepted modern classification scheme sel being the superior cerebellar artery. It is also thought that
for facial pain is the Burchiel classification, 2 which is sum- the brain settles within the cranial vault with age, and thus cre-
marized in ▶ Table 20.1. Classical idiopathic trigeminal neu- ates a greater likelihood for such an interaction between a
ralgia is known as type I trigeminal neuralgia in the blood vessel and the trigeminal nerve in the more elderly
Burchiel classification. It is defined as pain that is episodic population.
at least 50% of the time. It can be located within any of the Other etiologies that can produce pain syndromes similar to
three divisions of the trigeminal nerve and is commonly idiopathic trigeminal neuralgia include MS, tumors of the skull
described as sharp, stabbing, or electrical shock-like in qual- base (e.g., meningioma, acoustic neuroma, metastatic disease),
ity. The intensity of pain has been described using the Bar- Charcot-Marie-Tooth disease, lyme disease, herpes zoster, trau-
row Neurologic Institute (BNI) pain scale. This scale is matic nerve injury, and somatoform pain disorders. The impor-
commonly used in the scientific literature to describe pain tance of the various etiologies of facial pain with regard to the
before and after an intervention, and to compare results use of stereotactic radiosurgery (SRS) is the fact that radiosurgi-
between multiple series. The BNI scale is summarized in cal management has a high rate of response for type I pain, but
▶ Table 20.2. an inferior response rate for some of the other etiologies. The

160
Stereotactic Radiosurgery for Trigeminal Neuralgia

risk of SRS-related toxicity, though low, is another reason to dif- the pain may remain in remission. It is common, however, for
ferentiate between the various causes of facial pain. Patients patients to become refractory to medical management over
with herpetic neuralgia and neuropathic pain from traumatic time, and these patients will commonly require surgical or
nerve injury are less likely to respond to radiosurgery. ablative management. Furthermore, some of the antiepileptics
will commonly have associated toxicities such as sedation, cog-
nitive changes, and ataxia, which if intolerable, may lead to the
20.4 Treatment Options need for surgery or radiosurgery. Carbamazepine, in particular,
can have a high rate of such toxicities. Oxcarbazepine and gaba-
The major treatment options for trigeminal neuralgia include pentin may have lower rates of toxicity. Tricyclic antidepres-
antiepileptic medications, microvascular decompression (MVD) sants such as amitriptyline (Elavil; AstraZeneca, London, UK)
surgery, percutaneous rhizotomy, and SRS. The proper treat- are often used in patients with advanced age when there is a
ment option for each patient is generally dependent on such concern for intolerable toxicity.
factors as the patient’s age, patient preference, type of facial
pain, medical comorbidities, severity of pain, and previous
treatments that have either succeeded or failed. A proposed Microvascular Decompression
management algorithm is depicted in ▶ Fig. 20.1. Microvascular decompression is a surgical technique involving
a craniotomy and decompression of the trigeminal nerve from
the offending blood vessel. Intraoperative insertion of an inert
20.4.1 Medical Management implant (generally Teflon) allows for prevention of recurrent
The first-line therapeutic option for newly diagnosed trigeminal vascular compression. The chief advantage of MVD is the fact
neuralgia is medical management. In general, antiepileptics are that the pain relief is durable and likely curative. In general, 70%
the most common type of medication used for trigeminal neu- of patients treated with MVD continue to be pain-free 10 to 20
ralgia, although tricyclic antidepressants, benzodiazepines, and years after the operation.4 Operative morbidity and mortality is
narcotics can also lead to pain relief in selected cases. The single generally quite low, but may rise after the age of 70, the age
most effective medication for trigeminal neuralgia is carbama- that noninvasive alternatives may have a greater therapeutic
zepine (Tegretol; Novartis International AG, Basel, Switzerland). ratio.5 An endoscopic microvascular decompression technique
Other medications that have reported responses include pheny- has also been described.6
toin (Dilantin; Pfizer, New York, NY), baclofen (Gablofen; CNS A prospective study conducted at the Mayo Clinic has looked
Pharmaceutics, St. Paul, MN), oxcarbazepine (Trileptal; Novartis at the cost-effectiveness of SRS versus MVD as a definitive treat-
International), gabapentin (Neurontin; Pfizer), and lamotrigine ment option for trigeminal neuralgia. In this study, MVD
(Lamictal; GlaxoSmithKline plc, Brentford, London, UK). appeared to have a higher cost in the near term, but for patients
Patients who have an initial response to medical management with longer life expectancies, it appeared to be the more cost-
can undergo a trial of withdrawal of medications over time as effective treatment option.7

Fig. 20.1 Management algorithm for trigeminal


neuralgia. IV, intravenous.

161
Stereotactic Radiosurgery for Trigeminal Neuralgia

Stereotactic Radiosurgery chapter, the following radiosurgical outcomes will refer specifi-
cally to Gamma Knife results, with the exception of the section
Stereotactic radiosurgery represents a noninvasive treatment specifically dedicated to linac approaches.
option for trigeminal neuralgia, with its main advantage being
its noninvasiveness and low morbidity rate. The great majority
of the data that exist for radiosurgical management of trigemi- 20.5.1 Quality-of-Life and Pain
nal neuralgia involves the use of the Gamma Knife, although Outcomes
there is an emerging literature for linear accelerator (linac)
Successful responses to SRS are generally measured by several
approaches. The major disadvantage of SRS is the limited dura-
end points: pain relief, ability to come off of medications, and
bility of its pain response. Most series have shown that the
improvement of quality of life. A prospective quality-of-life
median duration of radiosurgical response to be on the order of
study performed by a group from Marseille, France, found
5 years. Although SRS can be repeated, for younger patients
improvement in all quality-of-life parameters and that 58 of 83
even a second application of SRS may not remain effective
(70%) responders were able to come off of medications after
throughout their life span. Because of issues with normal tissue
SRS.9
tolerance, patients are generally limited to two radiosurgical
Several large retrospective series have also been reported in
applications over a lifetime. Elderly patients and those with sur-
the scientific literature for trigeminal neuralgia after definitive
gical contraindications such as severe cerebro- or cardiovascu-
SRS. The results of the largest series of patients undergoing
lar disease or bleeding diatheses may also be best treated with a
Gamma Knife radiosurgery (GKRS) for the treatment of trigemi-
radiosurgical approach.
nal neuralgia are summarized in ▶ Table 20.3. The series that
have been reported have shown quite similar results regarding
Percutaneous Rhizotomy pain response. In a series from Wake Forest, North Carolina,
Percutaneous ablative techniques have also had success in the Marshall et al reported a cohort of over 400 patients with trige-
treatment of trigeminal neuralgia. These techniques include minal neuralgia and reported an 86% initial response to pain
radiofrequency rhizotomy, glycerol rhizotomy, and balloon rhi- within 3 months.10
zotomy. The chief advantages of percutaneous ablative techni- Among the greater concerns for treatment with SRS is the
ques are that pain relief is immediate and that they often do not increasing possibility of pain relapse with increasing time after
require general anesthesia. Comparisons of the percutaneous treatment. Riesenburger et al reported that pain relapse after
procedures have suggested that they likely have very similar re- SRS is a time-dependent phenomenon. Marshall et al reported
sponse rates and durability of response. However, the likelihood a median durability of 4.9 years for patients with type I trigemi-
of persistent hypesthesia or anesthesia dolorosa may be higher nal neuralgia.10 Lucas et al have recently reported that the ini-
than what is seen with MVD or a radiosurgical approach.8 The tial successful response and ability to discontinue medications
durability of response for percutaneous techniques is limited was the dominant factor predicting durable pain relief after
and similar to results with SRS. Patients with need for immedi- SRS.11 Other series have found prior surgical intervention to
ate pain relief, but who are not candidates for MVD, may be best lead to decreased response duration after SRS.12
treated with percutaneous rhizotomy.

20.5.2 Factors that Affect Response


20.5 Radiosurgery Outcomes Several factors have been identified that affect the likelihood of
As the great majority of the data for radiosurgical outcomes treatment success for SRS in the treatment of trigeminal neural-
have come using the Gamma Knife unit, for the purpose of this gia. The development of postradiosurgical numbness has been

Table 20.3 Selected large series of Gamma Knife radiosurgery for trigeminal neuralgia
Institution N Median dose (Gy) Response rate Any toxicity

Pittsburgh13 503 80 89% 11%

Marseille40 497 85 91% 14%

Wake Forest10 448 90 86% 44%

Columbia24 293 75 76% 5%

Beaumont41 149 80 92% 25%

UVA42 136 80 90% 19%

Mayo43 117 90 85% 37%

Maryland12 112 75 81% 6%a

Brussels26 109 90 82% 38%

Spokane44 108 86 82% 19%


aSeries reported bothersome numbness only.

162
Stereotactic Radiosurgery for Trigeminal Neuralgia

identified as a major factor that predicts treatment success in proportionately more than sensory fibers.21 At higher doses,
multiple series.13 Prior surgery for trigeminal neuralgia,14 and necrosis is seen more commonly and may contribute to the res-
particularly radiofrequency ablation of the nerve,10 appears to ponse to SRS; 90 Gy appears to be the upper limit of the accept-
decrease the likelihood of treatment response. Régis et al able dose.10,20,22
showed a sequential decrease in response with every previous
procedure performed.9 Having evidence based on magnetic res-
onance imaging (MRI) of contact between a blood vessel and 20.6.2 Isocenter Location
the trigeminal nerve appears to predict better response after
The technical goal of trigeminal neuralgia SRS is to place a
SRS.15 The dose rate of the Gamma Knife sources16 does not
radiosurgical 4-mm isocenter onto the trigeminal nerve as it
appear to affect the response rate.
runs through the prepontine cistern. A sample radiosurgical
plan is demonstrated in ▶ Fig. 20.2. The rationale for placing the
20.5.3 Radiosurgical Complications isocenter within the prepontine cistern is that the nerve can be
well visualized on MRI in this area, and that the nerve is also
Trigeminal nerve dysfunction is the major possible toxicity in
surrounded by cerebrospinal fluid, allowing for the precise tar-
patients who have received SRS for trigeminal neuralgia. The
geting and sharp dose falloff (i.e., penumbra) beyond the nerve,
mechanism for such radiosurgical toxicity is damage to the sen-
minimizing the risk of damage to surrounding structures such
sory fibers within the trigeminal nerve. There have been several
as the brainstem and temporal lobe.
series that have reported higher rates of numbness in patients
There are several hypotheses on the target of radiation effect
who have received higher doses and those who have a greater
when trigeminal neuralgia is treated with SRS. The putative tar-
length of nerve treated. There has also been an association
get of radiation damage is important because of the implica-
between patients who experience postradiosurgical numbness
tions it has on the ideal isocenter location. Kondziolka et al
and the durability of radiosurgical treatment response.11 Other
have published that the dorsal root entry zone is more radio-
series have suggested that excellent pain relief responses can be
sensitive than more distal portions of the nerve because of the
achieved in the absence of trigeminal nerve dysfunction. 17
transition between more radiosensitive oligodendrocytes and
Recent data from the University of Pittsburgh (Pittsburgh, PA)
more resistant Schwann cells.23 This was supported by data
suggest that patients receiving gabapentin may have a lesser
from Columbia University (New York, NY), which demonstrated
risk of GKRS-induced numbness.18 More severe toxicities that
improved pain outcomes in patients with greater volumes of
occur following SRS include corneal anesthesia and anesthesia
brainstem receiving a dose of 15 Gy.24 A strategy for targeting
dolorosa. The likelihood of more severe toxicity is rare, with
the dorsal root entry zone places the isocenter such that the
anesthesia dolorosa rates reported to be less than 1%.10
50% isodose line is tangential to the brainstem. However, data
from multiple other series support equivalent pain relief while
20.6 Radiosurgical Targeting and targeting a more distal portion of the nerve such as the pars
triangularis.
Treatment Planning Modern targeting strategies include targeting the pars trian-
gularis and using the 20% isodose line to determine isocenter
20.6.1 Prescription Dose location. The rationale for targeting the pars triangularis is that
Doses delivered for SRS generally range between 70 and 90 Gy it is a relatively distal portion of the nerve, but allows targeting
prescribed to the isocenter. There has been one series in which of the entire nerve circumference before it diverges into multi-
patients were treated in the repeat setting in which pain ple branches. Other series have placed the 20% isodose line such
responses were seen at doses as low as 45 Gy prescribed to the that it is tangential to the brainstem surface. The rationale for
isocenter.19 Pollock et al reported results from the Mayo Clinic this approach is to constrain the brainstem surface to < 20 Gy, as
(Rochester, MN) in which patients were treated with either 70 this dose has been implicated in treatment-related numbness.
or 90 Gy. Patients in the 90 Gy cohort experienced a greater With this approach, the majority of the high-dose region is
degree of pain relief, but also had a greater degree of numb- within the pars triangularis.
ness.20 The mechanism of pain relief is believed to be focal axo- Several series have compared the two isocenter locations. A
nal degeneration of the trigeminal nerve that affects pain fibers study from Wake Forest assessed dose delivered to the

Fig. 20.2 Targeting for trigeminal neuralgia ra-


diosurgery in the up-front and repeat settings. (a)
A Gamma Knife plan in which the target is the
pars triangularis; the 20% isodose line is tangen-
tial to the brainstem. (b) A plan after which the
patient’s pain recurred several years later. The
target is now more proximal so as to avoid
isocenter overlap from the previous plan; the 50%
isodose line is tangential to the brainstem.

163
Stereotactic Radiosurgery for Trigeminal Neuralgia

brainstem surface and root entry zone and found that although risk of relapsing over time as compared with patients with type
a higher dose to the brainstem did not predict a greater pain re- I pain.29 There have been much more limited series assessing
sponse, patients with shorter nerves did have increased treat- the outcomes of patients with non-type I trigeminal neuralgia
ment efficacy. The authors concluded that a “target” structure after SRS. This has been a difficult population to assess because
of radiation damage did exist. A recent series of 99 patients of the heterogeneity of the population in general, and the non-
from the University of Virginia (Charlottesville, VA) compared standardized classification systems used by various institutions.
cohorts receiving 80 Gy in which patients with a proximal iso- Dhople et al published a series of 35 patients with atypical tri-
center (50% isodose line entering brainstem) experienced great- geminal neuralgia from the University of Maryland.12 In this
er durability of pain relief than those with a distal isocenter series, the authors included patients with type II pain (continu-
(20% isodose line tangential to the brainstem).10 ous) as well as patients with burning as opposed to lancinating
Optimal isocenter location remains controversial. Both the pain. There was a trend toward longer time before pain relief as
Wake Forest and University of Virginia data also demonstrate well as shorter duration of pain relief in patients with atypical
that greater doses to the brainstem led to greater degree of trigeminal neuralgia in this series.
posttreatment numbness. A series from Wake Forest University compared outcomes of
patients with type I and type II trigeminal neuralgia. 10 In this
series, there were 61 patients with type II trigeminal neuralgia
20.6.3 Use of Dose Modulation and 32 patients with atypical facial pain. Patients with type II
Several studies have been published on the use of channel trigeminal neuralgia and atypical facial pain both had decreased
blocking or multiple isocenters as a means of blocking dose initial response rates after SRS as well as a decreased durability
from the brainstem and delivering dose to a greater length of of pain relief. Median durability of pain relief was 4.9 years for
nerve. type I, 1.7 years for type II, and 0.7 years for atypical facial pain.
A randomized prospective study of 87 patients was con-
ducted by the University of Pittsburgh in which patients were
randomized to one versus two isocenters.25 The rationale for 20.7.2 Multiple Sclerosis–Related
such a study was to determine whether increased length of Trigeminal Neuralgia
nerve treated resulted in any difference in either efficacy or tox-
icity of radiosurgical treatment. Although there was no change Multiple sclerosis–related, also called symptomatic, trigeminal
detected in efficacy, the incidence of complications correlated neuralgia constitutes approximately 1% of patients with trige-
with the nerve length irradiated—greater with two versus one minal neuralgia-like symptoms. Symptoms may manifest as epi-
isocenter. sodic or continuous pain, and it does not appear that the nature
Another controversy regarding targeting and delivery of SRS of pain affects the success rate of the treatment. 30 The most
for trigeminal neuralgia involves the question of collimator important distinction in this population with regard to thera-
plugging. “Plugging” blocks a portion of the collimator to shape peutic options is the difference in pathophysiology of the pain.
the beam to incorporate a greater length of nerve and to Multiple sclerosis–related trigeminal neuralgia is due to a
decrease the amount of brainstem exposure. A study from Brus- demyelinating process within the trigeminal neuronal pathway,
sels, Belgium, showed that the addition of plugging, although which can commonly be detected on MRI. As such, microvascu-
modestly improved the response to Gamma Knife, caused a lar decompression is not considered an adequate treatment op-
greater degree of bothersome numbness.26 The authors con- tion, as it does not address the pathophysiology of the disease.
cluded that plugging should be avoided. Medical management is considered to be first-line therapy, as it
is for idiopathic trigeminal neuralgia. Surgical options such as
glycerol rhizotomy and SRS have also been reported. Because of
20.6.4 Effect of Dose Rate the relative rarity of symptomatic trigeminal neuralgia, avail-
able published evidence is limited to small, single-institution
Multiple retrospective analyses have investigated if the dose
retrospective series.30,31 In the largest of such, a study from the
rate of the cobalt sources affected the efficacy of treatment. It
University of Pittsburgh, 37 patients were treated with GKRS
has been hypothesized that lower dose rates may have wors-
using a dose range between 70 and 90 Gy.32 The authors
ened pain responses because of the possibility of tissue repair
reported that 36 of 37 patients reported BNI I–IIIb pain at some
within the nerve.27 However, a series from the University of
point in their course with 23 patients experiencing a BNI I pain
Pittsburgh recently demonstrated no differences in efficacy
score. Five percent of patients experienced a new-onset pares-
over the span of a 2 half-life decay of cobalt-60 sources.28
thesia in this series.

20.7 Special Populations


20.7.3 Bilateral Trigeminal Neuralgia
20.7.1 Type II Trigeminal Neuralgia Bilateral trigeminal neuralgia is a complicated clinical entity
Several reports have demonstrated a decreased response rate that represents approximately 2% of patients with trigeminal
and durability of response in patients with type II trigeminal neuralgia. The etiology of bilateral trigeminal neuralgia is com-
neuralgia when treated with surgical or radiosurgical modal- monly related to either Charcot-Marie-Tooth disease or MS.33 It
ities. Tyler-Kabara et al showed that in series of 2,264 patients has been shown that patients with bilateral trigeminal neural-
with trigeminal neuralgia, those with type II pain had a greater gia are less likely to have blood vessel compression on MRI, 15

164
Stereotactic Radiosurgery for Trigeminal Neuralgia

and thus likely that a proportion of these patients have pain neuralgia as opposed to a consequence of previous SRS such as
that is not caused by vascular compression. The clinical com- deafferentation pain. Furthermore, a common practice is to
plexity of the bilateral pain is related to the possibility of bilat- select patients who had a prior response to SRS.36
eral trigeminal nerve dysfunction that can result from an It is common to prescribe a slightly lower dose at the second
ablative treatment. Such dysfunction can lead to significant tox- radiosurgical application. Doses as high as 90 Gy3and as low as
icity such as dysarthria and dysphagia resulting from damage to 45 Gy19 prescribed to the 100% isodose line have been shown to
the motor pathways of the trigeminal nerve. The efficacy of yield pain responses with the second radiosurgical treatment. 19
MVD is controversial given the difference in pathophysiology of Because the cumulative numbers in the literature are small, it is
the patients with bilateral pain. Stereotactic radiosurgery for unclear as to whether the likelihood of significant toxicity is
bilateral trigeminal neuralgia has been reported in an eight-pa- any worse than after the first radiosurgery.
tient series from Tufts Medical Center in Boston, Massachusetts, With regards to the isocenter location in the setting of repeat
without significant toxicity.34 However, long-term efficacy radiosurgery, data exist for placement of the isocenter in the
remains to be reported. One approach to avoid bilateral trige- exact same position, or in a different position to avoid over-
minal nerve dysfunction has been to treat the more sympto- lap.35 One common strategy has been to place the first isocenter
matic side first, then follow the patient for 6 to 12 months to distally at the first radiosurgical treatment, with the later iso-
assess for efficacy and toxicity before deciding on the manage- center being placed more proximally where the change from
ment of the other side. oligodendrocytes to Schwann cells may make the region more
radiosensitive.

20.8 Specific Clinical Scenarios


20.8.2 Use of Computed Tomography
20.8.1 Repeat Radiosurgery Alone for Treatment Planning
Because the median durability of pain relief after SRS is on There exists a population of patients with trigeminal neural-
the order of 5 years, some patients receiving primary SRS will gia who are candidates for radiosurgical management, but
have a recurrence of trigeminal neuralgia pain at some point have contraindications to MRI such as pacemaker placement,
in their lifetime. In this scenario, a second application of SRS a ferromagnetic implant, or shrapnel exposure. In such
is a reasonable treatment option. Several institutions have patients, computed tomography (CT)-based treatment plan-
now reported on the efficacy of repeat SRS and found that ning with or without cisternography has been reported in
the response rate and durability of a second response are which the targeting of the nerve is done based on anatomical
similar to what is seen with the first application. 35 Select landmarks such as the trigeminal impression on the temporal
results of repeat radiosurgical series are presented in bone.37,38 Further follow-up is likely necessary to ensure that
▶ Table 20.4. this population does not have a higher rate of late toxicity or
Patient selection is an important issue in patients who are late pain recurrence.
considered for a second radiosurgical procedure. A detailed
history is necessary to rule out the possibility that the pain the
patient is experiencing is truly a recurrence of trigeminal 20.8.3 Linear Accelerator–Based
Approaches
Table 20.4 Repeat radiosurgery series for relapsed trigeminal neuralgia Linear accelerator–based approaches for trigeminal neuralgia
Institution N Median re- Response Any toxicity are utilized less than GKSRS for several reasons: difficulty in
treatment rate accurately characterizing the output factor for a 4-mm collima-
dose (Gy) tor, the instability associated with a linac gantry, and the fact
that inaccuracies are cumulative. The potential magnitude of
Pittsburgh36 119 70 87% 21%
error (i.e., inaccuracy) estimated for linac treatment of trigemi-
Columbia45 45 40 62% 13% nal neuralgia is thought to be as great as 30%. For sufficient
treatment of trigeminal neuralgia on a linear accelerator using a
Wake For- 37 84 84% 57%
est35 4-mm collimator, a root mean square value of all errors likely
needs to be < 1 mm.39
Tangdu Hos- 34 71 97% 12% The largest series of linac-based SRS comes from the Univer-
pital (China)
46
sity of California, Los Angeles. In this series of 179 patients, the
authors demonstrated a response rate and durability of treat-
Tufts19 27 45 86% 29% ment similar to that which is seen with Gamma Knife series.22
The authors used a median dose of 90 Gy (range 70–90 Gy) with
Medical Uni- 22 74 100% 74%
versity of the 30% isodose line tangential to the pons.
Graz (Aus-
tria)47
20.9 Summary
Mayo48 19 76 95% 21%
Stereotactic radiosurgery represents a safe and effective nonin-
Maryland49 18 70 78% 11%
vasive treatment option for trigeminal neuralgia. The major

165
Stereotactic Radiosurgery for Trigeminal Neuralgia

limitation of SRS lies in its limited durability as compared with [21] Kondziolka D, Lacomis D, Niranjan A, et al. Histological effects of trigeminal
nerve radiosurgery in a primate model: implications for trigeminal neuralgia
MVD. Patients older than 70 years, with multiple sclerosis, or
radiosurgery. Neurosurgery 2000; 46: 971–976, discussion 976–977
with significant medical comorbidities represent populations [22] Smith ZA, Gorgulho AA, Bezrukiy N, et al. Dedicated linear accelerator radio-
that may be well suited for SRS. Patients are generally best surgery for trigeminal neuralgia: a single-center experience in 179 patients
managed by a multidisciplinary team to determine which treat- with varied dose prescriptions and treatment plans. Int J Radiat Oncol Biol
ment option is optimal for each patient. Phys 2011; 81: 225–231
[23] Kondziolka D, Lunsford LD, Flickinger JC, et al. Stereotactic radiosurgery for
trigeminal neuralgia: a multiinstitutional study using the gamma unit. J Neu-

References rosurg 1996; 84: 940–945


[24] Brisman R, Mooij R. Gamma knife radiosurgery for trigeminal neuralgia:
dose-volume histograms of the brainstem and trigeminal nerve. J Neurosurg
[1] Katusic S, Beard CM, Bergstralh E, Kurland LT. Incidence and clinical features
2000; 93 (Suppl 3): 155–158
of trigeminal neuralgia, Rochester, Minnesota, 1945–1984. Ann Neurol 1990;
[25] Flickinger JC, Pollock BE, Kondziolka D, et al. Does increased nerve length
27: 89–95
within the treatment volume improve trigeminal neuralgia radiosurgery? A
[2] Burchiel KJ. A new classification for facial pain. Neurosurgery 2003; 53:
prospective double-blind, randomized study. Int J Radiat Oncol Biol Phys
1164–1166, discussion 1166–1167
2001; 51: 449–454
[3] Jannetta PJ. Arterial compression of the trigeminal nerve at the pons in pa-
[26] Massager N, Nissim O, Murata N, et al. Effect of beam channel plugging on the
tients with trigeminal neuralgia. J Neurosurg 1967; 26 (Suppl): 159–162
outcome of gamma knife radiosurgery for trigeminal neuralgia. Int J Radiat
[4] Barker FG II, Jannetta PJ, Bissonette DJ, Larkins MV, Jho HD. The long-term
Oncol Biol Phys 2006; 65: 1200–1205
outcome of microvascular decompression for trigeminal neuralgia. N Engl J
[27] Stieber V, Robbins M, Balamucki C, et al. Determination of a clinical value for
Med 1996; 334: 1077–1083
the repair half-time (T1/2) of the trigeminal nerve based on outcome data
[5] Kalkanis SN, Eskandar EN, Carter BS, Barker FG II. Microvascular decompres-
from gamma knife radiosurgery for facial pain. Radiat Res 2007; 168: 143–
sion surgery in the United States, 1996 to 2000: mortality rates, morbidity
148
rates, and the effects of hospital and surgeon volumes. Neurosurgery 2003;
[28] Arai Y, Kano H, Lunsford LD, et al. Does the Gamma Knife dose rate affect out-
52: 1251–1261, discussion 1261–1262
comes in radiosurgery for trigeminal neuralgia? J Neurosurg 2010; 113
[6] Halpern CH, Lang SS, Lee JY. Fully endoscopic microvascular decompression:
(Suppl): 168–171
our early experience. Minim Invasive Surg 2013; 2013: 739432
[29] Tyler-Kabara EC, Kassam AB, Horowitz MH, et al. Predictors of outcome in
[7] Pollock BE, Ecker RD. A prospective cost-effectiveness study of trigeminal
surgically managed patients with typical and atypical trigeminal neuralgia:
neuralgia surgery. Clin J Pain 2005; 21: 317–322
comparison of results following microvascular decompression. J Neurosurg
[8] Burchiel KJ, Steege TD, Howe JF, Loeser JD. Comparison of percutaneous radio-
2002; 96: 527–531
frequency gangliolysis and microvascular decompression for the surgical ma-
[30] Weller M, Marshall K, Lovato JF, et al. Single-institution retrospective series of
nagement of tic douloureux. Neurosurgery 1981; 9: 111–119
gamma knife radiosurgery in the treatment of multiple sclerosis-related tri-
[9] Régis J, Metellus P, Hayashi M, Roussel P, Donnet A, Bille-Turc F. Prospective
geminal neuralgia: factors that predict efficacy. Stereotact Funct Neurosurg
controlled trial of gamma knife surgery for essential trigeminal neuralgia. J
2014; 92: 53–58
Neurosurg 2006; 104: 913–924
[31] Rogers CL, Shetter AG, Ponce FA, Fiedler JA, Smith KA, Speiser BL. Gamma
[10] Marshall K, Chan MD, McCoy TP, et al. Predictive variables for the successful
knife radiosurgery for trigeminal neuralgia associated with multiple sclerosis.
treatment of trigeminal neuralgia with gamma knife radiosurgery. Neurosur-
J Neurosurg 2002; 97 (Suppl): 529–532
gery 2012; 70: 566–572, discussion 572–573
[32] Zorro O, Lobato-Polo J, Kano H, Flickinger JC, Lunsford LD, Kondziolka D. Gam-
[11] Lucas JT Jr, Nida AM, Isom S, et al. Predictive nomogram for the durability of
ma knife radiosurgery for multiple sclerosis-related trigeminal neuralgia.
pain relief from gamma knife radiation surgery in the treatment of trigeminal
Neurology 2009; 73: 1149–1154
neuralgia. Int J Radiat Oncol Biol Phys 2014; 89: 120–126
[33] Tacconi L, Miles JB. Bilateral trigeminal neuralgia: a therapeutic dilemma. Br J
[12] Dhople AA, Adams JR, Maggio WW, Naqvi SA, Regine WF, Kwok Y. Long-term
Neurosurg 2000; 14: 33–39
outcomes of Gamma Knife radiosurgery for classic trigeminal neuralgia: im-
[34] Wu JK, Raval A, Salluzzo J, et al. Results of bilateral trigeminal neuralgia
plications of treatment and critical review of the literature. Clinical article. J
treated with gamma knife radiosurgery: Boston Gamma Knife Center experi-
Neurosurg 2009; 111: 351–358
ence. Paper presented at: 16th Meeting of the Leksell Gamma Knife Society;
[13] Kondziolka D, Zorro O, Lobato-Polo J, et al. Gamma Knife stereotactic radio-
March 25–29, 2012; Sydney, Australia
surgery for idiopathic trigeminal neuralgia. J Neurosurg 2010; 112: 758–765
[35] Aubuchon AC, Chan MD, Lovato JF, et al. Repeat gamma knife radiosurgery for
[14] Maesawa S, Salame C, Flickinger JC, Pirris S, Kondziolka D, Lunsford LD. Clini-
trigeminal neuralgia. Int J Radiat Oncol Biol Phys 2011; 81: 1059–1065
cal outcomes after stereotactic radiosurgery for idiopathic trigeminal neural-
[36] Park KJ, Kondziolka D, Berkowitz O, et al. Repeat gamma knife radiosurgery
gia. J Neurosurg 2001; 94: 14–20
for trigeminal neuralgia. Neurosurgery 2012; 70: 295–305, discussion 305
[15] Brisman R, Khandji AG, Mooij RB. Trigeminal nerve-blood vessel relationship
[37] Attia A, Tatter SB, Weller M, et al. CT-only planning for Gamma Knife radio-
as revealed by high-resolution magnetic resonance imaging and its effect on
surgery in the treatment of trigeminal neuralgia: methodology and outcomes
pain relief after gamma knife radiosurgery for trigeminal neuralgia. Neuro-
from a single institution. J Med Imaging Radiat Oncol 2012; 56: 490–494
surgery 2002; 50: 1261–1266, discussion 1266–1267
[38] Park KJ, Kano H, Berkowitz O, et al. Computed tomography-guided gamma
[16] Balamucki CJ, Stieber VW, Ellis TL, et al. Does dose rate affect efficacy? The
knife stereotactic radiosurgery for trigeminal neuralgia. Acta Neurochir
outcomes of 256 gamma knife surgery procedures for trigeminal neuralgia
(Wien) 2011; 153: 1601–1609
and other types of facial pain as they relate to the half-life of cobalt. J Neuro-
[39] Rahimian J, Chen JC, Rao AA, Girvigian MR, Miller MJ, Greathouse HE. Geo-
surg 2006; 105: 730–735
metrical accuracy of the Novalis stereotactic radiosurgery system for trigemi-
[17] Massager N, Lorenzoni J, Devriendt D, Desmedt F, Brotchi J, Levivier M. Gamma
nal neuralgia. J Neurosurg 2004; 101 (Suppl 3): 351–355
knife surgery for idiopathic trigeminal neuralgia performed using a far-anterior
[40] Tuleasca C, Carron R, Resseguier N, Donnet A, Roussel P, Gaudart J, Levivier M,
cisternal target and a high dose of radiation. J Neurosurg 2004; 100: 597–605
Régis J. Patterns of pain-free response in 497 cases of classic trigeminal neu-
[18] Flickinger JC Jr, Kim H, Kano H, et al. Do carbamazepine, gabapentin, or other
ralgia treated with Gamma Knife surgery and followed up for least 1 year. J
anticonvulsants exert sufficient radioprotective effects to alter responses
Neurosurg 2012; 117 (Suppl): 181–188
from trigeminal neuralgia radiosurgery? Int J Radiat Oncol Biol Phys 2012;
[41] Baschnagel AM, Cartier JL, Dreyer J, Chen PY, Pieper DR, Olson RE, Krauss DJ,
83: e501–e506
Maitz AH, Grills IS. Trigeminal neuralgia pain relief after gamma knife stereo-
[19] Dvorak T, Finn A, Price LL, et al. Retreatment of trigeminal neuralgia with
tactic radiosurgery. Clin Neurol Neurosurg 2014; 117: 107–111
Gamma Knife radiosurgery: is there an appropriate cumulative dose? Clinical
[42] Sheehan J, Pan HC, Stroila M, Steiner L. Gamma knife surgery for trigeminal
article. J Neurosurg 2009; 111: 359–364
neuralgia: outcomes and prognostic factors. J Neurosurg 2005; 102: 434–441
[20] Pollock BE, Phuong LK, Foote RL, Stafford SL, Gorman DA. High-dose trigemi-
[43] Pollock BE, Phuong LK, Gorman DA, Foote RL, Stafford SL. Stereotactic radio-
nal neuralgia radiosurgery associated with increased risk of trigeminal nerve
surgery for idiopathic trigeminal neuralgia. J Neurosurg 2002; 97: 347–353
dysfunction. Neurosurgery 2001; 49: 58–62, discussion 62–64

166
Stereotactic Radiosurgery for Trigeminal Neuralgia

[44] Elaimy AL, Lamm AF, Demakas JJ, Mackay AR, Lamoreaux WT, Fairbanks RK, [47] Gellner V, Kurschel S, Kreil W, Holl EM, Ofner-Kopeinig P, Unger F. Recurrent
Pfeffer RD, Cooke BS, Peressini BJ, Lee CM. Gamma knife radiosurgery for typ- trigeminal neuralgia: long term outcome of repeat gamma knife radiosur-
ical trigeminal neuralgia: An institutional review of 108 patients. Surg Neurol gery. J Neurol Neurosurg Psychiatry 2008; 79: 1405–1407
Int 2013; 4: 92 [48] Pollock BE, Foote RL, Link MJ, Stafford SL, Brown PD, Schomberg PJ. Repeat
[45] Brisman R. Repeat gamma knife radiosurgery for trigeminal neuralgia. Ster- radiosurgery for idiopathic trigeminal neuralgia. Int J Radiat Oncol Biol Phys
eotact Funct Neurosurg 2003; 81: 43–49 2005; 61: 192–195
[46] Wang L, Zhao ZW, Qin HZ, Li WT, Zhang H, Zong JH, Deng JP, Gao GD. Repeat [49] Herman JM, Petit JH, Amin P, Kwok Y, Dutta PR, Chin LS. Repeat gamma knife
gamma knife radiosurgery for recurrent or refractory trigeminal neuralgia. radiosurgery for refractory or recurrent trigeminal neuralgia: treatment out-
Neurol India 2008; 56: 36–41 comes and quality-of-life assessment. Int J Radiat Oncol Biol Phys 2004; 59:
112–116

167
Stereotactic Radiosurgery in Movement Disorders

21 Stereotactic Radiosurgery in Movement Disorders


Patrick Pezeshkian, Antonio De Salles, and Nader Pouratian

21.1 Introduction
Key Points
Despite initially being innovated for the purpose of treating
1. Radiosurgical thalamotomy functional disorders such as Parkinson disease (PD) and psychi-
● Stereotactic radiosurgery thalamotomy targets the atric disease, stereotactic radiosurgery (SRS) is now mostly
nucleus ventralis intermedius and is currently based on used to treat brain tumors and vascular lesions. Lesional techni-
anatomical magnetic resonance imaging alone and meas- ques, such as radiosurgery, have been progressively supplanted
urements relative to anterior and posterior commissures by technologies such as deep brain stimulation (DBS), which
(indirect targeting). We prefer a maximum dose of 140 Gy provide a reversible and modulatable treatment. Still there is
delivered with one 4-mm isocenter. considerable interest in the role of lesioning surgery, which
● The efficacy of stereotactic radiosurgery thalamotomy for when performed effectively obviates the need for hardware im-
controlling tremor is similar to that reported for radiofre- plantation and long-term maintenance, thereby significantly
quency thalamotomy and deep brain stimulation, with 85 reducing health care costs. Stereotactic radiosurgery in particu-
to 90% of patients showing significant improvements in lar is a very attractive treatment modality because it provides a
tremor in short-term follow-up studies. precise and noninvasive means of lesioning the brain to achieve
● Complications may arise from lesions that develop larger functional improvement. However, given the irreversible nature
than expected or from misplaced lesions. These include of the lesion and given the lack of precise and reliable means of
involvement of the internal capsule or ventral posterior neurophysiological testing and confirming the physiological
thalamic nucleus with associated deficits of weakness or target, SRS is generally reserved as an option for patients who
paralysis, dysphasia, or sensory loss. Four to five percent are not suitable for open neurosurgical procedures.
may experience side effects, but gradually recover over Advances in neuroimaging, computerized dosimetry, treat-
several months. ment-planning software systems, and the promising results of
2. Radiosurgical pallidotomy SRS in other brain diseases have reinvigorated interest in the
● Stereotactic radiosurgery pallidotomy uses indirect target- application of this technique in the treatment of movement dis-
ing of the globus pallidus internus with the isocenter orders since the 1990s. The role of lesioning in the treatment of
being placed such that 50% isodose line of a 4-mm colli- movement disorders is well documented in the radiofrequency
mator is within the globus pallidus internus and superior (RF) ablation literature, lending promise to the application of
to the optic tract. We prefer a radiosurgical dose of SRS for the treatment of movement disorders.1–8 However, the
140 Gy for creating a brain lesion with a 4-mm collimator. transition from RF lesions to noninvasive SRS lesions required
● The most consistent effect of pallidal ablation is relief of addressing additional questions, such as those related to dosing.
contralateral levodopa-induced dyskinesias. However, the Initially, the safety and effectiveness of the dose used for func-
amount of improvement in contralateral bradykinesia, tional radiosurgery was not known. Previous reports indicated
rigidity, and tremor varies among many studies. that doses of 180 to 200 Gy were capable of and necessary for
● Complications include visual field deficit, speech, and creating a focal lesion in the brain, but were associated with
swallowing difficulties, worsening of gait, and numbness potential complications such as uncontrolled lesion size and
in the contralateral hemibody. perilesional edema.9,10 These initial high doses are now
3. Radiosurgical subthalamotomy reduced, with a tendency toward fewer complications. Collima-
● Stereotactic radiosurgery subthalamotomy is rarely per- tor size and number of isocenters have also been a matter
formed. To our knowledge, the only report in the litera- of concern because the use of multiple isocenters and large-size
ture is a case report of a radiosurgical lesioning of the collimators have been related to increased complication
subthalamic nucleus with a single 120 Gy 4-mm shot rates.11–15
using indirect targeting. In this chapter, we present a broad review of the usefulness
● Outcomes are difficult to ascertain due to the rarity of of SRS for the treatment of movement disorders—its indica-
the procedure; however, the radiofrequency subthala- tions, techniques, applications, and complications.
motomy literature, albeit small, points to 45 to 60%
improvement in Unified Parkinson’s Disease Rating
Scale motor scores in the “off” period in the 16- to 24- 21.2 Definition, Patient Selection,
month follow-up.
● Given the paucity of stereotactic radiosurgery subthala-
and Indications
motomies, the main complication associated with subtha- Stereotactic radiosurgery has been applied for the treatment of
lamotomies, virtually all of which are reports of radiofre- such movement disorders as PD, essential tremor (ET), and dys-
quency subthalamotomies, is contralateral hemiballism, tonia. Before proceeding with a discussion of treatment and
which tends to resolve spontaneously within several outcomes, a brief definition of PD, ET, and dystonia is presented,
months. including patient selection criteria for neurosurgical interven-
tion and indications for intervention.

168
Stereotactic Radiosurgery in Movement Disorders

Parkinson disease is a progressive neurodegenerative disor- testing should also be performed because surgical interven-
der resulting in motor symptoms that are related to the death tion, as well as radiosurgical intervention, is generally not
of dopaminergic cells in the substantia nigra (SN). Early in the considered appropriate for patients with significant demen-
course of the disease, motor symptoms dominate and include tia.1,2,4,5,15–22
resting tremor, rigidity, axial and appendicular bradykinesia, Because neurophysiology-guided RF stereotactic surgery or
and difficulty with walking and gait. Later, cognitive and behav- DBS offer advantages over SRS and the permanent neurologic
ioral problems may arise, with dementia commonly occurring complication rate of SRS is worse than that of DBS (see discus-
in the advanced stages of the disease, whereas depression is the sion below), SRS is reserved for a small subset of patients who
most common psychiatric symptom. Other symptoms include have medical conditions that make them unacceptable candi-
sensory, sleep, and emotional problems. The Unified Parkin- dates for invasive stereotactic neurosurgical intervention, or
son’s Disease Rating Scale (UPDRS) is a scale that was developed require therapy but absolutely refuse invasive surgery under
as an effort to incorporate elements from existing scales to pro- any circumstances. These include elderly patients, patients suf-
vide a comprehensive, but efficient and flexible means to mon- fering from severe cardiac or pulmonary comorbidities, and
itor PD-related disability and impairment. The UPDRS itself has those who are anticoagulant dependent. In some cases, some
four components, largely derived from preexisting scales that patients may prefer SRS to avoid an invasive surgical proce-
were reviewed and modified by a consortium of movement- dure.15,17,19,20,21,23,24 Furthermore, invasive stereotactic surgery,
disorder specialists. These include Part I, Mentation, Behavior including both RF ablation and DBS, is associated with certain
and Mood; Part II, Activities of Daily Living; Part III, Motor; and morbidities and possible mortality. These procedures carry an
Part IV, Complications. The UPDRS scores are generally assessed inherent risk of intracerebral hemorrhage (1–2%), infection (4–
both off- and on-medication states. As patients with PD are 5%), seizures, brain displacement, tension pneumocephalus,
evaluated by a response to levodopa, PD patients should have at and direct injury from probe placement, among others.8,24,25,26
least a 30% decrease in UPDRS III scores in response to Stereotactic radiosurgery is a less invasive procedure that does
levodopa. not involve opening of the cranium or incisions; therefore,
Essential tremor is the most common movement disorder, there is no risk of hemorrhage or meningitis from postoperative
typically resulting in tremor of the arms, hands, or fingers, but infection. The postoperative patient care is also simpler;
sometimes involving the head or other body parts during vol- patients can return earlier to their regular activities with a
untary movements such as eating and writing. Essential tremor reduction in hospitalization time. The disadvantages of SRS
generally presents as a rhythmic tremor (4–12 Hz) that occurs include irreversibility of the procedure because it is an ablative
only when the affected muscle is exerting effort. Essential trem- procedure and often limited to unilateral application, variability
or is distinguished from PD by the postural and kinetic charac- in efficacy, and complications based on treatment technique
teristics of the tremor rather than the rest tremor associated such as dosage of radiation used, number of isocenters used,
with PD. Moreover, PD is marked by bradykinesia and rigidity, and collimator size. Furthermore, the exclusive reliance on ana-
whereas patients with ET do not have such motor symptoms. tomical targeting due to the impossibility of confirming the le-
Some individuals may have both conditions. The patient’s trem- sion site intraoperatively with electrophysiologic testing makes
or is evaluated with a standardized Tremor Rating Scale. The target determination less certain. Moreover, a mean delay of 6
scale describes the distribution and severity of the tremor at months for clinical improvement may be expected after SRS.15,
rest, with maintenance of a sustained posture and with action. 17,18,19,20,21,24

Because of the fluctuating nature of tremor severity and the


effects of antitremor drugs, repeated assessments that take into
account whether the patient is on or off drugs are most 21.3 Radiosurgical Thalamotomy
informative.
Dystonia is a neurologic movement disorder, in which sus- Stereotactic radiosurgery thalamotomy targeting the nucleus
tained muscle contractions cause twisting and repetitive move- ventralis intermedius (VIM) has been performed by several
ments or abnormal postures. The disorder may be hereditary or authors in the treatment of tremor in patients with tremor-
caused by other factors such as birth-related or other physical dominant PD, ET, and tremor related to multiple sclerosis, trau-
trauma, infection, poisoning (e.g., lead poisoning), or reaction ma, or other causes. The reported results and complications
to pharmaceutical drugs, particularly neuroleptics. with SRS thalamotomy are comparable to those achieved using
Patients are selected for SRS for the management of move- other methods.15,17,19,21,23,27,28,29
ment disorders in the same manner as for procedures such as
RF thalamotomy, pallidotomy, or DBS. Patients should be
assessed in a multidisciplinary movement disorder clinic to
21.3.1 Technique and Targeting
confirm the underlying diagnosis, in particular to exclude “Par- Currently targeting is based on anatomical magnetic resonance
kinson’s plus” syndromes such as multisystem atrophy, pro- imaging (MRI) alone and measurements relative to the anterior
gressive supranuclear palsy, and striatonigral degeneration. It is (AC) and posterior (PC) commissures—“indirect targeting.” Con-
important to ensure that adequate pharmacological response sequently, every effort must be made to eliminate or correct
has been documented and adequate treatment has been pro- sources of MRI distortion when calculating the stereotactic tar-
vided before any intervention is considered. Quantifying symp- get coordinates. The current technique for targeting the VIM
tom severity with the standard scoring systems, such as the thalamus is similar to conventional stereotactic procedures. In
UPDRS and Tremor Rating Scale, is important to define severity our institution, a frameless approach using the Novalis linear
of the disease and guide surgical timing. Neuropsychological accelerator (linac) system (Brainlab, Munich, Germany) is used.

169
Stereotactic Radiosurgery in Movement Disorders

High-resolution MR images are obtained with a 3.0-T scanner. thalamic nucleus is represented more constantly by the per-
Contrast-enhanced images are acquired, including through the centage of thalamic length than by the distance from the PC.
basal ganglia, midbrain, third ventricle, and AC and PC (1-mm Therefore, the regular coordinates for thalamotomy can be
isotropic resolution). Axial fast inversion recovery sequences adjusted for the VIM nucleus to be at a length of 45% from the
are performed for optimal gray–white differentiation and iden- anterior tip of the thalamus in the horizontal plane.31 Recent
tification of the internal capsule (0.9-mm in-plane resolution). developments in MRI diffusion tensor and colored fractional
High-resolution noncontrast 1-mm-cut head computed tomog- anisotropy techniques make possible the visualization of the
raphy (CT; reconstructed to 0.6-mm cuts) is obtained preproce- thalamic nuclei, including definition of the VIM, promising
durally and coregistered onto the high-resolution MRI improvement on targeting of thalamic sites.32
sequences. The VIM contralateral to the predominant tremor
extremity is targeted in the following way: anteroposterior
coordinate being one-quarter the AC distance plus 1 mm anteri-
21.3.2 Outcomes
or to PC (1-mm anterior displacement is intended to reduce the There are several case series of SRS thalamotomies in the litera-
risk of adversely affecting the ventral posterolateral [VPL] sen- ture for patients with PD or ET, documenting the success of this
sory nucleus of the thalamus); lateral coordinate being one-half technique for the treatment of tremor (▶ Table 21.1). The larg-
the third ventricle width plus 11 mm and superiorinferior coor- est series of SRS thalamotomy for ET 33 presented the results of
dinate being 2.5 mm superior to the AC–PC line. A simulated 161 patients, who underwent a total of 203 thalamotomies
target is placed in the inferior lateral corner of the thalamus (119 unilateral and 42 bilateral). The vast majority of the
such that 50% of the isodose line (4-mm collimator) is coinci- patients were treated with 140 Gy, varying from 140 to 150 Gy,
dent with the inferior lateral border of the thalamus and 20% using a single isocenter and 4-mm collimation. A statistically
isodose line of the 4-mm collimator is placed medial to the significant decrease in tremor scores for both writing and draw-
internal capsule. The maximum dose of 140 Gy is delivered with ing was observed after a mean postoperative follow-up dura-
one 4-mm isocenter. The anteromedial displacement from the tion of 56 ± 31 months. Overall, 81% of patients showed
actual target is done to avoid possible damage to the internal improvements in drawing and 77% showed improvement in
capsule and the sensory nucleus of the thalamus (▶ Fig. 21.1). writing scores. There were 14 patients who suffered neurologic
The knowledge that the high-signal zone surrounding a tha- side effects that were temporary (6 patients) or permanent (8
lamic lesion is functionally almost intact has led to some patients), which accounted for 6.9% of the 203 treatments. As in
authors changing the placement of the lesion at the real target other series in the literature, there was a correlation between
instead of shifting away to avoid capsular and sensory nucleus lesion volume and complications. The mean lesion volume for
involvement.30 A more conservative approach has been recom- the 157 procedures in which no complications was identified as
mended by some authors using 130 Gy and preventing the 10 188 ± 224 mm3, and for the 14 procedures for which complica-
to 15% isodose line from extending into capsular and sensory tions were identified, the mean lesion volume was 871 ±
nucleus.30 Another important point is the concept that each 742 mm3 (p < 0.001).

Fig. 21.1 Screenshot of planning magnetic reso-


nance imaging showing ventralis intermedius
(VIM) target in the left thalamus. A dose of
140 Gy using a 4-mm collimator to a single
isocenter was delivered.

170
Stereotactic Radiosurgery in Movement Disorders

Table 21.1 Literature review of radiosurgery thalamotomy for movement disorders


Study Pathology N Lesions FU Good (%) Mild Failed Complications Excellent
(mo) (%) (%)

Young et al, ET 51 51 26 (NR) 47 (92.1) — — 4 (7.8) 1 (1.9)


2000

Kondziolka et al, ET 27a 27 36 (4– 18 (66.6) 6 (22.2) — 3 (11.1) 2 (7.4)


2008 96)

Young et al, ET 161 203 56 ± 31 2 (14.2) 81 NR NR 14 (6.9)


2010

Elaimi, 2010 ET 1 1 72 1 (100) 1b — — —

Pan et al, PD 6 6 4.5 (2–9) 3 (50) 3 (50) — — 1 (16.6)


1996

Duma et al, PD 34 38 28 (6– 3 (50) 11 (29) 4 (10.5) 4 (10.5) No


1998 58)

Young et al, PD 102 102 47 (11– 78 (76.5) 12 (11.8) — 12 (11.8) 2 (1.9)


2000 93)

Ohye et al, PD and ET 30a 30 30 (24– 24 (80) — — 6 (20) No


2002 96)

Duma et al, PD and ET 42 46 30 (6– 25 (54) 13 (28) 4 (8.6) 4 (8.6) 1 (2.3)


2007 90)

Ohye et al, PD and ET 85 NR c 80 NR 4 (4.7) NR —


2009

Lim et al, PD and ET 14a 14 19.2 (7– Serious NR NR NR 3 (21.4)


2010 30) 1 (6)

Abbreviations: ET, essential tremor; FU, median and range of follow-up; NR, not reported; PD, Parkinson disease.
aPatients available for follow-up, and excellent and good.
bTransient.
cOver a 17-year interval (median follow-up and range not reported).

In addition to ET, thalamotomies have been used to treat evaluation. The physiological mechanism of such improvement
tremor dominant PD. Duma et al11 reported a series of 42 in rigidity and bradykinesia remains unexplained, but may be
patients submitted to 46 thalamotomies for the treatment of PD attributable to the open-label design of the study. In the whole
and ET with 6 to 90 months of follow-up (median 30 months). group, one transient (0.66%) and two permanent complications
No change in tremor was observed in 4 (8.6%) patients, mild (1.3%) were reported. In all three of these patients, the side
improvement in 4 (8.6%), good improvement (> 50%) in 13 effects were due to lesions that became larger than expected
(28%), excellent improvement in 13 (28%), and complete elimi- rather than due to targeting errors.
nation was observed in 12 (26%) patients. The median time of
improvement onset was 2 months (range 1 week–8 months).
Independent neurologist evaluation scores of patients’ response 21.3.3 Timing of Therapeutic Efficacy
to treatment were obtained at regular clinical follow-up inter-
vals. Complications were observed in one (2.3%) patient who
and Complications
was submitted to bilateral lesions and suffered a mild acute Some authors recommend that at least 1 year elapse before a
dysarthria 1 week after treatment. Young et al34 reported the contralateral lesion is performed and further recommend that
results of 102 PD patients who underwent SRS thalamotomy an MRI scan be obtained immediately before a second, contrala-
for the treatment of tremor. After a median follow-up of 47 teral lesion to confirm that the first lesion is of usual size, and
months (range 11–93 months), 78 (76.5%) were completely that there are no surrounding signal changes indicating an
tremor-free contralateral to the treated side, 12 (11.8%) were excess radiation reaction. Dose, volume, and time are the key
nearly free, and 12 (11.8%) had failed the treatment. Blinded components that determine the nature of functional ablation. 12,
35,37 In general, the MRI of a typical post-SRS thalamotomy pa-
assessments of UPDRS tremor scores showed statistically signif-
icant improvements in overall tremor, action tremor, and trem- tient demonstrates a well-demarcated, peripherally enhancing
or at rest. Although thalamotomies are not as effective as other lesion 4 to 5 mm in diameter, surrounding a hypoattenuating
targets for other symptoms of PD, the authors reported central region (▶ Fig. 21.2).
improvement in rigidity that was a statistically significant Complications may arise from lesions that develop larger
improvement and was maintained at 4-year follow-up than expected or from misplaced lesions. In the case of

171
Stereotactic Radiosurgery in Movement Disorders

Fig. 21.2 (a) T1-weighted axial magnetic resonance (MR) image with gadolinium contrast demonstrating a noncontrast-enhancing hypointense area
in the left ventralis intermediate nucleus of the thalamus 7 months after SRS thalamotomy. (b) T2-weighted axial MR image demonstrating a
hyperintense area in the left VIM. (c) Fluid-attenuated inversion recovery (FLAIR) image of the same lesion demonstrating no significant perilesional
edema.

thalamotomies, such lesions may involve the internal capsule because of the lack of neurophysiological confirmation of the
or ventral posterior thalamic nucleus with associated deficits of target and the sometimes unpredictable growth of the radio-
weakness or paralysis, dysphasia, or sensory loss. Approxi- surgical lesion, the neurologic complication rate of SRS thala-
mately 4 to 5% of patients experience side effects after SRS tha- motomy may be greater than that of open procedures.13,17,33,42
lamotomy, but usually there is gradual recovery over several Eighty-five to 90% of patients showed significant improvements
months. in tremor in short-term follow-up studies. These results have
Benefits and complications from SRS have been documented changed the use of the open lesioning technique in favor of SRS
to occur between 1 and 12 months after the procedure.38,39,40 thalamotomy in some centers.13,15,17,24 Lesioning of the thala-
The time needed for the radiation to damage the targeted tissue mus is an alternative treatment for tremor and SRS ablation
accounts for the delay in onset of benefits and complications. may be effective when properly performed. It should be
The lesional effect can be accelerated by increasing the dose of reserved for use in patients with severe disabling tremor who
radiation; however, the optimal dose of radiation has not been cannot tolerate open surgical procedures and should be per-
clearly established and with increased doses, higher rates of formed by neurosurgeons who have experience in DBS or RF
complications have been anticipated. The peak central doses thalamic procedures because targeting of the thalamus without
may range from 120 to 200 Gy. neurophysiological mapping can be very challenging.
In one study, a comparison was conducted between a sub-
group of patients in whom “low-dose” lesions (mean 120 Gy)
and those in whom “high-dose” lesions were made (mean 21.4 Radiosurgical Pallidotomy
160 Gy) for purposes of obtaining dose-response information. 17
Unilateral or bilateral pallidotomy may be accomplished with
Better tremor reduction in the high-dose group (78% mean
SRS, but is currently rarely used due to the superior effective-
improvement) than in the low-dose group (56% mean improve-
ness of globus pallidus internus (GPi) and subthalamic DBS in
ment) (p = 0.04) was reported and there were no neurologic
treating the full spectrum of PD symptoms (▶ Table 21.2). The
complications. However, Okun et al41 reported a higher rate of
goals of pallidotomy include relief of contralateral levodopa-
complications in a series of eight patients—seven of whom were
induced dyskinesias as well as improvement in contralateral
treated with a maximum dose of 200 Gy. These included death
bradykinesia, rigidity, and tremor.
secondary to dysphagia, and aspiration pneumonia, hemiplegia,
visual field deficits, aphasia, and pseudobulbar laughter. Most
neurosurgeons perform radiosurgical lesioning for functional 21.4.1 Technique
disorders at lower doses (e.g., 140 Gy).
For SRS pallidotomy, an axial image is selected that demon-
strates both AC and PC, and a coronal image is viewed at a point
2 to 3 mm anterior to the midcommissural point. The GPi and
21.3.4 Summary the underlying optic tract are visualized and the isocenter is
Although the efficacy of SRS thalamotomy for controlling trem- placed such that 50% isodose line of a 4-mm collimator is with-
or is similar to that reported for RF thalamotomy and DBS in the GPi and superior to the optic tract. It is crucial to define

172
Stereotactic Radiosurgery in Movement Disorders

Table 21.2 Literature review of radiosurgery pallidotomy for movement disorders


Study N Lesions Dose (Gy) FU Excellent Good Fair Poor Complications
(mo) (%) (%) (%) (%) (%)

Rand et al, 8 8 140–165 NR 2 (25) 4 (50) — 2 (25) No


1993

Friedman et al, 4 4 180 12 — 1 (25) — 3 (75) 1 (25)


1996

Bonnen et al, 1 1 140 NR — — 1 (100) — 1 (100)


1997

Young et al, 29 34 120–140 20.6 (6– 65.5–86.6a — — 1 (3.4)


1998 48)

Duma et al, 18 160 (90– 8 (6–40) — 6 (33) 3 (17) 9 (50) 9 (50)


2007 165)

Abbreviations: FU, median and range of follow-up; NR, not reported.


aExcellent and good, depending on the symptom—bradykinesia or dyskinesia.

that target in both the axial and coronal planes to properly (applied dose ranged between 120 and 140 Gy with a 4-mm
determine the vertical coordinate in relation to the optic tract. collimator) and submitted to 34 lesions; 22 were treated with
Furthermore, the 50% isodose line should be as far away from RF and submitted to 25 lesions. The median follow-up was 20.6
the optic tract as possible, within a location that includes the months (range 6–48 months), and the evaluations of motor
GPi completely within the 50% line, and limiting the dose to the performance and postoperative assessments were obtained by
optic tract to 8 Gy at most. At our institution, we prefer a radio- blinded observers. Improvement in dyskinesias was observed in
surgical dose of 140 Gy for creating a brain lesion with a 4-mm 86.6% versus 83.3% in the SRS and RF groups, and in bradykine-
collimator. With these parameters, a lesion of about 6 mm in sia and rigidity in 65.5% versus 63.6%, respectively. One patient
diameter (113-mm volume) will usually develop 3 to 9 months in the radiosurgery group (3.4%) presented with contralateral
after the procedure. Larger radiosurgical doses will produce homonymous hemianopsia secondary to a lesion larger than
lesions more rapidly, but the volume of the lesion may vary expected (volume 950 mm3) at 9 months postoperatively. Two
more, producing larger lesions that are associated with higher other patients developed larger lesions (520 mm3 and 700 mm3,
complication rates. Conversely, smaller doses may fail to pro- respectively), but they were not associated with any clinical
duce any definable lesion, therefore leading to no therapeutic side effects.
effect. According to the authors, the results were equally as good as
those obtained in the RF pallidotomies when electrophysiologi-
cal localization was used.
21.4.2 Outcomes
The most consistent effect of pallidal ablation is relief of con-
tralateral levodopa-induced dyskinesias. However, the amount
21.4.3 Complications
of improvement in contralateral bradykinesia, rigidity, and As mentioned for thalamotomies, the drawbacks of radiosurgi-
tremor varies among many studies.43 cal pallidotomy concern the latency between the procedure and
Stereotactic radiosurgery pallidotomy was first reported by the clinical benefit (2–3 months minimum) and the possibility
Rand in 1993,4 who used the technique in eight patients with that the lesion produced by radiosurgery will continue to
doses between 140 and 165 Gy. Significant improvement in enlarge on a delayed basis and involve adjacent normal struc-
contralateral rigidity, bradykinesia, and dyskinesias was tures. A review of the literature reveals only a few studies that
observed in four (25%) patients. Positive results without a major document the benefit of SRS pallidotomy in a blinded and
impact in quality of life were observed in two (25%) patients unbiased manner, with many more reports of complications
and there was no change in the course of the disease in the oth- after SRS pallidotomy. One such report by Duma et al 11 is a ser-
er two (25%) patients. Four cases of PD were described by Fried- ies of 18 patients with PD who underwent SRS pallidotomy. Fif-
man et al,18 who underwent unilateral pallidotomy using a 4- teen patients were treated using a single 4-mm collimator and
mm collimator and a dose of 180 Gy. No significant improve- three were treated using two 4-mm collimators with a median
ment occurred during the follow-up interval of 18 months; one maximum prescription dose of 160 Gy (range 90–165 Gy). Over
patient developed a stroke due to radiation vasculopathy with a median average follow-up of 8 months (range 6–40 months),
severe radiation changes in the blood vessels adjacent to the only six (33%) patients showed transient improvement in
radiosurgical lesion. rigidity and dyskinesia. Three (17%) patients were unchanged
Young et al44 reported a comparative study of 51 patients and nine (50%) were worsened by the treatment. Of the six
with PD who underwent pallidotomies. The patients were div- patients with improvement, two exhibited visual field deficits.
ided into two groups: 29 were treated with radiosurgery Overall, four (22%) patients had visual field deficit, three (16%)

173
Stereotactic Radiosurgery in Movement Disorders

had speech or swallowing difficulties, three (16%) had worsen- STN with a single 120-Gy 4-mm shot. After 3.5 years of follow-
ing of their gait, and one (5%) patient had numbness in the up, the STN lesion was stable and well placed and the patient
contralateral hemibody. Nine patients (50%) had one or experienced reduction in dyskinesias and improvement in
more complications related to the treatment, which were unre- tremor and rigidity.49
sponsive to steroid treatment and considered to be permanent.
The explanation of the high complication rate in this series was
related to the variability and unpredictability of the lesion size
21.5.1 Complications
when the GPi served as the target. Given the paucity of SRS subthalamotomies, the main complica-
The differences in outcome comparing VIM and GPi led the tion associated with subthalamotomies, virtually all of which
authors to conclude that there might be a difference in sensitiv- are reports of RF subthalamotomies being contralateral hemi-
ity to radiation between these two nuclei, probably represent- ballism. As shown in the Cuban report, three patients devel-
ing anatomical susceptibility to very small venous or arterial oped severe generalized chorea, dysarthria, and balance
infarctions in the area of the GPi, caused by the tapering end- instability that lasted 6 months before spontaneously resolving.
artery distribution of the lenticulostriate supply. 12,45 Similarly, in the Bristol report, one patient developed postope-
Homonymous hemianopsia after SRS pallidotomy is a serious rative hemiballisms on the side contralateral to the STN lesion,
deficit and is seen in 4 to 6% of patients. According to the litera- lasting for 3 months before spontaneously resolving.
ture, these visual field deficits are permanent and not com-
pletely unexpected in view of the proximity of the optic tract to
the GPi, which may receive as much as 15 to 25 Gy, which is
21.5.2 Summary
well beyond the usual 8-Gy-maximum radiation dose usually In contrast to SRS thalamotomy, which is an alternative to DBS
accepted for the optic apparatus. Other complications such as for the treatment of tremors associated with ET or PD, in
weakness or paralysis and dysphagia can occur due to involve- patients in whom surgical procedures are contraindicated, SRS
ment of the internal capsule by the lesion. pallidotomy and subthalamotomy currently play only a limited
role in the treatment of patients with movement disorders. This
is mainly owing to the high efficacy, relatively low complication
21.4.4 Summary rates, the possibility of reversibility, and fine adjustment of sti-
The number of centers that have been performing radiosurgery mulation parameters associated with DBS of basal ganglia tar-
pallidotomies compared with those performing thalamotomies gets in the surgical treatment of patients with movement
reflects the lack of reliability of the procedure and that other disorders.
therapeutic options are superior to SRS targeting the GPi. The
relatively high incidence of visual field deficits after SRS pallid-
otomy has caused many centers to limit its use to very rare 21.6 Conclusions
cases in which DBS or RF pallidotomy cannot be used and the Advances in stereotactic techniques associated with improve-
patient is significantly disabled by PD. ments in MRI targeting, planning software, and a better knowl-
edge of SRS parameters have brought the technique to a
21.5 Subthalamotomy precision capable of performing focal and precise lesions in the
basal ganglia for the treatment of movement disorders,
The subthalamic nucleus (STN) is considered one of the major although competing lesioning technologies are appearing in the
targets and DBS as the main option for the surgical treatment of market, as MRI-guided focused ultrasound50 and MRI-guided
the broad spectrum of symptoms in patients with idiopathic laser ablation,51 which may prove more effective and safer than
PD. Benabid et al were the first group to demonstrate the effi- SRS.
cacy of STN DBS in the treatment of PD in the mid-1990s. 46 Using modern functional SRS parameters, SRS thalamotomy
Despite the overwhelming preference of practitioners is a useful procedure for patients who are not suitable for an
towards DBS of the STN for treatment of PD symptoms, subtha- open surgical procedure and understand the potential risk of
lamotomy in the form of RF ablation is a seldom performed pro- neurologic complications. Complications appear to be mostly
cedure; therefore, case reports are limited to mainly centers related to the variability of lesion volumes using the same
outside of the United States that have reported on the benefits radiosurgical parameters rather than to the stereotactic target
of therapeutic RF subthalamotomy in patients with PD.47,48 precision. The factors related to this unpredictable thalamic
These include a Cuban series of 18 patients who underwent reaction to high single-dose radiation are still unknown.
bilateral RF subthalamotomies and exhibited a 58% improve- The results of radiosurgery pallidotomy are not homogene-
ment in UPDRS motor scores in the “off” period after average ous in the literature. Many reports disclosed an unacceptably
follow-up of 16 months. Likewise, the Bristol series of 50 unilat- high complication rate. Although just a few centers reported
eral RF subthalamotomies in 39 patients demonstrated 46% their results, the majority of them were not satisfactory, leading
improvement in the motor UPDRS score in 24 months. them to abandon the procedure. Moreover, further studies are
Stereotactic radiosurgery subthalamotomy as treatment for necessary to establish the role of SRS targeting the STN because
idiopathic PD is even more rarely performed. The only report of its small size and complex anatomical relationships make this
radiosurgery subthalamotomy in the literature described the target less suitable for the procedure.
case of an elderly patient previously submitted to a RF pallidot- In conclusion, although the advent of DBS, with its possibility
omy who underwent a contralateral radiosurgery lesion of the of reversibility and fine adjustments of stimulating parameters,

174
Stereotactic Radiosurgery in Movement Disorders

has supplanted lesioning as the first alternative in movement [22] Young RF, Vermeulen SS, Grimm P, Posewitz A. Electrophysiological target lo-
calization is not required for the treatment of functional disorders. Stereotact
disorder surgery, SRS might still be the only treatment option
Funct Neurosurg 1996; 66 (Suppl 1): 309–319
for selected patients. The development of alternative techniques [23] Friedman DP, Goldman HW, Flanders AE, Gollomp SM, Curran WJ Jr. Stereo-
of targeting with the visualization of the nuclei51,52 and novel tactic radiosurgical pallidotomy and thalamotomy with the gamma knife: MR
lesioning techniques such as MRI-guided focused ultrasound imaging findings with clinical correlation—preliminary experience. Radiology
may bring better efficacy and safety to pallidotomy and subtha- 1999; 212: 143–150
[24] Young RF, Shumway-Cook A, Vermeulen SS, et al. Gamma knife radiosurgery
lamotomy. This in turn may bring a better understanding of tar-
as a lesioning technique in movement disorder surgery. J Neurosurg 1998;
geting these structures, with SRS defining further the 89: 183–193
population of movement disorder patients who may benefit [25] Benabid AL, Benazzouz A, Hoffmann D, Limousin P, Krack P, Pollak P. Long-
from this technique. term electrical inhibition of deep brain targets in movement disorders. Mov
Disord 1998; 13 (Suppl 3): 119–125
[26] Starr PA, Vitek JL, Bakay RA. Ablative surgery and deep brain stimulation for
Parkinson’s disease. Neurosurgery 1998; 43: 989–1013, discussion 1013–
References 1015
[27] Elaimy AL, Demakas JJ, Arthurs BJ, et al. Gamma knife radiosurgery for essen-
[1] Dogali M, Fazzini E, Kolodny E, et al. Stereotactic ventral pallidotomy for Par- tial tremor: a case report and review of the literature. World J Surg Oncol
kinson’s disease. Neurology 1995; 45: 753–761 2010; 8: 20
[2] Goldman MS, Kelly PJ. Stereotactic thalamotomy for medically intractable es- [28] Ohye C, Shibazaki T, Hirato M, Inoue H, Andou Y. Gamma thalamotomy for
sential tremor. Sterotact Func Neurosurg 2000; 342: 461–468 parkinsonian and other kinds of tremor. Stereotact Funct Neurosurg 1996; 66
[3] Hariz MI, Bergenheim AT. A 10-year follow-up review of patients who under- (Suppl 1): 333–342
went Leksell’s posteroventral pallidotomy for Parkinson disease. J Neurosurg [29] Young RF, Jacques DS, Rand RW, Copcutt BR. Medial thalamotomy with the
2001; 94: 552–558 Leksell Gamma Knife for treatment of chronic pain. Acta Neurochir Suppl
[4] Jankovic J, Cardoso F, Grossman RG, Hamilton WJ. Outcome after stereotactic (Wien) 1994; 62: 105–110
thalamotomy for parkinsonian, essential, and other types of tremor. Neuro- [30] Ohye C, Shibazaki T, Ishihara J, Zhang J. Evaluation of gamma thalamotomy
surgery 1995; 37: 680–686, discussion 686–687 for parkinsonian and other tremors: survival of neurons adjacent to the tha-
[5] Laitinen LV, Bergenheim AT, Hariz MI. Leksell’s posteroventral pallidotomy in lamic lesion after gamma thalamotomy. J Neurosurg 2000; 93 (Suppl 3): 120–
the treatment of Parkinson’s disease. J Neurosurg 1992; 76: 53–61 127
[6] Linhares MN, Tasker RR. Microelectrode-guided thalamotomy for Parkinson’s [31] Ohye C, Shibazaki T. Location of the thalamic Vim nucleus. Its relation to the
disease. Neurosurgery 2000; 46: 390–395, discussion 395–398 whole thalamic length.] Funct Neurosurg 2002; 41: 52–53
[7] Lozano A, Hutchison W, Kiss Z, Tasker R, Davis K, Dostrovsky J. Methods for [32] Sedrak M, Gorgulho A, Frew A, Behnke E, DeSalles A, Pouratian N. Diffusion
microelectrode-guided posteroventral pallidotomy. J Neurosurg 1996; 84: tensor imaging and colored fractional anisotropy mapping of the ventralis in-
194–202 termedius nucleus of the thalamus. Neurosurgery 2011; 69: 1124–1129, dis-
[8] Tasker RR, Munz M, Junn FS, et al. Deep brain stimulation and thalamotomy cussion 1129–1130
for tremor compared. Acta Neurochir Suppl (Wien) 1997; 68: 49–53 [33] Young RF, Li F, Vermeulen S, Meier R. Gamma Knife thalamotomy for treat-
[9] Leksell L. Cerebral radiosurgery. I. Gammathalanotomy in two cases of in- ment of essential tremor: long-term results. J Neurosurg 2010; 112: 1311–
tractable pain. Acta Chir Scand 1968; 134: 585–595 1317
[10] Steiner L, Lindquist C, Forster D, Backlund EO, eds. Gamma knife thalamoto- [34] Young RF, Jacques S, Mark R, et al. Gamma knife thalamotomy for treatment
my for tremor: report of two cases. In: Radiosurgery. Baseline and Trends. of tremor: long-term results. J Neurosurg 2000; 93 (Suppl 3): 128–135
New York, NY: Raven Press; 1992:237–243 [35] Miles J, Redfern RM. The place of thalamotomy in the treatment of parkinson-
[11] Duma CM. Movement disorder radiosurgery—planning, physics and compli- ism. Br J Neurosurg 1987; 1: 311–315
cation avoidance. Prog Neurol Surg 2007; 20: 249–266 [36] Friehs GM, Noren G, Ohye C, et al. Lesion size following Gamma Knife treat-
[12] Friehs GM, Norén G, Ohye C, et al. Lesion size following Gamma Knife treat- ment for functional disorders. Stereotact Funct Neurosurg 1996; 66 (Suppl):
ment for functional disorders. Stereotact Funct Neurosurg 1996; 66 (Suppl 320–328
1): 320–328 [37] Konziolka D, Lunsford LD, Claassen D, Maiz AH, Flickinger JC. Radiobiology
[13] Kondziolka D, Ong JG, Lee JY, Moore RY, Flickinger JC, Lunsford LD. Gam- of radiosurgery, I: the normal rat brain model. Neurosurg 1992; 31: 271–
ma Knife thalamotomy for essential tremor. J Neurosurg 2008; 108: 279
111–117 [38] Duma CM, Jacques D, Kopyov OV. The treatment of movement disorders using
[14] Ohye C. From selective thalamotomy with microrecording to gamma tha- Gamma Knife stereotactic radiosurgery. Neurosurg Clin N Am 1999; 10: 379–
lamotomy for movement disorders. Stereotact Funct Neurosurg 2006; 84: 389
155–161 [39] Niranjan A, Kondziolka D, Baser S, Heyman R, Lunsford LD. Functional out-
[15] Ohye C, Shibazaki T. Treatment of functional disorders with gamma knife tha- comes after gamma knife thalamotomy for essential tremor and MS-related
lamotomy. Prog Neurol Surg 2009; 22: 170–181 tremor. Neurology 2000; 55: 443–446
[16] Bonnen JG, Iacono RP, Lulu B, Mohamed AS, Gonzalez A, Schoonenberg T. [40] Okun MS, Stover NP, Subramanian T, et al. Complications of gamma knife sur-
Gamma knife pallidotomy: case report. Acta Neurochir (Wien) 1997; 139: gery for Parkinson disease. Arch Neurol 2001; 58: 1995–2002
442–445(Wien) [41] Ohye C, Shibazaki T, Sato S. Gamma knife thalamotomy for movement disor-
[17] Duma CM, Jacques DB, Kopyov OV, Mark RJ, Copcutt B, Farokhi HK. Gamma ders: evaluation of the thalamic lesion and clinical results. J Neurosurg 2005;
knife radiosurgery for thalamotomy in parkinsonian tremor: a five-year ex- 102 (Suppl): 234–240
perience. J Neurosurg 1998; 88: 1044–1049 [42] Lang AE, Duff J, Saint-Cyr JA, et al. Posteroventral medial pallidotomy in Par-
[18] Friedman JH, Epstein M, Sanes JN, et al. Gamma knife pallidotomy in ad- kinson’s disease. J Neurol 1999; 246 (Suppl 2): II28–II41
vanced Parkinson’s disease Ann Neurol 1996; 39: 535–538 [43] Young RF, Vermeulen S, Posewitz A, Shumway-Cook A. Pallidotomy with the
[19] Friehs GM, Ojakangas CL, Pachatz P, Schröttner O, Ott E, Pendl G. Thalamoto- gamma knife: a positive experience. Stereotact Funct Neurosurg 1998; 70
my and caudatotomy with the Gamma Knife as a treatment for parkinsonism (Suppl 1): 218–228
with a comment on lesion sizes. Stereotact Funct Neurosurg 1995; 64 (Suppl [44] Friedman JH, Fernandez HH, Sikirica M, Stopa E, Friehs G. Stroke induced by
1): 209–221 gamma knife pallidotomy: autopsy result. Neurology 2002; 58: 1695–1697
[20] Pan L, Dai JZ, Wang BJ, Xu WM, Zhou LF, Chen XR. Stereotactic Gamma thala- [45] Benabid AL, Pollak P, Gross C, et al. Acute and long-term effects of subthala-
motomy for the treatment of parkinsonism. Stereotact Funct Neurosurg mic nucleus stimulation in Parkinson’s disease. Stereotact Funct Neurosurg
1996; 66 (Suppl 1): 329–332 1994; 62: 76–84
[21] Rand RW, Jacques DB, Melbye RW, Copcutt BG, Fisher MR, Levenick MN. [46] Alvarez L, Macias R, Lopez G, et al. Bilateral subthalamotomy in Parkinson’s
Gamma Knife thalamotomy and pallidotomy in patients with movement dis- disease: initial and long-term response. Brain 2005; 128: 570–583
orders: preliminary results. Stereotact Funct Neurosurg 1993; 61 (Suppl 1): [47] Gill SS, Heywood P. Bilateral dorsolateral subthalamotomy for advanced Par-
65–92 kinson’s disease. Lancet 1997; 350: 1224

175
Stereotactic Radiosurgery in Movement Disorders

[48] Keep MF, Mastrofrancesco L, Erdman D, Murphy B, Ashby LS. Gamma knife [51] Sedrak M, Gorgulho A, Bari A, et al. Diffusion tensor imaging (DTI) and col-
subthalamotomy for Parkinson disease: the subthalamic nucleus as a new ra- ored fractional anisotropy (FA) mapping of the subthalamic nucleus (STN)
diosurgical target. Case report. J Neurosurg 2002; 97 (Suppl): 592–599 and the globus pallidus interna (GPi). Acta Neurochir (Wien) 2010; 152:
[49] Elias WJ, Huss D, Voss T, et al. A pilot study of focused ultrasound thalamoto- 2079–2084
my for essential tremor. N Engl J Med 2013; 369: 640–648 [52] Pouratian N, Zheng Z, Bari AA, Behnke E, Elias WJ, Desalles AA. Multi-institu-
[50] Willie JT, Laxpati NG, Drane DL, et al. Real-time magnetic resonance-guided tional evaluation of deep brain stimulation targeting using probabilistic con-
stereotactic laser amygdalohippocampotomy for mesial temporal lobe epi- nectivity-based thalamic segmentation. J Neurosurg 2011; 115: 995–1004
lepsy. Neurosurgery 2014; 74: 569–584, discussion 584–585

176
Gamma Knife Radiosurgery for Obsessive–Compulsive Disorder

22 Gamma Knife Radiosurgery for Obsessive–Compulsive


Disorder
Nrupen Baxi, Andrew Brunswick, Eric Mazel, and Douglas Kondziolka

intervention include target brain lesioning or electrical brain


Key Points stimulation. Lesioning can be performed using radiofrequency
or radiation energy. Multiple small studies have shown that
● Obsessive–compulsive disorder has a lifetime prevalence of Gamma Knife radiosurgery (GKRS) may have the potential to
approximately 2.3% in the general population, with treat- provide significant benefits to some within this subset of
ment refractory obsessive–compulsive disorder causing patients. As is the case for all of our therapies, multiple ques-
significant disability. tions remain to be answered. In this chapter, we review the
● Neurosurgical interventions for refractory obsessive–compul- anatomy and pathophysiology of OCD as it pertains to surgical
sive disorder date to the 1950s and include target brain target selection, compare GKRS to deep brain stimulation (DBS)
lesioning with either radiofrequency or radiation energy, or as surgical modalities, review major studies of GKRS for OCD to
electrical brain stimulation. These interventions are designed date, and propose further avenues of research.
to disrupt circuits connecting the frontal lobe, striatum, and
thalamus that are believed to be overactive in patients with
severe obsessive–compulsive disorder. 22.2 Anatomy, Pathophysiology,
Radiosurgery and electrical stimulation each have distinctive

qualities.
and Surgical Target Selection
● For radiosurgical planning, we prefer two 4-mm isocenters in The neuroanatomy and neurobiology of OCD continue to be elu-
the inferior part of the anterior limb of the internal capsule cidated, but remain controversial.4 Researchers have made use
at the midputaminal point, separated in the superoinferior of brain volumetric studies as well as functional studies such as
plane to mimic the current spread of an electrode. positron emission tomography (PET), functional magnetic reso-
● Of the 27 patients described in the literature who have nance imaging (fMRI), and single-photon emission computed
undergone radiosurgery for obsessive–compulsive disorder, tomography (SPECT) to identify relevant anatomical struc-
20 (74%) have had at least a 25% reduction and 13 (48%) tures.5–10 Functional imaging studies have consistently shown
have had at least a 50% reduction in Yale-Brown Obsessive- abnormal activity in several key regions of the brain, including
Compulsive Scale score. the orbitofrontal cortex (OFC), anterior cingulate cortex (ACC),
and caudate head. Activity within these circuits is heightened
during symptom provocation and normalizes with treatment,
although some argue that this may correlate with rather than
22.1 Introduction cause symptoms.11,12 These areas are believed to be linked
together by a series of parallel circuits termed corticostriatal/
Obsessive-compulsive disorder (OCD) is a chronic, disabling thalamocortical (CSTC) circuits, which bring output from multi-
psychiatric disorder characterized by obsessions and/or com- ple cortical areas including the OFC through the striatum, into
pulsions that can significantly affect quality of life for afflicted the thalamus, and then return this input to the cortex, forming
patients. Obsessions are defined as recurrent, unwanted a loop.13
thoughts, urges, or images; compulsions are repetitive behav- To further complicate matters, some have made light of the
iors or mental actions a patient must perform in response to a fact that “refractory OCD” does not respond to traditional treat-
compulsion.1 Symptoms can leave patients overwhelmed by ments based on historical models of the disease. One hypothe-
anxiety and unable to effectively function in society. Approxi- sis is that refractory OCD is actually a separate disease requiring
mately 25 to 33% of patients with the disorder will contemplate different neuroanatomic and neurobiologic considerations. 14
suicide.1,2 The mainstay of treatment has been and remains Because DBS and GKRS have traditionally been used exclusively
medical. First-line therapy includes pharmacotherapy with se- for this subclass of OCD, one must wonder about the legitimacy
rotonin reuptake inhibitors (SSRIs) along with cognitive–behav- of using traditional models in the surgical treatment of this
ioral psychotherapy. Additional pharmacological agents include disease.
tricyclic antidepressants, low-dose antipsychotics, and benzo- Surgical interventions in OCD aim to interrupt the hyperac-
diazepines. The estimated lifetime prevalence of OCD in the tivity of CSTC circuits. This can be accomplished by lesioning
United States is 2.3%, with a slightly higher predominance of either key regions in the brain or by lesioning pathways that
males in childhood and a higher predominance of women in form the circuits connecting these regions. Because these path-
adulthood.3 The mean age of presentation is 19.5 years, with ways are diffuse, multiple targets have been proposed and
25% of cases having an onset before age 14.1 There is also a high attempted. Major targets of interest have included the anterior
incidence of comorbid conditions that accompany OCD, includ- limb of the internal capsule (ALIC), ACC, subcaudate tracts, and
ing tic disorder, anxiety disorder, posttraumatic stress disorder the inferior thalamic peduncle (ITP).
(PTSD), bipolar disorder, and major depressive disorder.1 The most common lesioning target for the treatment of OCD,
Neurosurgery has been considered for a select subset of the anterior internal capsule, was discovered somewhat fortui-
patients who have severe, medically refractory OCD. Types of tously. Varied initial success with leucotomies and other larger

177
Gamma Knife Radiosurgery for Obsessive–Compulsive Disorder

destructive procedures in the treatment of psychiatric disorders Sheehan et al has published the latest series of five patients
led researchers Meyer and Beck to carefully analyze postmor- treated with a single isocenter with 140 to 160 Gy. In their ser-
tem brain specimens for predictors of success. They speculated ies, 80% of the patients had a major reduction in their Y-BOCS
that a lesion in front of the anterior horn would sever connec- scores.24
tions to the frontal lobes.15,16 This lesion would interrupt fiber Patient selection for surgical OCD procedures varies slightly
tracts connecting the OFC to the caudate and thalamic nuclei, among studies, but common criteria are listed in ▶ Table 22.1.
thus disrupting circuitry vital to the pathophysiology of OCD. Generally, patients have a confirmed diagnosis of OCD without
Talairach was the first to perform this anterior capsulotomy in significant other comorbid conditions. They also have Y-BOCS
hopes of treating refractory schizophrenia. Leksell was able to values in the moderate, severe, or extreme range. Preprocedural
modify this technique to utilize his stereotactic system and a magnetic resonance imaging (MRI) is performed to evaluate for
radiofrequency thermal source for lesioning. Both noted poor any structural pathology. One or two isocenters are chosen in
results in treating schizophrenia, but satisfactory results in the inferior part of the anterior limb of the internal capsule at
nearly half the patients with anxiety or obsessional neuroses.15, the midputaminal point. We prefer two 4-mm isocenters sepa-
16 Leksell also performed a gamma capsulotomy where the rated in the superoinferior plane, mimicking the current spread
anterior capsule was lesioned using 100 Gy of stereotactically of an electrode, as seen in ▶ Fig. 22.1. The patient undergoes
delivered gamma radiation.16 placement of a stereotactic head frame on the day of the proce-
dure and is given a maximum dose of 140 to 180 Gy to the tar-
get. The permanent lesion within the internal capsule may take
22.3 Gamma Knife Radiosurgery: 2 to 4 months to fully form as tissue necrosis occurs. Patients
may have clinical improvements in their symptoms within 2 to
Outcomes and Technical 3 months postprocedure.23
Considerations Adverse events from radiosurgical lesioning procedures have
been described in older series. Mindus et al reported symptoms
The Yale-Brown Obsessive-Compulsive Scale (Y-BOCS) was of frontal lobe dysfunction including apathy and disinhibition,
developed in 1989 to allow researchers to measure the severity which were attributed to the high radiation doses used. 15 They
of OCD.17,18 A separate score is generated for both obsessions advocated for lower doses to limit these effects. They also pro-
and compulsions. Interrater variability is low, enabling its use posed proactively monitoring patients for personality changes,
by different members of a research team. The scale has been impulsiveness, and cognitive changes associated with possible
validated, with reduction in the total score correlating with decreased frontal lobe processing.15 Rück et al described one
improvement in OCD symptoms. Modern research studies patient with right-sided radiation necrosis and another with
including clinical drug trials and trials assessing neurosurgical malignant edema that was maximal at 1 year postprocedure.
procedures have used the Y-BOCS to quantify outcomes.17,18 This latter patient also developed apathy and executive prob-
The total score is assessed using a 10-item questionnaire, with a lems secondary to the edema. He and a third patient also devel-
maximum possible score of 40 points. The groups are then clas- oped urinary incontinence that was persistent.25 Rück et al also
sified in quintiles by severity, with a total score of 0–7 repre- noticed an average weight gain of 11 kg in long-term follow-up.
senting subclinical OCD, 8–15 mild OCD, 16–23 moderate OCD, One patient became sexually disinhibited and was subsequently
24–31 severe OCD, and 32–40 extreme OCD.18 convicted of rape.25 Frontal lobe symptoms were noticed in
Several early series of GKRS for OCD have been published. about 50% of patients overall. The majority of recent studies
Mindus et al published a series of 10 early patients with a 70% have not found significant adverse events; however, their sam-
response rate.15,19 Lopes et al first performed a randomized ple sizes remain small. One group (unpublished) noted that
clinical trial with five patients in which he reported a 60% res- some patients developed cystic changes around some lesions,
ponse rate of at least a 35% reduction in Y-BOCS score utilizing which can be seen after any brain lesioning procedure.
two isocenters on each side. Lesioning remained the mainstay
neurosurgical approach for refractory OCD until the develop-
Table 22.1 Criteria often used for patient selection
ment of DBS. Lopes et al performed a clinical trial after the ini-
tial reports of DBS in 2009, with three out of five patients Inclusions Exclusions
meeting response criteria at 48 months.20 Slavin et al discussed DSM criteria met for OCD Age < 20 y or > 55 y
four patients with marginal responses in 2009 at a neurosurgi-
cal meeting in Marseille, France, in 2009.21 Gouvea et al pub- Duration of symptoms > 5 y Axis I, II, or III complicating diag-
nosis
lished a report of one patient who underwent a sham
radiosurgical procedure in Brazil followed by therapeutic radio- Y-BOCS score > 26 Unable to provide informed con-
surgery 1 year later. She experienced no changes in her Y-BOCS sent
score after the sham procedure but had a dramatic benefit after
Deemed refractory to available
the second procedure.22 Kondziolka et al published a series of medical and cognitive–behavioral
three patients in 2011 with up to 5.5-year follow-up with sig- treatments by OCD specialists
nificant reductions in Y-BOCS scores for all patients, which cor-
Patient provides their own consent
related with improved clinical outcomes. No adverse events
were seen in this small series. They utilized two 4-mm isocen- Abbreviations: DSM, Diagnostic and Statistical Manual of Mental
ters on each side and limited the maximum dose to 140 to Disorders, 5th ed.; OCD, obsessive–compulsive disorder; Y-BOCS, Yale-
150 Gy instead of the 180 to 200 Gy seen in previous studies. 23 Brown Obsessive-Compulsive Scale.

178
Gamma Knife Radiosurgery for Obsessive–Compulsive Disorder

Fig. 22.1 Coronal magnetic resonance imaging


(T1 and T2) for radiosurgical planning in obses-
sive–compulsive disorder showing targeting of
inferior aspect of the anterior limb of the internal
capsule.

Results from the various GKRS studies have been pooled and up-front—three patients in OCD trials using DBS suffered intra-
listed in ▶ Table 22.2. Of the 27 patients who have undergone cranial hemorrhage, although only one had permanent defi-
the procedure in the literature, 20 (74%) of them have had at cits.26,27,28 Deep brain stimulation also carries the risk of sudden
least a 25% reduction and 13 (48%) have had at least a 50% failure, which can be unpredictable, and which can lead to the
reduction in Y-BOCS score. With the mean score of 32.5, a 25% return of symptoms. Deep brain stimulation requires further
reduction may drop patients into a less severe quintile on the invasive procedures for hardware changes, and there have been
Y-BOCS. On average, there was a 45% reduction (15 points) in significant problems associated with battery failure.29 There
Y-BOCS scores postprocedure. In many patients, severe to have been multiple reports of infections related to DBS hard-
extreme OCD lessened to moderate, more manageable OCD. ware.27,28,30,31 There have also been multiple reports of reversi-
ble stimulation-induced side effects such as hypomania,
agitation, anxiety, forgetfulness, and word-finding difficulties. 27,
22.4 Stereotactic Radiosurgery 28,30,32,33 Deep brain stimulation is also more expensive,

and Deep Brain Stimulation requires the use of complex devices that require specialized
programming, and is contraindicated in patients with excoria-
Interruption of circuits thought to be involved in OCD can be tion disorder or OCD with skin picking as a symptom due to
accomplished with both GKRS and DBS. Unfortunately, there wound-healing risks.34
are no long-term studies comparing the two techniques. Each
has its own distinct qualities; the choice may ultimately come
down to patient preference. We believe that from safety and
compliance standpoints for OCD patients, GKRS is superior to
22.5 Conclusion
DBS. Obsessive–compulsive disorder is a severely disabling psychiat-
Gamma Knife radiosurgery creates a permanent lesion in the ric illness that affects millions of people worldwide. Treatment
target of interest. The effect usually takes 2 to 4 months to of the condition can be challenging when symptoms are refrac-
become apparent, the time for tissue necrosis to occur. tory to medical and cognitive–behavioral therapy. Lesioning
Although the lesion is nonreversible and nonadjustable, there is procedures for OCD first developed in the 1950s showed mod-
no risk of hardware failure and there is no need for surgeries to erate success. With the introduction of SRS, lesioning could be
change hardware. Gamma Knife radiosurgery for OCD has risks, performed as an outpatient procedure and with less surgical
including the low risks of brain edema and cyst formation, as risk with similar rates of success. Neurosurgical treatment has
well as adverse events such as apathy, disinhibition, and decline been expanded with the advent of DBS, which provides a rever-
in executive function that have been seen in some series.20,25 sible treatment modality. However, renewed interest in GKRS
Unlike GKRS, DBS creates an immediate, reversible, and has resurfaced as the risks of DBS have been better understood.
adjustable interruption in the target of interest. Deep brain sti- Several small series have shown GKRS to be effective in refrac-
mulation requires an immediate procedure with significant risk tory OCD without the risk associated with deep brain electrode

179
Gamma Knife Radiosurgery for Obsessive–Compulsive Disorder

Table 22.2 Meta-analysis of radiosurgical lesioning patients to symptom dimensions and comorbid conditions. J Clin Psychiatry 2011; 72:
17–26, quiz 119–120
Study N Pre Y- Last Y- Follow-Up Gy [3] Ruscio AM, Stein DJ, Chiu WT, Kessler RC. The epidemiology of obsessive-
BOCS BOCS (mo) compulsive disorder in the National Comorbidity Survey Replication. Mol
Psychiatry 2010; 15: 53–63
Lopes et al, 200920 1 30 10 48 180
[4] Milad MR, Rauch SL. Obsessive-compulsive disorder: beyond segregated cor-
2 32 10 48 180 tico-striatal pathways. Trends Cogn Sci 2012; 16: 43–51
[5] Abelson JL, Curtis GC, Sagher O, et al. Deep brain stimulation for refractory
3 33 22 48 180 obsessive-compulsive disorder. Biol Psychiatry 2005; 57: 510–516
[6] Jenike MA, Breiter HC, Baer L, et al. Cerebral structural abnormalities in ob-
4 34 21 48 180 sessive-compulsive disorder. A quantitative morphometric magnetic reso-
nance imaging study. Arch Gen Psychiatry 1996; 53: 625–632
5 32 40 48 180
[7] Le Jeune F, Vérin M, N’Diaye K, et al. French Stimulation dans le trouble obses-
sionnel compulsif (STOC) study group. Decrease of prefrontal metabolism
Rück et al, 200825 1 28 24 156 200
after subthalamic stimulation in obsessive-compulsive disorder: a positron
2 36 5 144 200 emission tomography study. Biol Psychiatry 2010; 68: 1016–1022
[8] Robinson D, Wu H, Munne RA, et al. Reduced caudate nucleus volume in ob-
3 38 28 84 180 sessive-compulsive disorder. Arch Gen Psychiatry 1995; 52: 393–398
[9] Saxena S, Brody AL, Maidment KM, et al. Localized orbitofrontal and sub-
4 35 1 180 200 cortical metabolic changes and predictors of response to paroxetine treat-
ment in obsessive-compulsive disorder. Neuropsychopharmacology 1999;
5 27 15 180 200
21: 683–693
6 33 1 144 200 [10] Szeszko PR, MacMillan S, McMeniman M, et al. Brain structural abnormalities
in psychotropic drug-naive pediatric patients with obsessive-compulsive dis-
7 39 35 12 180 order. Am J Psychiatry 2004; 161: 1049–1056
[11] Rauch SL, Jenike MA. Neurobiological models of obsessive-compulsive disor-
8 30 9 96 180 der. Psychosomatics 1993; 34: 20–32
[12] Saxena S, Rauch SL. Functional neuroimaging and the neuroanatomy of ob-
Kondziolka et al23 1 34 24 55 140
sessive-compulsive disorder. Psychiatr Clin North Am 2000; 23: 563–586
[13] Aouizerate B, Guehl D, Cuny E, et al. Pathophysiology of obsessive-compulsive
2 39 8 42 140
disorder: a necessary link between phenomenology, neuropsychology, im-
3 39 18 28 150 agery and physiology. Prog Neurobiol 2004; 72: 195–221
[14] Atmaca M. Review of structural neuroimaging in patients with refractory ob-
4 22 8 14 140 sessive-compulsive disorder. Neurosci Bull 2011; 27: 215–220
[15] Mindus P, Rasmussen SA, Lindquist C. Neurosurgical treatment for refractory
5 35 30 22 140 obsessive-compulsive disorder: implications for understanding frontal lobe
function. J Neuropsychiatry Clin Neurosci 1994; 6: 467–477
Slavin et al, 200921 1 34 21 8 180
[16] Lévêque M, Carron R, Régis J. Radiosurgery for the treatment of psychiatric
2 22 15 8 180 disorders: a review. World Neurosurg 2013; 80: e1–e9
[17] Goodman WK, Price LH, Rasmussen SA, et al. The Yale-Brown Obsessive Com-
3 27 27 8 180 pulsive Scale. II. Validity. Arch Gen Psychiatry 1989; 46: 1012–1016
[18] Goodman WK, Price LH, Rasmussen SA, et al. The Yale-Brown Obsessive Com-
4 37 30 8 180 pulsive Scale: I. Development, use, and reliability. Arch Gen Psychiatry 1989;
46: 1006–1011
Sheehan et al, 1 31 12 15 140 [19] Mindus , Jenike MA. Neurosurgical treatment of malignant obsessive compul-
201324 sive disorder. Psychiatr Clin North Am 1992; 15: 921–938
[20] Lopes AC, Greenberg BD, Norén G, et al. Treatment of resistant obsessive-
2 34 13 6 140
compulsive disorder with ventral capsular/ventral striatal gamma capsuloto-
3 33 31 24 140 my: a pilot prospective study. J Neuropsychiatry Clin Neurosci 2009; 21:
381–392
4 32 13 33 160 [21] Slavin KJL, Mcgrath P, Bangera N, D’Agostino A. Radiosurgical capsulotomy for
refractory obsessive-compulsive disorder: structural and functional consider-
5 31 12 33 160 ations. Paper presented at: Joint Annual Meeting EANS-SFNC; March 27–31,
2009; Marseille, France
Abbreviation: Y-BOCS, Yale-Brown Obsessive-Compulsive Scale.
[22] Gouvea F, Lopes A, Greenberg B, et al. Response to sham and active gamma
ventral capsulotomy in otherwise intractable obsessive-compulsive disorder.
Stereotact Funct Neurosurg 2010; 88: 177–182
[23] Kondziolka D, Flickinger JC, Hudak R. Results following gamma knife radio-
placement or relapsing symptoms with device failure or mal- surgical anterior capsulotomies for obsessive compulsive disorder. Neurosur-
gery 2011; 68: 28–32, discussion 23–3
function. Gamma Knife radiosurgery remains an important
[24] Sheehan JP, Patterson G, Schlesinger D, Xu Z. knife surgery anterior capsulot-
treatment option for refractory OCD and may be preferred in omy for severe and refractory obsessive-compulsive disorder. J Neurosurg
patients with excoriation disorders or medical comorbidities. 2013; 119: 1112–1118
[25] Rück C, Karlsson A, Steele JD, et al. Capsulotomy for obsessive-compulsive dis-
order: long-term follow-up of 25 patients. Arch Gen Psychiatry 2008; 65:

References [26]
914–921
Blomstedt P, Sjöberg RL, Hansson M, Bodlund O, Hariz MI. Deep brain stimu-
[1] American Psychiatric Association and DSM-5 Task Force. Diagnostic and Stat- lation in the treatment of obsessive-compulsive disorder. World Neurosurg
istical Manual of Mental Disorders: DSM-5. 5th ed. Arlington, VA: American 2013; 80: e245–e253
Psychiatric Association; 2013 [27] Greenberg BD, Gabriels LA, Malone DA Jr, et al. Deep brain stimulation of the
[2] Torres AR, Ramos-Cerqueira AT, Ferrão YA, Fontenelle LF, do Rosário MC, Mi- ventral internal capsule/ventral striatum for obsessive-compulsive disorder:
guel EC. Suicidality in obsessive-compulsive disorder: prevalence and relation worldwide experience. Mol Psychiatry 2010; 15: 64–79

180
Gamma Knife Radiosurgery for Obsessive–Compulsive Disorder

[28] Mallet L, Polosan M, Jaafari N, et al. STOC Study Group. Subthalamic nucleus [32] Huff W, Lenartz D, Schormann M, et al. Unilateral deep brain stimulation of
stimulation in severe obsessive-compulsive disorder. N Engl J Med 2008; the nucleus accumbens in patients with treatment-resistant obsessive-com-
359: 2121–2134 pulsive disorder: outcomes after one year. Clin Neurol Neurosurg 2010; 112:
[29] Greenberg BD, Malone DA, Friehs GM, et al. Three-year outcomes in deep 137–143
brain stimulation for highly resistant obsessive-compulsive disorder. Neuro- [33] Jiménez-Ponce F, Velasco-Campos F, Castro-Farfán G, et al. Preliminary study
psychopharmacology 2006; 31: 2384–2393 in patients with obsessive-compulsive disorder treated with electrical stimu-
[30] Denys D, Mantione M, Figee M, et al. Deep brain stimulation of the nucleus lation in the inferior thalamic peduncle. Neurosurgery 2009; 65 (Suppl):
accumbens for treatment-refractory obsessive-compulsive disorder. Arch Gen 203–209, discussion 209
Psychiatry 2010; 67: 1061–1068 [34] Kondziolka D, Hudak R. Management of obsessive-compulsive disorder-re-
[31] Plewnia C, Schober F, Rilk A, et al. Sustained improvement of obsessive-com- lated skin picking with gamma knife radiosurgical anterior capsulotomies: a
pulsive disorder by deep brain stimulation in a woman with residual schizo- case report. J Clin Psychiatry 2008; 69: 1337–1340
phrenia. Int J Neuropsychopharmacol 2008; 11: 1181–1183

181
Stereotactic Radiosurgery for Hypothalamic Hamartomas

23 Stereotactic Radiosurgery for Hypothalamic


Hamartomas
Andrew G. Shetter, Heyoung L. McBride, and John F. Kerrigan

oligodendrocytes. The more numerous small neurons were


Key Points arranged in nodular clusters of varying size and density, inter-
mingled with and surrounded by the less numerous larger neu-
● Hypothalamic hamartomas are rare, nonneoplastic lesions rons. Wu and colleagues have shown that the small neurons are
that may produce intractable gelastic and generalized seiz- γ-Aminobutyric acid (GABA) ergic, and exhibit spontaneous
ures, cognitive and behavioral deterioration, and precocious pacemaker-like firing.5 Large HH neurons, which are excitatory
puberty. projection-type neurons, have the functionally immature prop-
● Stereotactic radiosurgery results in seizure control rates of erty of depolarizing and firing in response to GABA ligands. 6,7
50 to 60% with a very low incidence of complications com- Depth electrode recordings by Munari et al first demonstrated
pared with more invasive surgical alternatives. that the ictal discharges resulting in gelastic seizures arise from
● The principle disadvantage of radiosurgery is the long la- the hamartoma itself.8 This observation has been duplicated by
tency interval before a full therapeutic effect is achieved, other investigators and confirms the intrinsic epileptogenicity
which may be as long as 2 to 3 years. of HHs. Based on these and other studies, Fenoglio et al have
● A radiation dose of > 17 Gy to the margin of the hamartoma proposed a model whereby the small spontaneously firing
seems to be necessary for optimal results while limiting the GABAergic neurons synchronize the output of the normally qui-
dose to the optic apparatus to < 10 Gy. escent larger pyramidal neurons.9 These in turn likely project to
● A multidisciplinary team approach is mandatory to deter- extrahypothalamic sites such as the thalamus and hippocampus
mine the best treatment option for any given individual to produce clinically apparent seizures (▶ Fig. 23.1).
based on lesion size, topological location, and a full assess-
ment of the patient’s overall neurologic, behavioral, and cog-
nitive status. 23.3 Magnetic Resonance Imaging
Findings
Hypothalamic hamartomas are well visualized on high-resolu-
23.1 Introduction tion, thin-slice magnetic resonance images (MRIs), particularly
Hypothalamic hamartomas (HHs) are rare congenital lesions sagittal and coronal T2 fast spin-echo (FSE) images. Computed
that form on the tuber cinereum and inferior hypothalamus. tomography (CT) may demonstrate larger lesions, but is not ad-
The most common presenting symptoms are medically intract- equate to exclude small hamartomas. When compared with
able gelastic seizures, developmental delays, precocious pub- normal gray matter, HHs are usually hyperintense on T2-
erty, and behavioral disturbances. Their true incidence is weighted images, and hypointense or isointense on T1 stud-
unknown, but the prevalence of epilepsy associated with HHs ies.10 In the absence of prior treatment, they rarely enhance in
has been estimated to be 1 in 200,000 children and adoles- response to the administration of intravenous gadolinium con-
cents.1 As true hamartomas, they represent an aberrant devel- trast, and any sign of enhancement should raise suspicion of an
opment of normal tissues. They are nonneoplastic with no alternative diagnosis.
potential for growth after birth beyond that seen in normal
brain development. Hypothalamic hamartomas are usually
sporadic in nature and their exact embryological cause is uncer- 23.4 Classification
tain. Approximately 5% of patients with HHs have genetic
Based on MRI findings, classification schemes for HHs have
syndromes such as Pallister-Hall syndrome, Bardat-Biedl syn-
been proposed by several authors. A comparison of these differ-
drome, Waardenburg syndrome, and oral–facial–digital syndro-
ent approaches is nicely summarized in an article by Mittal et
me. Approximately 15% of sporadic HH patients have somatic
al.11 Arita et al distinguished intrahypothalamic hamartomas
(tumor only) mutations within GLI3, the gene that is also
that have a complete or partial base of attachment within the
responsible for Pallister-Hall syndrome.2 Despite their rarity,
third ventricle from parahypothalamic hamartomas, which are
HHs frequently give rise to an important epilepsy syndrome
attached only to the floor of the third ventricle.12 The most fre-
that can have devastating consequences, is usually refractory to
quently cited classification system is that of Delalande and Foh-
drug therapy, and may be cured by surgical treatment including
len (▶ Fig. 23.2).13 Under this classification, type I lesions are
radiosurgery.3
those that arise below the third ventricle with a horizontal
plane of attachment to the hypothalamus. The attachment may
23.2 Histology and Physiology sometimes be narrow (pedunculated) or in other instances
broader (sessile). Type II lesions are completely within the third
Coons et al characterized the histopathological findings in 57 ventricle and have a vertical plane of attachment to the hypo-
surgically resected HHs.4 All lesions were composed of histo- thalamus, usually clearly lateralized to either the right or left
logically normal small and large neurons, astrocytes, and side. Type III lesions occur above and below the floor of the

182
Stereotactic Radiosurgery for Hypothalamic Hamartomas

Fig. 23.1 Representation of cellular model for intrinsic epileptogenesis of hypothalamic hamartoma tissue. (a) The photomicrograph shows a cluster
of small neurons with hematoxylin and eosin staining. A single small neuron is indicated by the arrow, with loose nuclear material and dense nucleolus.
(b) The diagram represents a cluster of small neurons and two adjacent large neurons. Small neurons have intrinsic pacemaker-like firing and express
GABA as their main neurotransmitter. Large neurons, which are likely excitatory projection-type cells, have the functionally immature property of
depolarizing and firing in response to GABA. The network relationships between these cells are only partially understood. (Used with permission from
Barrow Neurological Institute.)

third ventricle and have both vertical and horizontal attach- abnormalities.”16 They are typically brief, lasting seconds, but
ments to the hypothalamus. Type IV hamartomas are “giant” may occur repeatedly throughout the day. Gelastic seizures
with broad attachments to the hypothalamus and mammillary usually begin during infancy or childhood, although they may
bodies, and frequently extend into the interpeduncular cistern. not be recognized as such until later in life. Some patients have
Régis et al have advocated a somewhat different classification clinical events that more closely resemble crying than laughing;
scheme, which they feel helps to predict the most appropriate these events have been termed dacrystic seizures. Over time, a
surgical strategy (▶ Fig. 23.3).14 Régis type I lesions are located majority of patients develop other seizure types, including com-
inside the hypothalamus with a small projection into the third plex partial seizures, generalized tonic-clonic seizures, tonic
ventricle. Type II lesions are predominately within the third seizures, and drop attacks. In older individuals, these alternate
ventricle and have a vertical attachment to the wall of the third seizure types may predominate, and gelastic seizures may
ventricle. Type III abnormalities are located in the floor of the become less frequent or stop all together. These observations
third ventricle with close proximity to the mammillary bodies have given rise to the concept of secondary epileptogenesis, in
and the columns of the fornix. Type IV lesions are located in the which the presence of one seizure focus causes the emergence
interpeduncular cistern with a small sessile attachment to the of an independent seizure focus from a distant and previously
hypothalamus. Type V lesions are pedunculated, and connect to normal brain region.17 Secondary epileptogenesis may be rever-
the inferior hypothalamus by a thin stalk. Type VI abnormalities sible when the original focus in the HH is excised or discon-
are giant lesions that extend both above and below the floor of nected, or it may become permanent over time. This concept
the third ventricle. Hypothalamic hamartoma lesions that com- may explain why some HH patients with an apparently com-
bine features from types I to VI are classified as “Mixed” within plete surgical resection of their lesion continue to have seiz-
the Régis classification system. ures.18 The success rate of HH treatment for controlling seizures
has an inverse correlation with patient age and lifetime dura-
tion of epilepsy at the time of surgery.19,20
23.5 Natural History A majority of patients with intrahypothalamic HHs demon-
Gelastic seizures are the hallmark of epilepsy produced by HHs. strate significant cognitive impairment. Prigatano et al per-
Trousseau in 1887 first recognized the possible epileptic nature formed neuropsychological assessments on 49 patients with
of compulsive bursts of laughter.15 Gascon and Lombroso intractable seizures who were undergoing a presurgical evalua-
defined gelastic seizure as “stereotypic recurrence of ictal tion and who varied in age from 5 to 55 years.21 Of the total
laughter, inadapted to context, associated with other signs group, 35% had no significant deficits and were functioning at a
compatible with seizure and with ictal/interictal EEG normal or near-normal intellectual level, 18% had deficits in

183
Stereotactic Radiosurgery for Hypothalamic Hamartomas

Fig. 23.2 Hypothalamic hamartoma classification


system as proposed by Delalande and Fohlen.
Type I lesions have a horizontal base of attach-
ment below the plane of the floor of the third
ventricle. Type II lesions have a vertical base of
attachment above the floor of the third ventricle.
Type III lesions are typically larger, with attach-
ment both above and below the floor of the third
ventricle (or the normal position of the floor, if it
can no longer be visualized). Type IV lesions are
characterized as “giant” lesions, although the size
boundary between III and IV remains undefined.
Types III and IV often have bilateral attachment to
the hypothalamus. (Used with permission from
the Barrow Neurological Institute.)

certain test subscores but were considered to be within the nor- connected to the inferior aspect of the hypothalamus (parahy-
mal range of overall intelligence, and 47% were intellectually pothalamic lesions of Arita et al and type V lesions of Régis
disabled. Despite the prevalence of cognitive and developmen- et al)12,14 usually have precocious puberty as their only clinical
tal delays, not all patients were affected, and there was a con- manifestation, and are rarely associated with epilepsy, cognitive
tinuous spectrum varying from severe retardation to those with impairment, or behavioral abnormalities. They can be treated
normal or above-average intelligence. medically with gonadotropin-releasing hormone agonists and
Many patients with HHs and intractable seizures also exhibit usually do not require surgery. Parvizi et al analyzed MRIs in
behavioral and emotional difficulties. These include affective 100 patients with gelastic seizures23; Chan et al reviewed scans
disorders, emotional lability, rage attacks, temper tantrums, in seven patients with precocious puberty.24 These two studies
and destructive or aggressive behavior directed against them- indicate that patients whose lesions are attached to the anterior
selves or others.22 This can be a source of extreme frustration region of the hypothalamus exhibit precocious puberty, where-
for parents and other family members. As with cognitive func- as those whose lesions are attached only to the posterior hypo-
tion, there is a wide range in the degree to which behavioral thalamus near the mammillary bodies have epilepsy alone.
and mood issues manifest themselves in any given patient. Patients with large hamartomas with anterior and posterior
Some infants and young children who present with gelastic hypothalamic attachment often have both epilepsy and preco-
seizures will develop worsening seizures and progressive intel- cious puberty.
lectual and behavioral deteriorations as they mature. This
downhill course has been termed epileptic encephalopathy. Both
secondary epileptogenesis and epileptic encephalopathy are
strong reasons to consider early surgical intervention in HH
23.6 Surgical Treatment
patients with intractable seizures before irreversible changes Because seizures resulting from HHs are almost always refrac-
occur.18 tory to medical therapy and may over time lead to secondary
Central precocious puberty may be present in as many as 40 epileptogenesis and epileptic encephalopathy, surgery is war-
to 50% of patients with HHs.3 Pedunculated hamartomas ranted in all but the mildest forms of this disorder. This is true

184
Stereotactic Radiosurgery for Hypothalamic Hamartomas

Fig. 23.3 Hypothalamic hamartoma classification system proposed by Régis. Type I lesions are located inside the hypothalamus with a small projection
into the third ventricle. Type II lesions are predominately within the third ventricle and have a vertical attachment to the wall of the third ventricle. Type
III abnormalities are located in the floor of the third ventricle with close proximity to the mammillary bodies and the columns of the fornix. Type IV
lesions are located below the floor of the third ventricle with a broad attachment to the hypothalamus. Type V lesions are pedunculated, and connect
to the inferior hypothalamus by a thin stalk. Type VI abnormalities are giant lesions that extend both above and below the floor of the third ventricle.
Hypothalamic hamartoma lesions that combine features from types I to VI are classified as “Mixed” within the Régis system. (Used with permission
from the Barrow Neurological Institute.)

185
Stereotactic Radiosurgery for Hypothalamic Hamartomas

despite the significant risks of surgical intervention. There are years. A complete lesion resection was obtained in 12 patients.
no randomized studies evaluating the various surgical Seizures were reduced by more than 90% in 70% of patients,
approaches that have been utilized, and given the rarity of HHs, and 49% of patients were seizure-free. Postoperative MRIs
evidence of this type is unlikely to be available in the foresee- showed small thalamic infarcts in 11 patients, although only 2
able future.3 Comparing uncontrolled surgical series with one were symptomatic with transient hemiparesis. The incidence of
another is made difficult by the often significant differences in permanent short-term memory impairment was 8%.
lesion size, anatomical location, patient age, and preoperative Pterional or orbitozygomatic approaches to intrahypothala-
neurologic status. For these reasons, dogmatic statements mic hamartomas result in poor seizure control rates,28 but they
about the “best” surgical option for HHs are not appropriate. In may be helpful for lesions with an inferior or horizontal plane
general, open surgical approaches for lesions within the third of attachment to the hypothalamus (Delalande type I). Abla et
ventricle are done from above, through the third ventricle. al reported their results in 10 such patients, 2 of whom had
Hamartomas attached to the floor of the hypothalamus can be combined orbitozygomatic and transventricular procedures for
accessed from below using pterional, orbitozygomatic, or sub- hamartomas both above and below the floor of the third ven-
frontal approaches. Larger lesions that have planes of attach- tricle.29 Complete seizure control was achieved in four patients
ment both within and below the third ventricle may require (40%) and seizure frequency decreased by more than 50% in an
combined approaches from above and below. When feasible, additional four patients (40%). Residual complications included
complete surgical resection is the optimal strategy, but good diabetes insipidus, visual deficit, and hemiparesis in one patient
seizure control can also be achieved by disconnecting the lesion each.
from the hypothalamus, even though a portion of the hamarto- Many investigators have noted a higher rate of complications
ma remains in place. Stereotactic approaches, including radio- and a lower degree of seizure control in older patients com-
surgery, are usually confined to abnormalities less than 2 to pared with pediatric patients undergoing open surgical resec-
3 cm in maximum diameter. tion. For example, Drees et al examined 40 patients 18 years of
age or older who underwent endoscopic, transcallosal, orbito-
zygomatic, or Gamma Knife stereotactic radiosurgery (GKSRS)
23.6.1 Open Surgical Procedures of their lesions.30 There were two postoperative deaths, and
Rosenfeld and his colleagues in Melbourne pioneered the use of one patient suffered a permanent hemianopia. Persistent hor-
a transcallosal anterior interforniceal approach for intrahypo- monal deficits were seen in 34% of patients, and 59% experi-
thalamic hamartomas.25 Their experience involved 29 patients enced a postoperative weight gain that averaged 12.7 kg. Only
ranging in age from 4 to 23 years, who were followed for a min- 29% of the total group became seizure-free, and 55% had a
imum of 12 months. A complete or near-complete resection greater than 90% seizure improvement. Of the four patients
was obtained in 62%, and a 75 to 95% resection was achieved in who elected radiosurgical treatment, two became seizure-free
an additional 24%. All patients exhibited both gelastic and other and none experienced a permanent complication.
seizure types preoperatively, and 52% became seizure-free after
surgery. Another 24% had a greater than 90% reduction in seiz-
ure frequency. Due to the surgical dissection and retraction
23.6.2 Stereotactic Procedures
required to visualize the third ventricle, the transcallosal A variety of stereotactic procedures for treating HHs have been
approach may potentially injure the columns of the fornix and developed in the hope that these less invasive techniques might
therefore cause short-term memory impairment. In the Rose- offer a lower risk alternative to open surgical resection.
nfeld series, 14% of patients had permanent short-term memo- Schulze-Bonhage et al treated 24 patients with gelastic epi-
ry deficits, and nearly 50% had temporary impairments. Other lepsy by stereotactically implanting a single radioactive seed
significant complications included hypernatremia secondary to into hamartomas with a mean volume of 1.2 cc, a procedure
syndrome of inappropriate antidiuretic hormone secretion they called interstitial radiosurgery.20 At a mean follow-up of 24
(SIADH), which was transient in all cases, and hyperphagia with months, 46% of patients were seizure-free or had > 90%
weight gain, which was permanent in 17%. decrease in the seizure frequency. More than half of their
Ng et al reported on 26 patients who underwent transcallosal patients (54%) underwent a second seed implant from 1 to 2.4
interforniceal HH resections at Barrow Neurological Institute years after the initial treatment. Permanent complications
(Phoenix, AZ), with findings similar to those of the Rosenfeld included memory decline in two patients and weight gain of
group.19 Complete seizure control was achieved in 54% of their more than 5 kg in four patients.
patients, and an additional 35% had at least 90% fewer seizures. Kameyama et al treated 24 patients with HHs and gelastic or
Persistent short-term memory disturbances were seen in 8% of mixed epilepsy by stereotactically inserting radiofrequency
their patients, and transient deficits in 58%. Other permanent (RF) thermocoagulation probes into the lesions.31 The electro-
complications included diabetes insipidus, hypothyroidism, des had an uninsulated tip 2 mm in length and were heated to a
hemianopsia, and communicating hydrocephalus (occurring in temperature of 74°C for 60 seconds. Multiple tracks (mean 3.8,
one patient each). range 1–8) and multiple lesions along the same track were used
Fohlen et al were among the first to advocate a transventricu- in most instances. Complete seizure freedom was achieved in
lar endoscopic approach to HHs, either to resect them com- 76% of their patients, and no long-term complications were
pletely or to disconnect their hypothalamic attachment. 26 Ng et noted. These outcomes are superior to any reported for open
al have reported the largest series using this technique.27 Forty- surgical procedures. They are particularly noteworthy because
two patients were treated and 37 were available for long-term of the size of the treated hamartomas (mean diameter 15.3 mm,
follow-up; the age range for this cohort was 8 months to 55 range 8–30 mm), and the apparent absence of any permanent

186
Stereotactic Radiosurgery for Hypothalamic Hamartomas

hemorrhagic complications despite the large number of elec- Complete seizure control was obtained in 37% of their
trode tracts employed. No other investigators have duplicated patients, and 22% had only rare nondisabling seizures. Nearly
this very favorable experience in a similar number of patients. all patients exhibited improvement in behavior, school per-
Recently, there has been considerable interest in the use of formance, and quality of life (QoL) as determined by parental or
stereotactic laser ablation (also called laser interstitial thermal patient reports and the subjective observations of the treating
therapy) to treat HHs. For this procedure, a thin diode laser group. Postoperative MRIs showed that the lesions were
catheter is inserted into the hamartoma using MRI and stereo- unchanged in the majority of patients, with two patients show-
tactic techniques. Laser energy deposited into the surrounding ing a slight decrease in the size of their lesion. Repeat radiosur-
tissues heats the tissues locally and results in necrosis by ther- gery was performed in nine patients who did not experience
mocoagulation. The extent of the thermal injury zone can be complete seizure abatement after 3 years, although no details
viewed in near real time using MRI thermography, and a feed- were given about the radiation doses utilized for these second
back loop can monitor temperatures in adjacent structures such procedures.
as the optic chiasm or mamillary bodies to shut off the laser if The main drawback of SRS noted in this and all other series is
predetermined temperatures are exceeded. Laser stereotactic the long latency period between treatment and the final effect.
ablation has the potential to be less invasive and have a lower Régis et al described a typical five-stage response pattern
risk than open surgery, be more precise than RF thermal abla- occurring over a minimum of 2 years.14,34 In the first phase,
tion, and produce a more rapid therapeutic response than SRS. patients experienced an immediate mild improvement in
Curry and his colleagues have accumulated the largest expe- behavior, and seizure frequency and severity. By an average of 2
rience using stereotactic laser ablation for HHs, although a to 6 months posttreatment, seizures had returned to their pre-
number of other centers are also actively investigating this operative state, although they were sometimes briefer in dura-
technique.32 They have reported 14 patients with medically tion. During the third phase, seizures suddenly increased in
intractable gelastic epilepsy treated with stereotactic laser abla- frequency. This period lasted several days up to 1 month, and
tion.31 Seizure freedom was achieved in 86% with a mean fol- was followed by a fourth phase in which seizures gradually
low-up of 9 months. A single patient sustained an diminished. The fifth phase was considered a consolidation
asymptomatic subarachnoid hemorrhage, but no permanent period leading to complete seizure freedom. Although there
neurologic or endocrine deficits were seen. Despite these prom- were individual variations in these patterns, patients or their
ising initial results, not enough patients have been treated to parents need to be aware that seizures may increase in fre-
date to assess the true complication rate of this procedure, and quency before they eventually subside, and that 2 to 3 years
the published follow-up intervals are still too short to deter- may be required to see the full effects of radiosurgical
mine the incidence of long-term seizure control. treatment.
The Marseille group emphasized the use of their topologi-
cal classification system to determine which patients were
Stereotactic Radiosurgery
best treated with GKSRS versus surgical options. They felt
Hypothalamic hamartomas are appealing targets for SRS that type I and type II lesions were the optimal candidates
because of their small size; their distinct borders, which can be for radiosurgery because of its superior safety profile com-
precisely imaged on MRIs; and their deep-seated locations, pared with endoscopic or interforniceal transcallosal resec-
which make open surgical approaches difficult. In 1998, Arita et tion. They also preferred radiosurgery for small- to medium-
al were the first to describe the successful Gamma Knife radio- size type III abnormalities unless seizures were rapidly pro-
surgical treatment of gelastic epilepsy in a 25-year-old man gressive. Type IV and type V (rarely epileptogenic) hamarto-
with a small hamartoma.33 Their patient was seizure-free at a mas could be treated with either GKSRS, or pterional or
21-month follow-up interval and sustained no new neurologic orbitozygomatic disconnections, depending on the size of the
or endocrine deficits. Interestingly, an MRI 12 months post- lesions and the severity of the seizures. They did not recom-
treatment showed complete disappearance of the lesion— mend radiosurgery as a primary treatment for type VI
something that has not been reported in subsequent series. lesions, but they did advocate it as a second procedure after
The acknowledged leaders in this field are Jean Régis and surgical resection if some hamartoma remained and seizures
his coworkers in Marseille, France. They have reported 60 persisted.
patients with HHs treated with a 201-source cobalt-60 Gam- Several other centers have reported their experience using
ma Knife unit.14,34 Twenty-seven patients were available for the Gamma Knife for HHs in smaller series of patients, with
detailed prospective analysis with a minimum of 3-year fol- results that generally parallel those of Régis et al (▶ Table 23.1).
low-up. In this group, median age at the time of treatment Abla et al described 19 patients treated at the Barrow Neurolog-
was 17.25 years (range 3–50 years), and the median maxi- ical Institute from 2003 to 2010.35 Ten patients were available
mum lesion diameter was 11.7 mm (range 7.2–25 mm). The for long-term follow-up (mean 43 months, range 18–81
median radiation dose delivered to the margins of the hamar- months) sufficient to permit an assessment of their outcomes.
tomas was 17 Gy (range 13–26 Gy), usually prescribed to the The mean age in this group was 15.1 years (range 5.7–29.3
50% isodose line. Beam-blocking strategies were used to mini- years), and the mean lesion volume was 695 mm3 (range 169—
mize dose to the optic pathways, mammillary bodies, and for- 3000 mm3). The median margin dose prescribed to the 50% iso-
nices. The median maximum doses to the optic chiasm, optic dose line was 18 Gy (range 16–20 Gy). Six patients became free
tract, and mammillary bodies were 3.7 Gy (range 3–12.2 Gy), of seizures, although two of these patients underwent endo-
6.4 Gy (range 3–12.2 Gy), and 18.6 Gy (range 0.9– 2.2 Gy), scopic resections 14.5 and 32 months after their GKSRS. Tele-
respectively. phone interviews or patient surveys documented improvement

187
Stereotactic Radiosurgery for Hypothalamic Hamartomas

Table 23.1 Hypothalamic hamartoma treatment with Gamma Knife radiosurgery


Study N Age (y) Margin dose Lesion volume Follow-Up Outcome Side effects
(Gy) (mm3) (mo)

Régis et al, 200614 27a 17.25 17 646.7 > 36 37% Seizure-free; 11% transient poikilother-
(Median) (Median) (Median) 22% rare seizure; mia
improved behavior
60%

Abla et al, 201029 10b 15.1 18 695 43 60% Seizure-free 1 Patient transient poikilo-
(Mean) (Mean) (Mean) (Mean) thermia; 2 patients tempo-
rary weight gain

Barajas et al, 200538 3 3, 12, 15 12.5, 14, 15 530, 591, 1800 30–50 All improved, none None
seizure-free

Mathieu et al, 200637 4 5–29 17.5 370 22 2/4 Rare seizures None
(Mean) (Mean) (Median)

Mathieu et al, 201036 9 12–57 14–20 300–1800 6–56 4/9 Seizure-free; None
(Range) 1/9 rare seizure;
improved verbal
memory and QoL

Abbreviation: QoL, quality of life.


aSixty patients were followed prospectively; 27 with > 3-year follow-up data.
bTwenty-six patients treated; 10 with adequate follow-up data.

in overall QoL in nine patients, and improvement of behavioral Gamma Knife series referred to above, there were no instances
symptoms in five patients. of death, visual deficits, cognitive or short-term memory
Mathieu et al treated six patients with small HHs (with a vol- decline, endocrine dysfunction, permanent weight gain, or
ume of 0.3 to 1.0 cc) using margin doses of 14 to 20 Gy. 36 Four paresis. All of these complications have been described to some
of these became seizure-free, and an additional patient had degree in surgical series using endoscopic, transcallosal, and
seizures only rarely after treatment. They also treated three pterional approaches. Transient morbidity has been seen in a
larger lesions (Régis type IV or VI) by targeting their area of few patients undergoing radiosurgical treatment and has
attachment to the hypothalamus in an attempt to produce a included poikilothermia (four patients) and hyperphagia with
radiosurgical disconnection. None of these patients had worth- weight gain (two patients). The absence of permanent short-
while seizure reduction, and they did not recommend this term memory loss following GKSRS is a particularly important
strategy for future use. consideration in older patients or those functioning at a normal
The University of Pittsburgh (Pittsburgh, PA) group37 treated level preoperatively.
four patients with HHs varying from 0.20 to 0.55 cc in volume A single institution using stereotactic RF thermocoagulation
with margin doses of 16 to 20 Gy. No individuals became com- has also reported no long-term cognitive or endocrine deficits
pletely seizure-free, but two exhibited rare persistent seizures using their technique.31 Complication rates following stereotac-
following radiosurgery. tic laser ablation have been low as well, but experience with
Barajas et al reported their results in three HH patients using this procedure is still preliminary.32
lower margin radiation doses (12.5, 14, and 15 Gy) than other Visual loss as a side effect of GKSRS for HHs has not been
centers.38 All patients had cessation of their tonic–clonic seiz- reported to date, but this is a potential concern because of the
ures; gelastic seizures persisted, although at a reduced rate. often close proximity of these lesions to the optic chiasm and
A small number of HH patients have received SRS using lin- optic tracts and the known radiosensitivity of the optic path-
ear accelerator (linac)-based devices rather than the Gamma ways. In patients who have not had prior external beam radia-
Knife. Selch et al treated three patients using 15 to 18 Gy mar- tion therapy, the risk of radiation-induced optic neuropathy
gin doses prescribed to the 90 to 95% isodose line.39 With fol- following SRS is 0 to 2% if dose to the optic apparatus is 10 Gy or
low-ups of 9 to 17 months, two patients were free of seizures less, and still low at a dose of 12 Gy or less.41 These limits need
and the third had only rare seizures. Papayannis et al treated to be respected when treating HHs, even if to do so may result
four patients with doses of 12 to 18 Gy, none of whom had in slight under treatment of the lesion itself.
any worthwhile seizure reduction.40 There is insufficient infor- The possibility that ionizing radiation may induce secondary
mation at present to judge whether linac-based radiosurgery benign or malignant tumors is always a concern when treating
can produce comparable results to those documented for children or young adults with an anticipated life span of many
GKSRS. decades. There are radiobiological reasons to believe the inci-
dence of secondary oncogenesis will be much lower using
single-session SRS than that reported for conventional fractio-
23.6.3 Radiosurgical Complications nated radiotherapy.42 Lunsford has estimated the risk of devel-
The outstanding advantage of GKSRS for HHs is its very low oping a secondary tumor after radiosurgery at < 1 in 20,000
complication rate relative to open surgical resection. In the five treated patients.43 Despite the unlikelihood of such an event,

188
Stereotactic Radiosurgery for Hypothalamic Hamartomas

patients or their parents need to be informed of this possible Although successful seizure control has been reported
complication. with lower doses, most centers have advocated a HH mar-
gin dose of 18 Gy or higher if at all possible. There is still
some uncertainty about the optimal treatment dose for
23.6.4 Gamma Knife Radiosurgical these lesions that represents the best compromise between
Techniques safety and efficacy. However, maximal dose to the optic
pathways should be kept below 10 Gy. We also try to mini-
With minor modifications, Gamma Knife radiosurgical tech-
mize radiation to the mamillary bodies and fornices,
niques for treating HHs are identical to those for other small
although the radiation tolerance of these structures is not
lesions in vital locations (e.g., vestibular schwannomas). The
well understood.
procedure may need to be performed under general anes-
Examples of typical treatment plans for two HHs treated at
thesia in younger patients at the discretion of their parents
our institution are shown in ▶ Fig. 23.4 and ▶ Fig. 23.5.
and treating physicians. We have found it helpful to angle
the stereotactic frame forward from back to front in an
attempt to place the entire extent of the optic pathways and
23.6.5 Mechanism of Action
the hamartoma in the same imaging plane. Coronal and
axial MRIs are obtained using 1-mm-thick T1 SPGR (spoiled The radiobiology and pathophysiology of the treatment effect
gradient echo) and 2-mm-thick T2 FSE sequences. The optic on HHs treated by GKSRS is not well understood. The neuropa-
pathways and the hamartoma are outlined, and small colli- thology of surgically resected HH tissue in patients with a pre-
mator shots (usually 4 mm) are employed to conform the vious history of GKSRS was compared with surgically resected
50% isodose line as closely as possible to the margins of the tissue from a cohort of patients with no history of radiosur-
lesion. We believe the Leksell Gamma Knife Perfexion unit gery.44 Tissue necrosis was not observed. When examined with
(Elekta Instruments AB, Stockholm, Sweden) has significant thick-section stereology, the total nucleated cell density in
advantages over earlier Gamma Knife models because it GKSRS-treated HH tissue was significantly lower than in
facilitates the use of multiple treatment isocenters and untreated tissue. Total neuron density was lower in a GKSRS-
allows composite shots that enable custom blocking dose-dependent fashion in a manner that approached signifi-
patterns. cance (p = 0.06). Gamma Knife stereotactic radiosurgery-treated

Fig. 23.4 A 39-year-old woman with gelastic seizures since early childhood was undiagnosed until age 21 when she experienced a generalized seizure.
Her generalized seizures were controlled by medications, but she continued to experience multiple daily gelastic seizures. Lesion volume was
297 mm3. She was treated using a margin dose of 18 Gy to the 50% isodose line via six isocenters using 4-mm collimators and custom beam blocking.
The lesion was classified as a Delalande type II. (Used with permission from the Barrow Neurological Institute.)

189
Stereotactic Radiosurgery for Hypothalamic Hamartomas

Fig. 23.5 A 10-year-old boy with daily gelastic seizures since age 5 and rare generalized seizures. His precocious puberty was treated with Lupron
(AbbVie, Inc., North Chicago, IL). He exhibited behavioral difficulties and possible cognitive decline. Lesion volume was 431 mm 3. He was treated using
a margin dose of 18 Gy to the 50% isodose line via 10 isocenters using 4-mm collimators and custom beam blocking. The lesion was classified as a
Delalande type III. (Used with permission from the Barrow Neurological Institute.)

tissue exhibited significant increases in reactive gliosis, endo- severity of the type of seizure and the impact of the seizure
thelial thickening, and microglial activation. These results sug- activity on the patient’s and family’s QoL. When the patient’s
gest that nonnecrotizing doses of GKSRS act in part by seizures are well tolerated and the patient is otherwise stable
mediating cell death, rather than by playing a purely neuromo- with respect to cognitive function and behavior, then the
dulatory role, and that they therefore contribute to the delay in efficacy is acceptable and a treatment decision favor-
decreased excitation in the neuronal network that is responsi- ing GKSRS is appropriate to take advantage of its superior
ble for seizures in HH tissue. safety profile. From our perspective, treatment decisions can
and should be revisited if clinical conditions deteriorate,
favoring the use of one of the surgical therapies with imme-
23.7 Summary diate efficacy (surgical resection/disconnection or lesion
Gamma Knife stereotactic radiosurgery is an effective and safe destruction by stereotactic thermoablation) even if GKSRS
treatment modality for patients with HH associated with was previously performed. Régis et al have advocated a sec-
intractable epilepsy. It should be part of the treatment arma- ond Gamma Knife treatment after 3 years in patients who
mentarium of any center that claims expertise in treating this improve following an initial procedure, but are not com-
disorder. pletely seizure-free. There are no reports from other centers
Hypothalamic hamartomas result in diverse clinical symp- utilizing this strategy; to date we have preferred to consider
toms and a complicated natural history that includes disability alternative approaches if the first Gamma Knife treatment is
and deterioration in most patients with concomitant epilepsy. unsuccessful.
These patients are best managed by a multidisciplinary team We believe that GKSRS is an attractive treatment modality
experienced in managing this rare and clinically significant for those patients who have undergone surgical resection, but
condition. have residual HH tissue. This situation is not uncommon, as the
The treatment algorithm that guides clinical decision mak- surgical margin between normal hypothalamus and HH tissue
ing at our institute is shown in ▶ Fig. 23.6. The clinical feature is not always obvious, even under direct visualization with
most favorable to selecting GKSRS is clinical stability of the assistance from intraoperative neuronavigation systems: The
patient in terms of seizure type and frequency. This is a surgeon should err on the side of conservative (rather than
highly individualized determination that must take into overly aggressive) resection to minimize injury to normal
account not just the absolute number of seizures, but also the hypothalamus.

190
Stereotactic Radiosurgery for Hypothalamic Hamartomas

Fig. 23.6 Treatment algorithm for hypothalamic hamartoma. This algorithm is based upon the surgical anatomy of the hypothalamic hamartoma as
classified by the system proposed by Delalande and Fohlen (types I through IV). 13 This approach is utilized at our institution (Barrow Neurological
Institute, Phoenix, AZ) as a guide to treatment decision making. The algorithm was developed on the basis of expert opinion and the available
literature as discussed in this chapter. Controlled, randomized treatment trials do not exist. Decision making should always be individualized to the
clinical circumstances of each patient and the experience of the local institution. Ideally, patients should be evaluated and treated at hypothalamic
hamartoma referral centers with established multidisciplinary programs. Hypothalamic hamartoma type is based on the Delalande classification.
(Used with permission from the Barrow Neurological Institute.)

[6] Kim Y, Fenoglio KA, Simeone TA, et al. GABAA receptor-mediated activation
References of L-type calcium channels induces neuronal excitation in surgically resected
human hypothalamic hamartomas. Epilepsia 2008; 49: 861–871
[1] Brandberg G, Raininko R, Eeg-Olofsson O. Hypothalamic hamartoma with ge- [7] Wu J, DeChon J, Xue F, et al. GABA(A) receptor-mediated excitation in dissoci-
lastic seizures in Swedish children and adolescents. Eur J Paediatr Neurol ated neurons from human hypothalamic hamartomas. Exp Neurol 2008; 213:
2004; 8: 35–44 397–404
[2] Craig DW, Itty A, Panganiban C, et al. Identification of somatic chromosomal [8] Munari C, Kahane P, Francione S, et al. Role of the hypothalamic hamartoma
abnormalities in hypothalamic hamartoma tissue at the GLI3 locus. Am J in the genesis of gelastic fits (a video-stereo-EEG study). Electroencephalogr
Hum Genet 2008; 82: 366–374 Clin Neurophysiol 1995; 95: 154–160
[3] Pati S, Sollman M, Fife TD, Ng YT. Diagnosis and management of epilepsy as- [9] Fenoglio KA, Wu J, Kim Y, et al. Hypothalamic hamartoma: basic mechanisms
sociated with hypothalamic hamartoma: an evidence-based systematic re- of intrinsic epileptogenesis. Semin Pediatr Neurol 2007; 14: 51–59
view. J Child Neurol 2013; 28: 909–916 [10] Freeman JL, Coleman LT, Wellard RM, et al. MR imaging and spectroscopic
[4] Coons SW, Rekate HL, Prenger EC, et al. The histopathology of hypothala- study of epileptogenic hypothalamic hamartomas: analysis of 72 cases. AJNR
mic hamartomas: study of 57 cases. J Neuropathol Exp Neurol 2007; 66: Am J Neuroradiol 2004; 25: 450–462
131–141 [11] Mittal S, Mittal M, Montes JL, Farmer JP, Andermann F. Hypothalamic hamar-
[5] Wu J, Xu L, Kim DY, et al. Electrophysiological properties of human hypo- tomas. Part 1. Clinical, neuroimaging, and neurophysiological characteristics.
thalamic hamartomas. Ann Neurol 2005; 58: 371–382 Neurosurg Focus 2013; 34: E6

191
Stereotactic Radiosurgery for Hypothalamic Hamartomas

[12] Arita K, Ikawa F, Kurisu K, et al. The relationship between magnetic resonance [30] Drees C, Chapman K, Prenger E, et al. Seizure outcome and complica-
imaging findings and clinical manifestations of hypothalamic hamartoma. J tions following hypothalamic hamartoma treatment in adults: endo-
Neurosurg 1999; 91: 212–220 scopic, open, and Gamma Knife procedures. J Neurosurg 2012; 117:
[13] Delalande O, Fohlen M. Disconnecting surgical treatment of hypothalamic ha- 255–261
martoma in children and adults with refractory epilepsy and proposal of a [31] Kameyama S, Murakami H, Masuda H, Sugiyama I. Minimally invasive mag-
new classification. Neurol Med Chir (Tokyo) 2003; 43: 61–68 netic resonance imaging-guided stereotactic radiofrequency thermocoagula-
[14] Régis J, Scavarda D, Tamura M, et al. Epilepsy related to hypothalamic hamar- tion for epileptogenic hypothalamic hamartomas. Neurosurgery 2009; 65:
tomas: surgical management with special reference to gamma knife surgery. 438–449, discussion 449
Childs Nerv Syst 2006; 22: 881–895 [32] Wilfong AA, Curry DJ. Hypothalamic hamartomas: optimal approach to clini-
[15] Trousseau A. De l’epilepsie. In: Peter MM, ed. Clinique Medicale de l’Hotel- cal evaluation and diagnosis. Epilepsia 2013; 54 (Suppl 9): 109–114
Dieu de Paris. Paris, France: Librarie J. B. Bailliere; 1877 [33] Arita K, Kurisu K, Iida K, et al. Subsidence of seizure induced by stereotactic
[16] Gascon GG, Lombroso CT. Epileptic (gelastic) laughter. Epilepsia 1971; 12: 63–76 radiation in a patient with hypothalamic hamartoma. Case report. J Neuro-
[17] Cibula JE, Gilmore RL. Secondary epileptogenesis in humans. J Clin Neurophy- surg 1998; 89: 645–648
siol 1997; 14: 111–127 [34] Régis J, Scavarda D, Tamura M, et al. Gamma knife surgery for epilepsy related
[18] Kerrigan JF, Ng YT, Chung S, Rekate HL. The hypothalamic hamartoma: a to hypothalamic hamartomas. Semin Pediatr Neurol 2007; 14: 73–79
model of subcortical epileptogenesis and encephalopathy. Semin Pediatr [35] Abla AA, Shetter AG, Chang SW, et al. Gamma Knife surgery for hypothalamic
Neurol 2005; 12: 119–131 hamartomas and epilepsy: patient selection and outcomes. J Neurosurg
[19] Ng YT, Rekate HL, Prenger EC, et al. Transcallosal resection of hypothalamic 2010; 113 (Suppl): 207–214
hamartoma for intractable epilepsy. Epilepsia 2006; 47: 1192–1202 [36] Mathieu D, Deacon C, Pinard CA, Kenny B, Duval J. Gamma Knife surgery for
[20] Schulze-Bonhage A, Trippel M, Wagner K, et al. Outcome and predictors of in- hypothalamic hamartomas causing refractory epilepsy: preliminary results
terstitial radiosurgery in the treatment of gelastic epilepsy. Neurology 2008; from a prospective observational study. J Neurosurg 2010; 113 (Suppl): 215–
71: 277–282 221
[21] Prigatano GP, Wethe JV, Gray JA, et al. Intellectual functioning in presurgical [37] Mathieu D, Kondziolka D, Niranjan A, Flickinger J, Lunsford LD. Gamma knife
patients with hypothalamic hamartoma and refractory epilepsy. Epilepsy Be- radiosurgery for refractory epilepsy caused by hypothalamic hamartomas.
hav 2008; 13: 149–155 Stereotact Funct Neurosurg 2006; 84: 82–87
[22] Weissenberger AA, Dell ML, Liow K, et al. Aggression and psychiatric comor- [38] Barajas MA, Ramírez-Guzman MG, Rodríguez-Vázquez C, Toledo-Buenrostro
bidity in children with hypothalamic hamartomas and their unaffected sib- V, Cuevas-Solórzano A, Rodríguez-Hernández G. Gamma knife surgery for hy-
lings. J Am Acad Child Adolesc Psychiatry 2001; 40: 696–703 pothalamic hamartomas accompanied by medically intractable epilepsy and
[23] Parvizi J, Le S, Foster BL, et al. Gelastic epilepsy and hypothalamic hamarto- precocious puberty: experience in Mexico. J Neurosurg 2005; 102 (Suppl):
mas: neuroanatomical analysis of brain lesions in 100 patients. Brain 2011; 53–55
134: 2960–2968 [39] Selch MT, Gorgulho A, Mattozo C, Solberg TD, Cabatan-Awang C, DeSalles AA.
[24] Chan YM, Fenoglio-Simeone KA, Paraschos S, et al. Central precocious pub- Linear accelerator stereotactic radiosurgery for the treatment of gelastic seiz-
erty due to hypothalamic hamartomas correlates with anatomic features but ures due to hypothalamic hamartoma. Minim Invasive Neurosurg 2005; 48:
not with expression of GnRH, TGFalpha, or KISS1. Horm Res Paediatr 2010; 310–314
73: 312–319 [40] Papayannis CE, Consalvo D, Seifer G, Kauffman MA, Silva W, Kochen S. Clinical
[25] Harvey AS, Freeman JL, Berkovic SF, Rosenfeld JV. Transcallosal resection of spectrum and difficulties in management of hypothalamic hamartoma in a
hypothalamic hamartomas in patients with intractable epilepsy. Epileptic developing country. Acta Neurol Scand 2008; 118: 313–319
Disord 2003; 5: 257–265 [41] Leavitt JA, Stafford SL, Link MJ, Pollock BE. Long-term evaluation of radiation-
[26] Fohlen M, Lellouch A, Delalande O. Hypothalamic hamartoma with refractory induced optic neuropathy after single-fraction stereotactic radiosurgery. Int J
epilepsy: surgical procedures and results in 18 patients. Epileptic Disord Radiat Oncol Biol Phys 2013; 87: 524–527
2003; 5: 267–273 [42] Rowe J, Grainger A, Walton L, Silcocks P, Radatz M, Kemeny A. Risk of malig-
[27] Ng YT, Rekate HL, Prenger EC, et al. Endoscopic resection of hypothalamic hamar- nancy after gamma knife stereotactic radiosurgery. Neurosurgery 2007; 60:
tomas for refractory symptomatic epilepsy. Neurology 2008; 70: 1543–1548 60–65, discussion 65–66
[28] Palmini A, Chandler C, Andermann F, et al. Resection of the lesion in patients [43] Lunsford L.D. In discussion of: Rowe J, Grainger A, Walton L, Silcocks P, Radatz
with hypothalamic hamartomas and catastrophic epilepsy. Neurology 2002; M. Risk of malignancy after Gamma Knife stereotactic radiosurgery. Neuro-
58: 1338–1347 surgery 2007; 60:60–66
[29] Abla AA, Rekate HL, Wilson DA, et al. Orbitozygomatic resection for hypo- [44] Kerrigan JF, Parsons A, Rice SG, et al. Hypothalamic hamartomas: neuropa-
thalamic hamartoma and epilepsy: patient selection and outcome. Childs thological features with and without prior gamma knife radiosurgery. Stereo-
Nerv Syst 2011; 27: 265–277 tact Funct Neurosurg 2013; 91: 45–55

192
Stereotactic Radiosurgery for Epilepsy

24 Stereotactic Radiosurgery for Epilepsy


Mark Quigg and Nicholas M. Barbaro

anticonvulsant effect may arise from a nondestructive, neuro-


Key Points modulatory effect. For example, a dose-dependent reduction in
seizures was shown in kainic-acid-treated epileptic rats4,5,6 and
● The anticonvulsant effect of stereotactic radiation has both in electrical-stimulation epileptic rats7 despite the lack of gross
modulatory, nondestructive components and destructive, le- neuronal injury. Cognitive functions were spared; for example,
sional components; in successful radiosurgery, focal ischemia water maze performance was unimpaired after treatment.5,6
and neuronal destruction of the epileptic target Experiments with kindled animals demonstrate that SRS may
predominate. alter seizure threshold through mechanisms of plasticity and
● Treatment of space-occupying lesions that have epilepsy as a repair.8 For example, if brain irradiation is introduced before,
symptom—tumors or arteriovenous malformations—with during, and after a kindling protocol, seizure thresholds are
Gamma Knife radiosurgery results in high rates of seizure worsened before kindling, unaffected at midkindling, and
remission along with treatment of the lesion. reduced at the final stage of kindling. Thus, kindled epileptic
● Gamma Knife radiosurgery appears to be a reasonable option tissue may lack plasticity and the ability to recover from irradi-
for minimally invasive surgery for the gelastic seizures associ- ation, whereas the recovery process in normal tissue may ren-
ated with hypothalamic hamartoma. der circuits hyperexcitable.
● Gamma Knife radiosurgery for the syndrome of mesial tem- Neuromodulatory effects, however, may not be entirely bene-
poral lobe epilepsy is safe, and in some studies has been ficial. Sometimes proconvulsant effects are observed, as out-
shown to have efficacy similar to that of open surgery, lined in the above animal experiments in which kindled
although worse outcomes have been associated with differ- seizures worsened in animals treated before kindling compared
ences in dose or patient selection. with those treated after kindling.8 This point is emphasized by
results of low-dose human protocols—designed to emulate non-
destructive doses in animal models—that demonstrated transi-
ent exacerbations in auras or sometimes complex partial
24.1 Introduction seizures.8,9 Other studies have explored, in a preliminary fash-
ion, if differential susceptibility to irradiation or ischemia
In association with epilepsy, stereotactic radiosurgery (SRS) is
among different neuronal populations—excitatory versus inhib-
most often used in the treatment of epileptic lesions such as
itory populations—within epileptogenic circuits occurs.9
tumors or vascular malformations; epilepsy in these cases is
Although neurons are resistant to radionecrosis, mitotically
often one of several presenting symptoms and frequently
active structures—vasculature and glia—are not. The other main
improves as a benefit of the treatment of the lesion. Recently,
hypothesis, therefore, is that neuronal destruction results from
the use of SRS has expanded into the treatment of physiological
ischemia caused by radiation vasculitis. Accordingly, the major
lesions such as seen in mesial temporal lobe epilepsy or pre-
pathological findings following focal radiation consist of endo-
viously inoperable epileptic lesions such as hypothalamic
thelial damage.10 Rat brains treated with SRS at 75 Gy and
hamartomas. In this chapter, we outline the radiosurgical
examined 3 months later have, through the method of vascular
management of epilepsy.
casting, a markedly decreased vascular density.11 Electron
microscopy demonstrates thickening of the vascular basement
24.2 Experimental Models and membrane. These vascular changes precede development of
necrosis within the radiosurgical target.
Mechanisms of Radiation Treatment of human mesial temporal lobe epilepsy demon-
A coherent mechanism has not been demonstrated that strates that radionecrosis in the surgical target corresponds
accounts for the anticonvulsant—and occasionally, proconvul- with successful treatment of seizures. Magnetic resonance
sant—effects of SRS. Fundamental to SRS is the stripping of elec- imaging (MRI) and mass spectroscopy12 data from the U.S. Mul-
trons called ionization, resulting in the alteration of chemical ticenter Pilot Study show that the volume of contrast enhance-
bonds or the production of free radicals.1,2 Susceptibility to ion- ment and T2 hyperintensity seen on MRI obtained 12 months
ization is proportional to DNA synthesis. Neurons, therefore, after SRS correlate strongly with outcome. No patients with T2-
are relatively radioresistant; actively proliferating cells, such as weighted volumes of edema less than 200 mL at 12 months
capillary cells, are radiosensitive. Ionizing radiation also dam- went on to experience seizure remission between 24 and 36
ages glial cells such as oligodendroglia that produce myelin and months.12 Furthermore, magnetic resonance spectroscopy
astrocytes.3 Furthermore, although animal models of epilepsy, (MRS) within the target showed evidence of frank ischemia;
usually rats, are helpful in evaluating mechanisms, rat brains one year after SRS, lactate (evidence of anaerobic metabolism)
are remarkably radioresistant, and “scaling up” directly to appeared; choline, creatine, and N-acetylaspartate (NAA) levels
humans to predict dose is not straightforward. (evidence of normal neuronal activity) were largely absent. 12
Hypotheses of the pro- and anticonvulsant effects of SRS fall Similar findings of decreased markers of neuronal activity were
into two basic camps: neuromodulation and tissue necrosis. observed with MRS greater than 1 year after SRS in a trial of six
Studies with rat models of limbic epilepsy emphasize that an patients (without seizure responses results reported).13

193
Stereotactic Radiosurgery for Epilepsy

Histopathology of epileptic targets in humans is difficult to epilepsy.19 A later study22 described dose-volume effects on
interpret because tissue is obtained from failed rather than suc- epileptogenic AVMs opposite to that reported for epileptogenic
cessful SRS in the course of a “rescue” open surgery. For exam- tumors in that seizure remission is better with smaller AVMs 22;
ple, patients who underwent open surgery after failed SRS the amount of radiation to the margin had no clear effect. Nei-
demonstrated hippocampal sclerosis and “radiational changes” ther study found a relationship between obliteration of the
in operative samples14 or changes thought to be confined to AVM and seizure remission.19,22
“normal” hippocampal sclerosis.15
In the U.S. Multicenter Pilot Study, one patient who under-
went temporal lobectomy for steroid-dependent symptoms
24.3.3 Cavernous Malformations
(and who was seizure-free for ~ 3 months before open surgery) The enthusiasm for the use of SRS for cavernous malforma-
was found to have hippocampal sclerosis as well as evidence of tions (CMs) remains mixed. The across-study proportion of
chronic infarcts with prominent hyalinization, thickening, and seizure remission reported in retrospective case series is
closure of small vessels. Similar findings—necrosis with evi- 50%.23 Remission is associated with higher doses. Represent-
dence of vascular damage—were seen in four patients who had ing the extremes in efficacy, Shih et al 24 reported mean mar-
anterior temporal lobectomy (ATL) between 18 and 22 months ginal doses of 13 Gy and central doses of 21 Gy for a
after SRS.16 Therefore, limited data from human histopathology remission rate of 25%, whereas Kim et al 25 used a mean mar-
show a range of findings from no changes to ischemic changes ginal dose of 15 Gy and central dose of 26 Gy for a remission
after failed SRS. rate of 70%. The volume or marginal dose applied to the brain
In summary, the two mechanisms discussed above, neuro- surrounding the CMs, thought important in epileptogenic
modulation and neuronal destruction, may not be mutually tumors17 and AVMs,19 has not been systematically studied
exclusive. We will see below that SRS of mesial temporal lobe with respect to seizure control. Retrospective studies of open
epilepsy helps to define a therapeutic window of treatment resection suggest that removal of the hemosiderin-stained
dose bounded by ineffectiveness, paradoxical exacerbation, or tissue surrounding CM is associated with better outcome 26;
delayed remission at the lower doses and by effective tissue therefore, variable outcomes following SRS may also stem
destruction at higher doses. from inconsistent inclusion of this potentially epileptogenic
region in the treatment volume.
Unfortunately, excess morbidity in terms of radiation toxicity
24.3 Treatment of Lesional with higher doses and posttreatment hemorrhage with lower
Epilepsy doses remains a concern. For example, the early Swedish expe-
rience determined that SRS did not appreciably alter the natural
In other chapters of this book, the details of SRS for mass lesions course of CMs while exposing patients to radiation-induced
are reviewed; in this chapter, we briefly discuss the anticonvul- complications that exceeded by 7 times those expected for the
sant effects of SRS for these lesions. same dose for an AVM.27 A case of a patient treated with SRS for
bilateral CMs and intractable epilepsy reported delayed (> 2-
year postoperative) radiation necrosis and intractable edema
24.3.1 Epileptogenic Tumors requiring craniotomy and hyperbaric oxygen treatments.28 A
Given the variety of types, pathology, and locations of central recent retrospective comparison concluded that open resection
nervous system (CNS) tumors, the studies on effects of SRS on resulted in better seizure control and hemorrhage avoidance
tumor-associated epilepsy are few. However, Schröttner et al than SRS.24 This study, as well as comparisons with the approx-
concentrated on the dose absorbed by tissue in tumor penumb- imately 70 to 80% seizure remission rate seen after open sur-
ra, dividing patients into two groups according to the volume of gery for a CM,29,30 suggests that any benefits of noninvasive SRS
tissue outside the tumor that had received 10 Gy or more.17 over open surgery for a CM must be weighed against risks of
Outcome was retrospectively ranked at mean duration of less efficacy and possibly increased toxicities.
approximately 2 years into excellent (Engel class I–II) or not.
High-volume patients achieved a 66% improvement rate com-
pared with 42% for the low-volume group. Because all patients
24.3.4 Hypothalamic Hamartomas
achieved tumor control with SRS (thus removing tumor res- Hypothalamic hamartomas (HHs) are an important cause of an
ponse as a confounder), the differing rates of seizure improve- epileptic encephalopathy marked by medically intractable
ment suggest that higher SRS volumes delivered to tumor gelastic and other seizures usually accompanied by behavioral
penumbra are important in modifying tumor-associated and cognitive decline. Although HHs are difficult to excise with
epilepsy. the use of standard surgical techniques, SRS has an advantage
of noninvasive access. Series of SRS treatment of HHs33–38 dem-
onstrate a seizure remission rate of 27% across studies.
24.3.2 Arteriovenous Malformations Although this rate appears low, seizure remission alone under-
The potential efficacy of SRS in the treatment of symptomatic estimates the benefits seen with respect to reduced morbidity
localization-related epilepsies is most evident in the treatment and need for less custodial care required in these cases of severe
of arteriovenous malformations (AVMs), with an across-study epilepsy. Behavioral outcomes have not been rigorously quanti-
mean seizure remission rate of 70%.18–22 Representative is the fied, but most reports cited above note that the majority of
large series accumulated by Steiner et al who reported that patients undergo anecdotal improvement in behavior, sleep
seizures remit after SRS in 69% of patients with AVM and quality, and learning.

194
Stereotactic Radiosurgery for Epilepsy

A European, multicenter, prospective trial of SRS for HHs has et al37 who treated 44 patients between 1959 and 1973 with the
enrolled 60 patients, 27 of whom exceeded 3-year follow-up.31 use of yttrium 90, yielding a 75% rate of seizure remission over a
This study emphasizes the evolution of seizure changes during mean follow-up duration of 5.7 years.37 Unfortunately, initial
the postoperative period that track the development of the attempts in Sweden with Gamma Knife38 and elsewhere with the
radiosurgical lesion. Within the first 2 months, a slight improve- use of fractionated radiotherapy and other radiosurgical techni-
ment in seizure rate typically occurs. Seizures transiently wor- ques39 were not encouraging. Many of these patients, however,
sen in frequency before reduction and remission occur. were treated before uniform definitions of MTLE existed.
Behavioral improvements, along with electroencephalographic
(EEG) normalization, tend to occur in a more linear fashion.
Morbidity is low, with no ill effects except for one instance of
24.4.2 Outcomes: Seizure Remission
poikilothermia noted among the above reports. Some reports Régis et al resurrected the Gamma Knife technique for treat-
emphasize the importance of the marginal dose of radiation31 as ment of MTLE in 1995,40 and then conducted a subsequent trial
noted in tumors and AVMs. Patients treated with doses exceed- of seven MTLE patients.41 In this series, a target comprising the
ing 17 Gy to the margin of HH had greater rates of seizure parahippocampal gyrus, head and anterior body of the hippo-
remission than those receiving less than 13 Gy.32 campus, and amygdala, comprising approximately 6.5 mL 50%
More recent studies compared, in nonrandomized and retro- isodose volume, was treated with 25 Gy. Since this publication,
spective fashions, open surgical treatment for HHs. For exam- nine studies of 83 patients report a wide range of efficacies of
ple, Shim et al treated 14 patients: 1 with standard surgery, 4 seizure remission ranging from 0 to 86%14,15,16,47–52 with an
with SRS, and endoscopic disconnection in 11. 33 Of the 11 across-study average of 51%. In comparison, the rate of seizure
patients who underwent endoscopic disconnection, 6 were remission in the prospective, randomized trial of open surgery
seizure-free immediately after surgery. None of the four for MTLE was 58%.35
patients who underwent SRS experienced remission. Outcome Stereotactic radiosurgery studies fall into two groups. The first
may have been limited by the morphology of the lesions; the comprise a variety of single-center case reports and case series.14,
authors note that they were unable to target a sessile base for 15,16,42,43 With one exception,43 these series used doses smaller

“disconnection.” than 25 Gy, and rates of seizure remission were low or nil.
Stereotactic radiosurgery may be one of several nontradition- The second group consists of two larger, prospective, multi-
al surgical approaches that can improve what is otherwise a center trials, one European44 and one American.45 The Euro-
devastating epileptic encephalopathy. Consideration of higher pean trial demonstrated a 2-year postoperative seizure
marginal doses and the morphology of the HH lesion may pro- remission rate of 62% with the use of a treatment protocol iden-
vide guidance in further trials. tical to the group’s earlier studies.
Barbaro et al in the U.S. Multicenter Pilot Study45 randomized
30 patients to a high (24 Gy, n = 13) or low (20 Gy, n = 17) dose
24.4 Treatment of Physiological delivered to the target as specified by Régis et al44 with the
added specification that 50% isodose volumes were restricted
Epilepsies to 5.5 to 7.0 mL attained with two to six isodoses. Ten patients
in each group were seizure-free at 36-month follow-up, result-
24.4.1 Mesial Temporal Lobe Epilepsy ing in a remission rate of 77% in the high-dose and 59% in
Mesial temporal lobe epilepsy (MTLE) consists of atrophy, glio- the low-dose group, for an overall remission rate of 67%
sis, and selective neuronal loss within the hippocampus and (▶ Fig. 24.1).
associated limbic system. Mesial temporal lobe epilepsy is the
most frequent cause of medically intractable epilepsy in adults
(although more recent data suggest that the prevalence may be
dropping, at least among those available to the larger, estab-
lished epilepsy surgery centers34). Stereotactic radiosurgery
may have several potential benefits that drive its consideration
despite the well-established efficacy of open surgery for
MTLE.35 First, open surgery has a small, but real risk of surgical
complications such as bleeding, infection, stroke, and postope-
rative pain. Second, open surgery is expensive. The average cost
per patient for open microsurgery is nearly 50% more than that
for SRS for treatment of similar disorders36; admittedly, relative
costs of open versus SRS have not been studied. Third, some of
the adverse events of open surgery involve changes in neuro-
psychological testing. Fourth, some patients as well as physi- Fig. 24.1 Distribution of seizure outcome as defined by Engel’s criteria
cians hold persistent biases against open surgery despite between months 24 and 36 of the National Institutes of Health-
proven efficacy; noninvasive techniques may broaden the sponsored, multicenter pilot study of stereotactic radiosurgery for
acceptance of epilepsy surgery. unilateral mesial temporal lobe epilepsy divided by dose.45 The overall
The first surgical series employing SRS in a systematic fashion rate of seizure remission (Engel class IB or better) at 36 months was
in the treatment of physiological focal epilepsy (as opposed to 76.9% for the high-dose and 58.8% for the low-dose group (p = 0.12 by
Fisher’s exact test).
epilepsy resulting from mass lesions) was reported by Talairach

195
Stereotactic Radiosurgery for Epilepsy

Vojtěch et al published a retrospective analysis of 14 patients and anatomy important in the pathophysiology of MTLE.
with markedly different results.46 Initial doses were identical to Patients with epilepsies that may differ from the stereotypic
that reported by Régis et al; in fact, six of the patients overlap unilateral MTLE lesion may not be suitable for SRS. For example,
with the European multicenter trial.44 Doses decreased with most patients in the long-term French cohort who failed SRS
accumulated experience; of the 14 patients in the report, the also had evidence for extension of the “epileptic zone” beyond
last 6 were treated with 18 Gy because of clinically significant mesial structures.47 One reason that was offered to explain poor
“prominent radiosurgical responses” associated with 25 Gy. outcomes of the experience of Vojtěch et al was that the pri-
None of the 14 patients were seizure-free after 39 months. mary epileptic injury in some patients could be considered
A recent report on longer term follow-up (≥ 5 years, mean fol- symptomatic “MTLE plus.”46 Patients with evidence of involve-
low-up of ~ 8 years) of 15 patients shows that seizure remission ment outside of the SRS target or with symptomatic etiologies
remains durable with 60% of patients experiencing either seiz- may not experience seizure remission with the highly selective
ure remission or nondisabling auras.47 Because long-term fol- technique of SRS.
low-up was not conducted with intent-to-treat criteria, the
results may be susceptible to sampling bias.
24.4.4 Morbidities
Morbidities in the multicenter protocols do not exceed those of
24.4.3 Treatment Protocols open surgery and fall in line with those seen commonly after
The variability in seizure remission rates underscores the diffi- SRS.44 Both trials outlined the typical postoperative course and
culties in the determination of basic parameters of anatomical development of the radiosurgical lesion, and in many respects,
target, dose, and target volume. The anatomical targets in SRS it follows the polyphasic course described after SRS of a HH. 31
of MTLE are the least variable factor, with all studies14,15,47,48,49, Most clinical or neuroimaging changes occur between 9 and 15
50 specifying that the 50% isodose volume contains regions months. Nearly all patients experience exacerbations in auras
thought most important in generation of mesial seizures: the before seizures decrease or remit (▶ Fig. 24.3). The most dra-
amygdala, the head and anterior body of the hippocampus, and matic drop in seizure rate occurs between 12 and 18 months,
the parahippocampal gyrus. coinciding with the development and resolution of maximal
Dose and target volume are more variable. Comparison of the MRI changes.12
above studies suggests that low-dose protocols (≤ 20 Gy)14,15,42 Other morbidities reported by the two multicenter studies
are less successful than higher-dose protocols (> 20 Gy). 47,50,51 include visual field deficits. Superior quadrant field defects
As noted above, in patients randomized by dose (high = 24 Gy, are an expected morbidity after standard temporal lobec-
low = 20 Gy), Barbaro et al noted that the high-dose group expe- tomy, with rates of visual field defects between 52 to 100%. 48
rienced a higher rate of seizure remission, experienced a short- Quantified visual fields were measured in the U.S. Multicen-
er latency to seizure remission, and experienced fewer auras ter Study48 at 24 months after SRS. No patients reported vis-
during development of the radiosurgical lesion (▶ Fig. 24.2).45 ual changes. Fifteen (62.5%) of 24 patients had postoperative
Variability in seizure remission among the above trials also visual field changes, all homonymous superior quadranta-
emphasizes the importance of patient selection. Stereotactic nopsias. No changes were consistent with injury to the optic
radiosurgery is, in effect, a “superselective” amygdalohippo- nerve or chiasm. Consistent with above findings that found
campectomy involving the destruction of the minimum volume seizure remission correlating with dose, patients with seizure
remission had a higher rate of visual field defects. In the
European study, one patient had a hemianopia (indicating
direct involvement of the optic tract) and another had mixed
deficits.44
Other morbidities consisted of headache, nausea, vomiting,
and depression. Headache requires special comment, as it coin-
cides in some patients with postoperative edema. Steroids, in
response to headache, visual field changes, or MRI changes,
were used in 62% of patients.45
One potential problem of SRS compared with open surgery is
that the latency between SRS and remission may expose
patients to continued seizures. Two deaths occurred during the
latency period in a case series of five patients treated with low-
er-dose radiation.14 Given the known risk of death from uncon-
trolled seizures (SUDEP), and the possible proconvulsive effects
of lower radiation doses, it is possible that radiosurgery con-
Fig. 24.2 Distribution of continuous duration of seizure remission
tributed to the death in these cases. Pathological evaluation of
(defined as Engel class IB or better) from the final outcome point of 36 tissue obtained in these cases only showed evidence of MTS.
months postoperatively in the National Institutes of Health–sponsored Allowing enough time for maturation of the radiosurgical lesion
multicenter pilot study.45 Patients who dropped out early because of may have allowed some patients treated with lower doses to
“escape” temporal lobectomy or because of loss of follow-up are demonstrate remission, given that animal models treated with
counted as a duration of zero months. p value = 0.05 from two-sample lower doses demonstrated improvements at a slower rate than
t test. Bar, average.
those treated with higher doses.6,49 Future trials of SRS may

196
Stereotactic Radiosurgery for Epilepsy

Fig. 24.3 Mean ± standard error of the number of


auras per seizure diary interval in patients of the
National Institutes of Health–sponsored multi-
center pilot study.45 As seen in earlier studies, the
onset of auras frequently preceded frank changes
on magnetic resonance imaging and coincided
with the diminishment of complex partial seiz-
ures.

need to consider anticonvulsant prophylaxis during the latency open surgery suggest that the highly selective radiosurgical le-
period. sion may provide some benefit. The ROSE Trial (Radiosurgery or
Open Surgery for Epilepsy), a prospective randomized trial of
standard open surgery versus SRS, is currently underway. The
24.4.5 Secondary Outcomes/ ROSE Trial may provide further evidence regarding the effects
Neurocognition of SRS on neuropsychological functioning and whether cogni-
tive abilities may be less adversely affected by SRS relative to
Some studies reported secondary outcome measures such as
traditional open surgery.
cognition. In fact, SRS of MTLE may serve as an interesting mod-
Stereotactic radiosurgery for MTLE appears to have promise.
el of the effects of focal radiation on cognition, especially
Higher-dose protocols appear to have better results. Patient
because controversies over the cognitive sequelae of focal or
selection and relative benefits compared with open surgery
fractionated radiation therapy are often engendered by the con-
have yet to be clearly demarcated.
founding effects of tumors or other CNS lesions.50 Epileptic rats
demonstrated no cognitive effects attributable to SRS. 6
So far, four studies in the treatment of MTLE have reported
neurocognitive outcomes. Two single-center series reported
24.5 Other Nonlesional Epilepsies
mixed results. Srikijvilaikul et al reported no group mean 24.5.1 Neocortical Foci
changes at 6-month follow-up, although some individuals
showed decline in at least one cognitive domain.14 McDonald et There are no published reports on the use of SRS for nonstruc-
al reported on 27-month follow-up in three patients who tural lesion neocortical foci. Régis has presented a small series
underwent dominant hemisphere low-dose SRS treatment. 51 of cases where radiosurgery was delivered to perisylvian
No long-term consistent changes in neurocognitive parameters regions including the insular cortex following localization of
were found, although each patient showed decline in a measure the seizure focus with stereo-EEG recordings (J. Régis, personal
of verbal memory. They concluded that neurocognitive changes communication). Because localization of seizure foci in nonle-
following SRS appeared similar to those of temporal lobectomy. sional neocortical epilepsies usually requires invasive techni-
The European prospective trial reported no mean neurocog- ques, the noninvasive nature of SRS loses some advantage. One
nitive changes over a 2-year follow-up.44 may speculate that if noninvasive localization and brain map-
Detailed evaluation of neurocognitive outcomes were pro- ping were rigorous enough to supplant invasive methods, then
vided from the U.S. Multicenter Trial.52 Mean scores of tests of SRS may have a future role.
language and verbal memory at 24 and 36 months after SRS did
not differ from preoperative baselines. Specifically, significant
24.5.2 Corpus Callosotomy
verbal memory impairment was seen in 25% of dominant-
surgery patients and in 7% of nondominant patients. In fact, sig- Transection of the corpus callosum may decrease the severity
nificant improvement was seen in 16% of dominant and 7% of and number of primary generalized or rapidly propagating sec-
nondominant patients. In comparison, rates of significant ondarily generalized seizures in patients who are not otherwise
impairment following dominant hemisphere temporal lobec- good surgical candidates. Small case series report that improve-
tomy range from 10 to 60%.53–57 Furthermore, mood remained ment in seizures following SRS resection of the corpus callosum
stable, and quality-of-life scores improved in those patients was comparable to that reported after open callosotomy with
who experienced seizure remission. lack of notable complications.58,59 The future role of SRS in cor-
In summary, seizure remission rates following SRS vary, but pus callosotomy may be more difficult to determine because
the larger, multicenter trials with higher-dose protocols more epileptologists are currently opting for vagal nerve stimu-
showed remission rates similar to the gold standard of open lation in patients who might otherwise qualify for corpus
surgery. Favorable neurocognitive outcomes compared with callosotomy.

197
Stereotactic Radiosurgery for Epilepsy

report of five cases. Neurosurgery 2004; 54: 1395–1402, discussion 1402–


24.6 Summary 1404
[15] Cmelak AJ, Abou-Khalil B, Konrad PE, Duggan D, Maciunas RJ. Low-dose ster-
Stereotactic radiosurgery offers an alternative to open surgery eotactic radiosurgery is inadequate for medically intractable mesial temporal
in selected patients with epilepsy caused by mass lesions such lobe epilepsy: a case report. Seizure 2001; 10: 442–446
as tumors or AVMs with the choice of technique guided by the [16] Prayson RA, Yoder BJ. Clinicopathologic findings in mesial temporal sclerosis
location and type of the lesion rather than any epilepsy-specific treated with gamma knife radiotherapy. Ann Diagn Pathol 2007; 11: 22–26
[17] Schröttner O, Eder HG, Unger F, Feichtinger K, Pendl G. Radiosurgery in le-
properties of the lesion. Stereotactic radiosurgery for the seiz-
sional epilepsy: brain tumors. Stereotact Funct Neurosurg 1998; 70 (Suppl
ures arising from CMs appears to have a less than enthusiastic 1): 50–56
endorsement given some studies’ findings of excessive rebleed- [18] Heikkinen ER, Konnov B, Melnikov L, et al. Relief of epilepsy by radiosurgery
ing and lower seizure-free rates than with open surgery. Stereo- of cerebral arteriovenous malformations. Stereotact Funct Neurosurg 1989;
53: 157–166
tactic radiosurgery for HHs, in contrast, appears promising
[19] Steiner L, Lindquist C, Adler JR, Torner JC, Alves W, Steiner M. Clinical out-
given the combination of inaccessibility to traditional surgery, come of radiosurgery for cerebral arteriovenous malformations. J Neurosurg
severe morbidity of gelastic epilepsy, and good safety profile. 1992; 77: 1–8
Finally, recent trials suggest that certain protocols of SRS for [20] Gerszten PC, Adelson PD, Kondziolka D, Flickinger JC, Lunsford LD. Seizure
MTLE may offer a noninvasive alternative to open surgery while outcome in children treated for arteriovenous malformations using gamma
knife radiosurgery. Pediatr Neurosurg 1996; 24: 139–144
yielding similar rates of seizure remission. Other possible situa-
[21] Kurita H, Kawamoto S, Suzuki I, et al. Control of epilepsy associated with cer-
tions merit consideration of SRS; one authority proposes that ebral arteriovenous malformations after radiosurgery. J Neurol Neurosurg
SRS may be useful in cases of (1) epilepsy of temporal lobe ori- Psychiatry 1998; 65: 648–655
gin in those who have medical contraindications to open sur- [22] Schäuble B, Cascino GD, Pollock BE, et al. Seizure outcomes after stereotactic
radiosurgery for cerebral arteriovenous malformations. Neurology 2004; 63:
gery, (2) temporal lobe epilepsy after failed open resection, and
683–687
(3) extratemporal epilepsy that can be noninvasively localized, [23] Bartolomei F, Régis J, Kida Y, et al. Gamma Knife radiosurgery for epilepsy as-
especially following a failed open surgery.60 Further work is sociated with cavernous hemangiomas: a retrospective study of 49 cases.
needed to clarify risks and benefits of SRS for these less com- Stereotact Funct Neurosurg 1999; 72 (Suppl 1): 22–28
mon but important clinical situations. [24] Shih YH, Pan DH. Management of supratentorial cavernous malformations:
craniotomy versus gammaknife radiosurgery. Clin Neurol Neurosurg 2005;
107: 108–112
[25] Kim MS, Pyo SY, Jeong YG, Lee SI, Jung YT, Sim JH. Gamma knife surgery for
References intracranial cavernous hemangioma. J Neurosurg 2005; 102 (Suppl): 102–
106
[1] Khan FM. The Physics of Radiation Therapy. 4th ed. Philadelphia, PA: Lippin- [26] Baumann CR, Schuknecht B, Lo Russo G, et al. Seizure outcome after resection
cott Williams & Wilkins; 2010 of cavernous malformations is better when surrounding hemosiderin-stained
[2] Haffty BG, Wilson LD. Handbook of Radiation Oncology: Basic Principles and brain also is removed. Epilepsia 2006; 47: 563–566
Clinical Protocols. Sudbury, MA: Jones and Bartlett; 2009 [27] Karlsson B, Kihlström L, Lindquist C, Ericson K, Steiner L. Radiosurgery for
[3] Fike JR. Physiopathology of radiation-induced neurotoxicity. Rev Neurol cavernous malformations. J Neurosurg 1998; 88: 293–297
(Paris) 2011; 167: 746–750 [28] Takenaka N, Imanishi T, Sasaki H, et al. Delayed radiation necrosis with exten-
[4] Sun B, DeSalles AA, Medin PM, et al. Reduction of hippocampal-kindled seiz- sive brain edema after gamma knife radiosurgery for multiple cerebral caver-
ure activity in rats by stereotactic radiosurgery. Exp Neurol 1998; 154: 691– nous malformations—case report. Neurol Med Chir (Tokyo) 2003; 43: 391–
695 395
[5] Maesawa S, Kondziolka D, Balzer J, Fellows W, Dixon E, Lunsford LD. The be- [29] Cappabianca P, Alfieri A, Maiuri F, Mariniello G, Cirillo S, de Divitiis E. Supra-
havioral and electroencephalographic effects of stereotactic radiosurgery for tentorial cavernous malformations and epilepsy: seizure outcome after lesio-
the treatment of epilepsy evaluated in the rat kainic acid model. Stereotact nectomy on a series of 35 patients. Clin Neurol Neurosurg 1997; 99: 179–183
Funct Neurosurg 1999; 73: 115 [30] Cohen DS, Zubay GP, Goodman RR. Seizure outcome after lesionectomy for
[6] Maesawa S, Kondziolka D, Dixon CE, Balzer J, Fellows W, Lunsford LD. Subne- cavernous malformations. J Neurosurg 1995; 83: 237–242
crotic stereotactic radiosurgery controlling epilepsy produced by kainic acid [31] Régis J, Scavarda D, Tamura M, et al. Epilepsy related to hypothalamic hamar-
injection in rats. J Neurosurg 2000; 93: 1033–1040 tomas: surgical management with special reference to gamma knife surgery.
[7] Chen ZF, Kamiryo T, Henson SL, et al. Anticonvulsant effects of gamma sur- Childs Nerv Syst 2006; 22: 881–895
gery in a model of chronic spontaneous limbic epilepsy in rats. J Neurosurg [32] Régis J, Bartolomei F, de Toffol B, et al. Gamma knife surgery for epilepsy re-
2001; 94: 270–280 lated to hypothalamic hamartomas. Neurosurgery 2000; 47: 1343–1351, dis-
[8] Jenrow KA, Ratkewicz AE, Zalinski DN, Roszka KM, Lemke NW, Elisevich KV. cussion 1351–1352
Influence of ionizing radiation on the course of kindled epileptogenesis. Brain [33] Shim KW, Chang JH, Park YG, Kim HD, Choi JU, Kim DS. Treatment modality
Res 2006; 1094: 207–216 for intractable epilepsy in hypothalamic hamartomatous lesions. Neurosur-
[9] Tsuchitani S, Drummond J, Kamiryo T, et al. Selective vulnerability of inter- gery 2008; 62: 847–856, discussion 856
neurons to low dosage radiosurgery. Abstract presented at: Society for Neu- [34] Englot DJ, Ouyang D, Garcia PA, Barbaro NM, Chang EF. Epilepsy surgery
roscience Annual Meeting; October 8–12, 2003; New Orleans, LA trends in the United States, 1990–2008. Neurology 2012; 78: 1200–1206
[10] Kondziolka D, Lunsford LD, Claassen D, Maitz AH, Flickinger JC. Radiobiology [35] Wiebe S, Blume WT, Girvin JP, Eliasziw M, Effectiveness and Efficiency of Sur-
of radiosurgery: Part I. The normal rat brain model. Neurosurgery 1992; 31: gery for Temporal Lobe Epilepsy Study Group. A randomized, controlled trial
271–279 of surgery for temporal-lobe epilepsy. N Engl J Med 2001; 345: 311–318
[11] Kamiryo T, Lopes MB, Kassell NF, Steiner L, Lee KS. Radiosurgery-induced mi- [36] Wellis G, Nagel R, Vollmar C, Steiger HJ. Direct costs of microsurgical manage-
crovascular alterations precede necrosis of the brain neuropil. Neurosurgery ment of radiosurgically amenable intracranial pathology in Germany: an
2001; 49: 409–414, discussion 414–415 analysis of meningiomas, acoustic neuromas, metastases and arteriovenous
[12] Chang EF, Quigg M, Oh MC, et al. Epilepsy Radiosurgery Study Group. Predic- malformations of less than 3 cm in diameter. Acta Neurochir (Wien) 2003;
tors of efficacy after stereotactic radiosurgery for medial temporal lobe epi- 145: 249–255
lepsy. Neurology 2010; 74: 165–172 [37] Talairach J, Bancaud J, Szikla G. Approche nouvelle de la neurochirurgie de
[13] Hájek M, Dezortová M, Liscák R, Vymazal J, Vladyka V. 1H MR spectroscopy of l’epilepsie. Méthodologie stéréotaxique et résultats thérapeutiques. In: Neu-
mesial temporal lobe epilepsies treated with Gamma knife. Eur Radiol 2003; rochirurgie: Congrés Annuel de la Société de Langue Française. Marseille,
13: 994–1000 France: Masson; 1974:205–213
[14] Srikijvilaikul T, Najm I, Foldvary-Schaefer N, Lineweaver T, Suh JH, Bingaman [38] Lindquist C, Kihlström L, Hellstrand E. Functional neurosurgery—a future for
WE. Failure of gamma knife radiosurgery for mesial temporal lobe epilepsy: the gamma knife? Stereotact Funct Neurosurg 1991; 57: 72–81

198
Stereotactic Radiosurgery for Epilepsy

[39] Barcia JA, Barcia-Salorio JL, López-Gómez L, Hernández G. Stereotactic radio- [50] Armstrong CL, Gyato K, Awadalla AW, Lustig R, Tochner ZA. A critical review
surgery may be effective in the treatment of idiopathic epilepsy: report on of the clinical effects of therapeutic irradiation damage to the brain: the roots
the methods and results in a series of eleven cases. Stereotact Funct Neuro- of controversy. Neuropsychol Rev 2004; 14: 65–86
surg 1994; 63: 271–279 [51] McDonald CR, Norman MA, Tecoma E, Alksne J, Iragui V. Neuropsychological
[40] Régis J, Peragui JC, Rey M, et al. First selective amygdalohippocampal radio- change following gamma knife surgery in patients with left temporal lobe ep-
surgery for ‘mesial temporal lobe epilepsy’. Stereotact Funct Neurosurg 1995; ilepsy: a review of three cases. Epilepsy Behav 2004; 5: 949–957
64 (Suppl 1): 193–201 [52] Quigg M, Broshek DK, Barbaro NM, et al. Radiosurgery Epilepsy Study Group.
[41] Régis J, Bartolomei F, Rey M, et al. Gamma knife surgery for mesial temporal Neuropsychological outcomes after Gamma Knife radiosurgery for mesial tempo-
lobe epilepsy. Epilepsia 1999; 40: 1551–1556 ral lobe epilepsy: a prospective multicenter study. Epilepsia 2011; 52: 909–916
[42] Kawai K, Suzuki I, Kurita H, Shin M, Arai N, Kirino T. Failure of low-dose ra- [53] Stroup E, Langfitt J, Berg M, McDermott M, Pilcher W, Como P. Predicting ver-
diosurgery to control temporal lobe epilepsy. J Neurosurg 2001; 95: 883–887 bal memory decline following anterior temporal lobectomy (ATL). Neurology
[43] Hoggard N, Wilkinson ID, Griffiths PD, Vaughan P, Kemeny AA, Rowe JG. The 2003; 60: 1266–1273
clinical course after stereotactic radiosurgical amygdalohippocampectomy [54] Hermann BP, Wyler AR, Bush AJ, Tabatabai FR. Differential effects of left and
with neuroradiological correlates. Neurosurgery 2008; 62: 336–344, discus- right anterior temporal lobectomy on verbal learning and memory perform-
sion 344–346 ance. Epilepsia 1992; 33: 289–297
[44] Régis J, Rey M, Bartolomei F, et al. Gamma knife surgery in mesial temporal [55] Hermann BP, Seidenberg M, Dohan FC Jr, et al. Reports by patients and their
lobe epilepsy: a prospective multicenter study. Epilepsia 2004; 45: 504–515 families of memory change after left anterior temporal lobectomy: relation-
[45] Barbaro NM, Quigg M, Broshek DK, et al. A multicenter, prospective pilot ship to degree of hippocampal sclerosis. Neurosurgery 1995; 36: 39–44, dis-
study of gamma knife radiosurgery for mesial temporal lobe epilepsy: cussion 44–45
seizure response, adverse events, and verbal memory. Ann Neurol 2009; [56] Seidenberg M, Hermann B, Wyler AR, Davies K, Dohan FC Jr, Leveroni C. Neu-
65: 167–175 ropsychological outcome following anterior temporal lobectomy in patients
[46] Vojtěch Z, Vladyka V, Kalina M, et al. The use of radiosurgery for the treat- with and without the syndrome of mesial temporal lobe epilepsy. Neuropsy-
ment of mesial temporal lobe epilepsy and long-term results. Epilepsia 2009; chology 1998; 12: 303–316
50: 2061–2071 [57] Chelune GJ, Najm IM. Risk factors associated with postsurgical decrements in
[47] Bartolomei F, Hayashi M, Tamura M, et al. Long-term efficacy of gamma memory decline after anterior temporal lobectomy. In: Luders HO, Comair Y,
knife radiosurgery in mesial temporal lobe epilepsy. Neurology 2008; 70: eds. Epilepsy Surgery. 2nd ed. Philadelphia, PA: Raven; 2000:497–504
1658–1663 [58] Eder HG, Feichtinger M, Pieper T, Kurschel S, Schroettner O. Gamma knife ra-
[48] Hensley-Judge H, Quigg M, Barbaro NM, et al. Visual field defects after radio- diosurgery for callosotomy in children with drug-resistant epilepsy. Childs
surgery for mesial temporal lobe epilepsy. Epilepsia 2013; 54: 1376–1380 Nerv Syst 2006; 22: 1012–1017
[49] Mori Y, Kondziolka D, Balzer J, et al. Effects of stereotactic radiosurgery on an [59] Pendl G, Eder HG, Schroettner O, Leber KA. Corpus callosotomy with radio-
animal model of hippocampal epilepsy. Neurosurgery 2000; 46: 157–165, surgery. Neurosurgery 1999; 45: 303–307, discussion 307–308
discussion 165–168 [60] Anschel DJ, Romanelli P. Letter in response to Quigg and Barbaro. Arch Neurol
2008

199
25 Stereotactic Radiosurgery for Ocular
Part VI Disorders 202

Ocular Disorder and Pediatric 26 Stereotactic Radiosurgery for Pediatric


Brain Tumors 210
Tumor Indications

VI
Stereotactic Radiosurgery for Ocular Disorders

25 Stereotactic Radiosurgery for Ocular Disorders


Roman Liscak and Gabriela Simonova

targets, and stereotactic coordinates are derived from the posi-


Key Points tion of the coordinate head frame, which is rigidly fixed to the
skull. Proper eye fixation or an eye motion-monitoring system
● The eye bulb has to be reliably immobilized before the is required because the eye can move during the treatment pro-
radiosurgery. cedure. Very eccentric target volume location can also cause
● Build-up over the eye bulb is necessary for precise dose some inaccuracies in the treatment-planning calculations,
calculations. including relative dose distribution and absolute dose calcula-
● Uveal melanoma is the most frequent indication for ophthal- tions. Finally, there may be some technical inconveniences in
mic radiosurgery, which has a high rate of preserving of the treating patients due to limitations in the range of the coordi-
eye bulb. nate system in this eccentric target location. Despite these diffi-
culties, radiosurgery is an attractive alternative treatment for
ophthalmic lesions mainly because of its accurate and precise
25.1 Introduction delivery of high conformal doses to the treatment target.

Due to its underlying radiobiological principles, radiosurgery is


generally used to treat small-volume lesions, and volumes of 25.3 Eye Fixation
ocular disorders usually fall within the volume constraints of Several different eye immobilization systems and techniques
radiosurgery. The first radiosurgical experience with ophthal- have been described in the literature.1,9–16 Tokuuye et al
mologic indications involved the treatment of patients suffering described a mask fixation technique with a plastic mold gently
from uveal melanomas.1–8 However, it appears that the poten- pressed down over the orbit to restrict ocular movements.15
tial of radiosurgery to provide effective treatment for ophthal- Zehetmayer et al reported on a suction immobilization techni-
mologic indications is broader. que for radiosurgery of intracranial malignancies using the
Radiosurgery of ocular disorders necessitates a close coopera- GK.16 Langmann et al used a fixation method of the globe by
tion with ophthalmologists. This multidisciplinary approach is means of a retrobulbar anesthetic block for eye treatments
critical for patient selection, follow-up, and treatment. using the GK.1 In the Prague (Czech Republic) center, the
treated eye was fixed under a retrobulbar local anesthesia by
25.2 Technical Issues Specific to suturing two rectus eye muscles to the stereotactic frame; this
procedure was done by an ophthalmologist. To ensure adequate
Stereotactic Radiosurgery for dose build-up and homogeneous propagation of radiation
Ocular Disorders between the tissues and the air, a partial tarsorrhaphy was per-
formed, and a plastic cover filled with tissue-equivalent gel was
The Leksell Gamma Knife (GK; Elekta Instruments AB, Stock- used (▶ Fig. 25.1). This simultaneously acted as protection for
holm, Sweden) was originally designed to treat deep brain the cornea. This eye fixation technique showed very good

Fig. 25.1 Fixation of the eye. (a) Retrobulbar local


anesthesia; (b) Suturing two rectus eye muscles;
(c) Plastic cover filled with tissue-equivalent gel;
(d) Build-up with fixation of rectus eye muscles to
the stereotactic frame.

202
Stereotactic Radiosurgery for Ocular Disorders

stability and rigidity in time. Having the patient in a prone ver- to the treated eye was recommended (▶ Fig. 25.1). Such materi-
sus supine position had no effect on stereotactic target and eye- al should reduce discrepancies between the planned and the
structure localization.12,13 delivered radiation dosimetry. Relative dose distributions
Active eye immobilization techniques, where the patient con- measured by polymer gel dosimetry in the region of the target
trols the eye position, for example, by fixating on a light source, volumes were in good agreement with the Leksell GammaPlan
have been described for proton/helium ion beam therapy and (Elekta AB), and measurements demonstrated that calculation
also for linear accelerator (linac)-based stereotactic radiother- errors should not exceed more than 10% even at surface struc-
apy of malignant tumors.9,11 For proton/helium ion beam ther- tures when built-up material is used.
apy, a surgical intervention is performed to position tantalum Because of the marginal position of the eye, an extreme gam-
clips at the border of the visible tumor under diaphanoscopical ma angle has to be used to avoid collision of the skull with the
control. These clips are used during treatment planning to collimator helmet of the GK model B or C. Patients usually do
define the target volume and to verify the position of the tumor not tolerate excessive backward bending of the head in a supine
during treatment delivery by using X-rays.10 Petersch et al position. Therefore, the prone position was used in most cases.
described a noninvasive eye fixation system for the application The problems of collisions and coordinate limits have been
of linac-based stereotactic radiotherapy in the treatment of overcome by the enlarged space of the collimator helmet in the
uveal melanoma.14 The computer-controlled eye-monitoring latest model of the Leksell Gamma Knife Perfexion (Elekta AB),
system is based on the patient’s fixation on a point of light. and patients are treated now comfortably in the supine position
When the eye position exceeds the preset geometrical limits to without any difficulty.
that point of light, the system automatically initiates a cessation
of the radiation beam until the eye is repositioned to the proper
alignment. 25.5 Specific Disorders
25.5.1 Uveal Melanoma
25.4 Dosimetry and Treatment
Natural History and Clinical Presentation
Planning Uveal melanoma represents the most common primary malig-
Due to very eccentric target volume location, some inaccuracies nant tumor of the eye in adults. There is a peak incidence
in the treatment-planning calculations, including relative dose between 55 and 70 years of age, with a prevalence of six or sev-
distributions and absolute dose calculations (calculation of en cases per million people per year. The incidence is rare in
treatment time or monitor units), can be expected. This issue is patients younger than 20 years of age. Approximately 50% of
even more prominent for surface structures of the eye (e.g., eye patients with diagnosed melanomas of the choroid or ciliary
lens, eyelid, and cornea) than for target volumes itself. body will die from this tumor within 15 years.17,18
Petersch et al reported excellent agreement between actual Uveal melanomas are neoplasms arising from the uveal tract
measurements and the treatment-planning system calculations that includes the iris, ciliary body, and choroid. The uveal tract
for uveal melanomas at a depth of 15 to 20 mm.14 However, at is a vascular structure with no lymphatic drainage. Lymphatic
small depths (< 10 mm), the treatment-planning system cannot node involvement (preauricular, submandibular, and cervical
model the influence of absorption in the eye motion-monitor- nodes) can be diagnosed on rare occasions when subconjuncti-
ing device accurately if the beam passed perpendicularly val extension of the primary tumor has also been observed. The
through it. Consequently, as a means of reducing dosimetric major routes of extension are local growth and hematogenous
errors, avoidance of frontal beams during radiosurgery to ocu- spread. The common sites of hematologic dissemination are the
lar lesions has been suggested. liver, lung, and brain.
Detailed dosimetric measurements were also performed at Wide variations in the grading of malignancy can be
the Prague Na Homolce Hospital. In this case, treatment-plan- observed, from relatively benign types with several years of
ning system calculations of target volume relative dose distri- survival without dissemination, to other tumors with fast mul-
butions and absolute dose calculations in different depths were tiorgan dissemination, which can lead to the patient’s death
compared with experimental measurements. 13 Various treat- within a few months. Tumor growth is also influenced by the
ment plans for different ophthalmic indications (e.g., uveal mel- potential cell-doubling time, which varies widely from 60 to
anoma, glaucoma, and retinoblastoma) calculated for the GK 350 days (median ~ 70 days).19 A longer doubling time as com-
were evaluated. Absolute doses measured by pinpoint ion pared with a significant proportion of other tumors can explain
chamber at depths of 10 mm and deeper showed relatively the relatively long survival of these patients and also the rela-
good agreement with the treatment-planning system calcu- tively slow tumor response after irradiation.
lated data. Absolute doses delivered during head phantom irra- The correct diagnosis for a majority of patients with uveal
diation were systematically lower than calculated ones, usually melanoma can be made by taking a history and performing a
within 5% or even within 3%. Absolute doses measured by pin- complete ocular examination. The ophthalmologic examination
point ion chamber at depths < 10 mm showed a relatively large includes external ocular examination, indirect ophthalmoscopy,
deviation compared with the treatment-planning system’s cal- fluorescein angiography, and ultrasonography, which plays a
culated data. In this situation, absolute doses delivered during basic role at the time of diagnosis. The ophthalmologic exami-
phantom irradiation were systematically lower by 15 to 20%. To nation also plays a role in follow-up and can be used to obtain
improve on inaccuracies in absolute dose calculations in surface precise measurement of the tumor base and height. The height
structures, proper tissue-equivalent built-up material attached of the tumor in particular represents one of the most important

203
Stereotactic Radiosurgery for Ocular Disorders

Table 25.1 Toxicity scoring system (RTOG/EORTC LENT SOMA)


Grade 1 Grade 2 Grade 3 Grade 4

Cornea Increased tearing on Noninfectious keratitis Infectious keratitis; corneal ulcer Panophthalmitis; corneal scar;
examination ulceration leading to perforation
of globe/loss of globe

Iris Rubeosis only Rubeosis; increased intraocular Neovascular glaucoma; ability to Neovascular glaucoma without
pressure count fingers 1 m ability to count fingers 1 m;
complete blindness

Optic nerve Afferent pupillary defect with ≤ ¼ Pallor with asymptomatic > Pallor or central scotoma Profound optic atrophy; com-
normal-appearing nerve visual field defect plete blindness

Lens Asymmetric lenticular opac- Moderate lenticular changes Moderate lenticular changes Severe lenticular changes
ities; no visual loss with mild-moderate visual loss with severe visual loss

Retina Microaneurysms, nonfoveal Cotton wool spots Massive macular Opaque vitreous hemorrhage;
exudates, minor vessel attenu- exudation; focal retinal detach- complete retinal detachment;
ation, and extrafoveal pigment ment blindness
changes

Sclera Loss of episcleral vessels ≤ 50% Scleral thinning > 50% Scleral thinning Scleral or periosteal graft re-
quired due to perforation

Abbreviations: EORTC, European Organization for Research and Treatment of Cancer; LENT SOMA, late effects of normal tissues, subjective, objective,
management, and analytic; RTOG, Radiation Therapy Oncology Group.

factors influencing the treatment decision, and it is used during tumor height > 10 mm or extrascleral growth were indicated for
follow-up to evaluate the tumor’s response. enucleation (5% in our series).
The most extensively published charged particle center is the
Paul Scherrer Institute in Switzerland.28 In their group of 2,258
Results of Radiosurgery, Stereotactic
patients, 5- and 10-year local tumor control rates were 96 and
Radiotherapy, and Proton Beam Radiotherapy 95%, respectively. Five- and 10-year survival rates were 82 and
The first experiments with radiosurgery for the treatment of 62%, respectively. Seven percent of patients underwent enuclea-
uveal melanomas were conducted using the rabbit eye melano- tion after proton therapy. Preservation of the eye 5 and 10 years
ma model. The reported tumor lethal dose ranged from 60 to after the treatment was 89 and 85%, respectively. Dendale et al29
90 Gy applied in a single session.5 Several authors reported the reported a large series of 1,406 patients treated by proton beam
results of Gamma Knife stereotactic radiosurgery (GKSRS) in therapy (four daily fractions); the 5-year local tumor control rate
uveal melanomas, and some observed relatively high radiation- was 96%. The 5-year enucleation for complications rate was
related toxicity.1,2,3,4,6,7,20–25 The minimum effective dose for 7.7%. Caujolle et al30,31 reported a local recurrence rate of 6.1% at
long-lasting local tumor control (10–15 years), as well as the 5 years from series of 1,102 patients after proton beam therapy.
tolerance doses for critical ocular structures, is not yet well Stereotactic irradiation using linac with applied doses of 45
defined. Toktas et al24 reported 35 patients treated using GK to 70 Gy delivered usually in three fractions is also an effective
with standardized dose 30 Gy on 50% isodose curve for all method, with tumor height reduction achievement in 97% of
patients; the 3-year local tumor control rate was 83%. The most cases, secondary enucleation performance in 13%, and the inci-
frequent complication was retinal detachment (17%). Schirmer dence of secondary glaucoma in 20%, also including iris or
et al26 used lower doses (< 25 Gy marginal dose), but the mini- chamber angle neovascularization.8,9,32,33 Kunaprayoon23 irradi-
mum follow-up was relatively short (median 20.9 months) and ated patients with a single fraction dose 14 to 20 Gy with the
the patients series was relatively small (14 patients). following late complications: 13% patients had neovasculariza-
Our own experience represents a group of 126 patients with tion, 22% patients had vitreous hemorrhage, 28% radiation ret-
uveal melanoma treated using the GK over a period of 8 years. inopathy, and 19% optic papillopathy.
The LENT SOMA (Late Effects Normal Tissues, subjective, objec-
tive, management, and analytic) scoring system27 was used to Complications from Radiosurgery
measure the radiation side effects (▶ Table 25.1). The tumor
marginal dose for radiosurgery ranged from 35 to 45 Gy, with a The most common late toxicity complications for all types of
median dose of 41 Gy. Tumor regression was defined as a irradiation are retinopathy, cataract, secondary glaucoma, and
decrease in tumor height registered by ultrasonography scans optic neuropathy. The most common cause of the toxicity-
and by control magnetic resonance imaging (MRI). The tumor related enucleation is secondary neovascular glaucoma. Secon-
regression was achieved in 70% of patients (▶ Fig. 25.2). The dary glaucoma is typically a late complication, and its incidence
maximum local effect has been recorded within 24 months increases with longer follow-up. Although the risk of enuclea-
after radiosurgery. Patients with tumor growth progression or tion from this reason is approximately 6% for at 2-year
without any response 30 months after treatment were candi- follow-up after radiosurgery, it increases to 19% with at 8-year
dates for retreatment by radiosurgery, and patients with a follow-up.25,34,35 Secondary glaucoma often accompanies uveal

204
Stereotactic Radiosurgery for Ocular Disorders

Fig. 25.2 (a) A 51-year-old man with uveal melanoma, 44 Gy at 50% isodose. (b) Seven years after Gamma Knife treatment—complete regression of
the melanoma with preserved vision of the eye.

melanoma before the treatment. The question remains of how and in the iris when the maximum dose did not exceed 15 Gy
much of the treatment itself contributes to its development or (incidence of late toxicity grade 3–4 in 4.6%).
accentuation. Differentiation of the natural development of this
complication from accentuation of secondary glaucoma caused
by radiosurgery is not possible at present. Prognostic Factors
Eyes with secondary enucleations presented an initial tumor The prognosis of patients with uveal melanoma is determined
height > 9 mm. The second frequent reason for enucleation was by tumor size, cell type, tumor location (poor results for ciliary
the origin of the tumor in the region of the ciliary body. 22 In a body tumors), and the extent of the disease.7,10,28,35,36 The most
prospective study of helium ion therapy, the incidence of secon- important prognostic factor is the presence or absence of organ
dary glaucoma was 29% after a mean observation period of 53 dissemination at the time of uveal melanoma diagnosis. Some
months. Incidence of rubeosis iridis in 34% of patients has been authors maintain that dissemination from uveal melanomas
observed using proton beam therapy after a median follow-up starts when the tumor is > 7 mm in diameter, and growth from
of 36 months.36 A major predictive factor for the development a 7 to 10 mm diameter increases the risk of metastatic disease
of rubeosis iridis and later neovascular glaucoma was a large to approximately 16%. This fact was supported by clinical obser-
tumor size unsuitable for brachytherapy. The enucleation vations.19 Five-year survival rates of patients with small tumors
rate was 19%: 3% for local failure and 16% because of (heights < 8 mm) and with no detected metastatic disease out-
complications.35 side the eye are > 80%. For larger tumors (> 10 mm in height)
An analysis of late toxicity of 126 patients of our series with metastases outside the eye, the 5-year survival rate ranges
recorded severe grade 3–4 retinopathy in 15% of treated from 10 to 30%. There still exists a knowledge gap regarding
patients, radiation-related late toxicity grade 3–4 in the lens growth of melanomas in situ and the documentation of the
was observed in 26%, optic neuropathy in 9%, and secondary time intervals between the diagnosis of primary tumor and the
glaucoma in 18% of cases. Eleven percent of patients were appearance of the other organ dissemination.
enucleated for worsening glaucoma during the first 2 years The best prognosis in our group of patients was for patients
after radiosurgery. The median time to occurrence of secondary with age younger than 50 years, with a preequatorial location
neovascular glaucoma was 18 months, so a higher incidence of of the tumor; tumor height not exceeding 5 mm; gross tumor
enucleation during a longer follow-up cannot be excluded. The volume not larger than 0.5 cm3; and no other organ dissemina-
worsening of useful vision caused by radiation-related toxicity tion existed.7
during 3 years after radiosurgery was observed in 34% of
patients and was due to tumor progression in 3%, optic nerve
neuropathy in 8%, retinopathy in 9%, and neovascular glaucoma Treatment Decision
in 14%. Rationale criteria for selecting a treatment modality in our cur-
Significantly lower toxicity in the optic nerve was observed rent practice are the following. Small tumors (< 10 mm in
when the maximum dose to that structure was < 10 Gy (inci- height) localized preequatorially can be successfully treated
dence of late toxicity grade 3–4 in 2.4%), in the cornea when using brachytherapy. Tumors localized equatorially or posterior
the maximum dose did not exceed 10 Gy (incidence of late tox- to the equator and not higher than 10 mm can be irradiated
icity grade 3–4 in 3%), in the lens when the maximum dose did using the GK. Tumors > 10 mm in height, especially melanomas
not exceed 7 Gy (incidence of late toxicity grade 3–4 in 7.7%), of the ciliary body, progression of growth after irradiation,

205
Stereotactic Radiosurgery for Ocular Disorders

tumors combined with neovascular glaucoma, and vitreous short. Four isocenters using the 8-mm collimator were dis-
hemorrhage are candidates for enucleation. placed in a cruciform manner ▶ Fig. 25.3. We delivered 20 Gy at
50% isodose, but for patients with some preserved vision, the
dose was lowered to 15 Gy at 50% isodose. The patients were
25.6 Glaucoma ophthalmologically followed in regular 3-month intervals dur-
Glaucoma is a chronic, slowly progressive, usually bilateral neu- ing the first year after the irradiation, then twice yearly to
ropathy of the optic nerve. Untreated glaucoma eventually assess the irradiation influence on pain, IOP, and neovasculari-
leads to complete loss of vision. About 10% of patients with zation. In a nonrandomized prospective study, a total of 107
glaucoma become unilaterally or bilaterally blind.37 We cur- eyes in 103 patients were irradiated in this way. All the patients
rently understand the pathophysiology of glaucoma to be a pro- had a long history of the disease and had exhausted all tradi-
gressive loss of ganglion cells resulting in visual field damage tional forms of therapy. Because of advanced painful glaucoma
related to the intraocular pressure (IOP).38 Although many clini- and progressive clinical symptoms, we have focused on follow-
cians now feel that there are several factors involved in the ing the decrease in pain and the effect on IOP, as well as simpli-
pathogenesis of glaucoma, the only rigorously proven treat- fying pharmacotherapy after the radiosurgery. Positive effect
ment method is the lowering of IOP. This could be achieved in was recorded in all patients in at least one of the following
two ways: by increasing the chamber-liquid drainage or by parameters. Intraocular pressure decreased usually within 12
decreasing its production. The conventional antiglaucoma weeks after the radiosurgery to the painless level in 52% of
treatment—local and systemic pharmacotherapy, laser and cry- cases; residual elevation of IOP remained in 45% of cases. Pain,
otherapy, and incisional surgery—takes effect in this way. Pro- which was present in all patients before the treatment,
duction of intraocular liquid can be decreased by affecting the regressed within 1 to 8 weeks after the treatment. Patients
site of its origin, which is the ciliary body. Irradiation of the cil- were completely pain-free in 66% of cases, and partial relief of
iary body using the GK is one such process. The intention of pain was recorded in 29% of cases. During the follow-up, which
radiosurgery is not complete destruction of the ciliary body, but was at least 2 years, there was no further deterioration of the
reduction of intraocular liquid production through action on its vision (if preserved before the treatment), and there were no
vessels. It thereby achieves a decrease of IOP. significant signs of postirradiation eye-bulb irritation, retinitis,
In secondary glaucoma, the goal of the radiosurgical treat- or changes in the anterior segment of the eye. In case of neovas-
ment is determined by the stage of the disease. When vision cularization, at least partial regression was observed in all
has been totally or partially lost, the main aims are to abolish affected eyes, usually 4 to 5 months after the radiosurgery.
the elevated IOP and to eliminate severe pain. In the early stages Pharmacotherapy was reduced in 39% of patients within 2 to 3
of the disease, when conventional therapy has insufficient months after the radiosurgery. Enucleation was performed in
effect and visual acuity is still preserved, the treatment has the eight blind eyes: in five of them the radiosurgery was not suffi-
more difficult task of preventing any further progress of the ciently effective, in two eyes the reason was cosmetic, and in
pathogenic mechanisms. one eye there was a trauma during the follow-up.
Because there was no previous experience in the radiosurgi- Treatment complications were not significant: short-term
cal treatment of glaucoma to refer to, we started to evaluate an postoperative lacrimation in 61% of patients caused by mechan-
effective and safe irradiation of the corpus ciliare as the source ical irritation after the eye fixation, postirradiation cataracts in
of intraocular aqueous humor.39,40 Gamma Knife stereotactic two patients, and noninfectious keratitis in another two
radiosurgery (SRS) of the ciliary body led to a significant allevi- patients.
ation of pain and reduction of IOP in advanced glaucoma. It was Patients with primary open-angle glaucoma suffer less pain
effective in secondary glaucoma, especially in terms of neovas- or no pain, their IOP is less elevated, and their disease is mainly
cularization. The latency of the treatment effect was relatively expressed by defects in their visual field as a manifestation of

Fig. 25.3 A plan to irradiate the ciliary body using four 8-mm collimators (50% isodose displayed) in the eye bulb with aphakia; therefore, the lens is
not visible on magnetic resonance imaging.

206
Stereotactic Radiosurgery for Ocular Disorders

the optic neuropathy. A prospective 5-year clinical study to halt serous detachment of the retina surrounding the process does
progressive optic neuropathy in the early stages of glaucoma not have any influence on the initial effect. Even where massive
had been concluded in our department, but the effects of radio- serous retinal detachment in the macular region has occurred,
surgery on patients with primary glaucoma does not seem to we have found the retina to be completely reattached after
be permanent and the results of GKSRS did not meet all our radiosurgery. However, GK treatment can stabilize the activity
expectations with recurrences in the majority of treated of CNVM, and in a majority of our patients, it can also stabilize
patients. and increase visual functions (or subjective evaluation of these
functions) for a few months. In nearly half of all patients the
recurrence of neovascularization was observed over a period of
25.7 Age-Related Macular 18 to 24 months after radiosurgery and the recurrence of neo-
vascularization was observed after 3 years in three quarters of
Degeneration the patients. Overall results of radiosurgery in this studied
Age-related macular degeneration (ARMD) is the most common group of patients have not reached all expectations and the
cause of legal blindness in patients 50 years or older. The two effect in the majority of patients was temporary.
main forms are nonexudative and exudative. Exudative ARMD
forms abnormal blood vessels called choroidal neovascular
25.7.1 Rare Ocular Disorders Treated by
membranes (CNVMs), which develop under the retina, leak flu-
id and blood, and ultimately cause a blinding disciform scar in Radiosurgery
and under the retina.
Retinoblastoma
Age-related macular degeneration is a degenerative process
with a proliferative component, and irradiation can play an Retinoblastoma represents the most common primary malig-
antiproliferative role. Choroidal neovascular membranes, which nant tumor of the eye in young children (in the first 2 years of
are composed of endothelial cells and proliferate more rapidly life). The treatment strategy depends on clinical staging (dis-
than the endothelial cells of the retina, may be more sensitive ease confined to the retina, disease confined to the globe, extra-
than the retinal vasculature. Consequently, radiation therapy ocular extension, and presentation of distant metastases) and
has been suggested as a treatment for subfoveal CNVMs. Many includes tumor resection, enucleation, chemotherapy, and
pilot studies for exudative ARMD show stabilization or regres- external beam radiotherapy.
sion of CNVMs with different kinds of radiation therapy.41–50 Our experience covers treatment of four children who were
Initial results from patients treated with radiosurgery for admitted for stereotactic irradiation using the GK and had vitre-
ARMD show that the GK is able to affect tissues beneath the ret- ous seeding of malignant cells. All patients were pretreated by
ina without damaging the overlying retinal structures. Haas et the International Society of Pediatric Oncology protocol and
al47 in their pilot study investigated the effect of single-fraction relapsed after standard treatment. The only remaining possi-
(10 Gy at the 90% isodose). Gamma Knife SRS in patients with bility was enucleation. The whole vitreous body was irradi-
subfoveal CNVMs due to ARMD. Ten patients were followed for ated using the GK in a single session, with a collimator
12 months. Fluorescein angiography and indocyanine green diameter of 14 mm. To avoid growth retardation of the irradi-
chorioangiography demonstrated a regression of the neovascu- ated eye and bony structures, the minimal dose delivered to
lar complex in one patient and stabilization in three patients. the eye globe did not exceed 15 Gy in all these patients. For
Enlargement of the CNVM was found in six patients and was this reason, and also bearing in mind the risk of complications
associated with a decrease of visual acuity in four patients. Hen- of previous treatment, the applied doses were relatively low.
derson et al50 followed seven patients with ARMD after GKSRS Local progression was observed in three out of four patients,
using a marginal dose of 12 Gy. With a median patient follow- and three patients were enucleated a median 2 months after
up of 2 years, they observed stabilization of disease in five radiosurgery.
patients; improvement of vision was recorded in one, and
impairment also in one patient. Orbital and Uveal Metastases of Carcinomas
We have constructed a prospective, nonrandomized study in
a group of 30 patients. A dose of 30 Gy in the center and a dose Metastatic lesions in every location of the eye and the orbit
of 15 Gy to the periphery of CNVM at 50% isodose were applied have been described, and autopsy studies estimate the inci-
in all 30 patients. Gamma Knife therapy in the studied group of dence of these metastases to be 4 to 12% in all types of cancer
patients was safe. No severe radiation-related side effects, such patients.51 Radiosurgery may extend treatment options for
as optic neuropathy, cataract formation or keratitis, were uveal metastases because of its specific advantages: the possi-
observed over a 2-year follow-up. bility of applying a high radiation dose and evidence of its spar-
Stabilization and regression of the choroidal neovasculariza- ing effect on the surrounding critical structures. The
tion (CNV) process was closely related to a reduction of activity, recommended minimal doses are similar to those for brain
subretinal fluid, and to a decrease of leakage. We have observed metastases, ranging from 20 to 30 Gy—depending on the tumor
that the most prominent initial response to the treatment can volume, location, and histology.
be expected in active (serous and/or hemorrhagic activity of
the membrane) well-defined membranes with a size < 10 mm
(volume of irradiated tissue under 300 mm3). The type of the
25.8 Other Ocular Disorders
membrane also seems to be important, with a major effect in We have recorded positive experience with treatment of ocular
predominantly occult forms. On the other hand, the degree of hemangioblastomas (▶ Fig. 25.4). Four patients with von

207
Stereotactic Radiosurgery for Ocular Disorders

Fig. 25.4 (a) A 16-year-old patient with hemangioblastoma and bulous elevation of retina (hemangioblastoma is covered by a 70% isodose
representing 15 Gy): 1 = lens, 2 = corpus vitreum, 3 = bulous retinal detachment, 4 = tumor, 5 = build-up filled with gel. (b) Four years after the
treatment the tumor and the retinal detachment have completely regressed.

Hippel-Lindau disease suffering from retinal or choroidal


hemangioblastomas were treated using GKSRS. Radiosurgery
25.10 Acknowledgments
had a stable effect, with follow-up exceeding 6 years in all We thank our colleagues from the Department of Ophthalmol-
patients, even when several lesions in other organs progressed ogy, first medical faculty, Charles University and the Central
and had to be surgically treated. Regression of the lesion vol- Military Hospital, Prague: J. Pašta, MD, PhD; J. Vladyková, MD,
ume by 50% or more was observed. DSc; J. Ernest, MD, PhD; P. Němec, MD; and L Nováček, MD, for
We irradiated the ciliary ganglion in a 32-year-old woman their care for our patients.
suffering from severe vegetative orbital pain after a longer his-
tory of penetrating bulbar injury, three previous operations for
amotio retinae, and a cyclocryocoagulation for secondary glau- References
coma. The affected eye was blind. The pain disappeared within
a week. Two and a half years after treatment, the patient was [1] Langmann G, Pendl G, Schröttner O. Gamma Knife of uveal melanoma radio-
surgery for intraocular melanomas: preliminary report. Spectrum Augen-
still pain-free after several years of previous continual suffering.
heilkd 1995; 9 (Suppl 1): 16–21
A positive treatment result was observed in a case of spinocel- [2] Marchini G, Gerosa M, Piovan E, et al. Gamma Knife stereotactic radiosurgery
lular carcinoma of the lacrimal saccus. for uveal melanoma: clinical results after 2 years. Stereotact Funct Neurosurg
Radiosurgery proved to be effective in the treatment of 1996; 66 (Suppl 1): 208–213
[3] Mueller AJ, Talies S, Schaller UC, Horstmann G, Wowra B, Kampik A. Stereo-
exophthalmos refractory to conservative treatment. Irradiation
tactic radiosurgery of large uveal melanomas with the gamma-knife. Oph-
of intraorbital muscles or inflammatory intraorbital tissue led thalmology 2000; 107: 1381–1387, discussion 1387–1388
to a regression of exophthalmos within 3 months of the [4] Pochop P, Pilbauer J, Krepelková J, et al. [2 years’ experience with treatment
treatment. of uveal melanoma using the Leksell gamma knife]. Cesk Slov Oftalmol 1998;
54: 222–234
[5] Rand RW, Khonsary A, Brown WJ, Winter J, Snow HD. Leksell stereotactic ra-

25.9 Summary diosurgery in the treatment of eye melanoma. Neurol Res 1987; 9: 142–146
[6] Rennie I, Forster D, Kemeny A, Walton L, Kunkler I. The use of single fraction
Leksell stereotactic radiosurgery in the treatment of uveal melanoma. Acta
Radiosurgery of ocular tumors and other eye or orbital lesions
Ophthalmol Scand 1996; 74: 558–562
represents a reasonable therapeutic option. If successful, it can [7] Simonová G, Novotný J Jr, Liscák R, Pilbauer J. Leksell gamma knife treatment
help to preserve the eye globe and visual function. Further clin- of uveal melanoma. J Neurosurg 2002; 97 (Suppl): 635–639
ical studies using radiosurgery in ocular disease are necessary [8] Zehetmayer M, Kitz K, Menapace R, et al. Local tumor control and morbidity
to optimize effective doses and to analyze factors influencing after one to three fractions of stereotactic external beam irradiation for uveal
melanoma. Radiother Oncol 2000; 55: 135–144
the length of survival, with a careful evaluation of the side
[9] Bellmann C, Fuss M, Holz FG, et al. Stereotactic radiation therapy for malig-
effects, as well as to improve local treatment effects while nant choroidal tumors: preliminary, short-term results. Ophthalmology
decreasing radiation-related complications. 2000; 107: 358–365

208
Stereotactic Radiosurgery for Ocular Disorders

[10] Courdi A, Caujolle JP, Grange JD, et al. Results of proton therapy of uveal mela- [32] Dieckmann K, Georg D, Zehetmayer M, Bogner J, Georgopoulos M, Pötter R.
nomas treated in Nice. Int J Radiat Oncol Biol Phys 1999; 45: 5–11 LINAC based stereotactic radiotherapy of uveal melanoma: 4 years clinical ex-
[11] Debus J, Fuss M, Engenhart-Cabillic R, et al. [Stereotactic conforming irradia- perience. Radiother Oncol 2003; 67: 199–206
tion of choroid metastases]. Ophthalmologe 1998; 95: 163–167 [33] Muller K, Naus N, Nowak PJ, et al. Fractionated stereotactic radiotherapy
[12] Liscák R, Vladyka V. Radiosurgery in ocular disorders: clinical applications. for uveal melanoma, late clinical results. Radiother Oncol 2012; 102: 219–
Prog Neurol Surg 2007; 20: 324–339 224
[13] Novotny J Jr, Novotny J, Liscák R, et al. Assessment of the accuracy in ophthal- [34] Collaborative Ocular Melanoma Study Group. Histopathologic characteristics
mic radiosurgery. In: Kondziolka D, ed. Radiosurgery. Vol. 6. Basel, Switzer- of uveal melanomas in eyes enucleated from the Collaborative Ocular Mela-
land: Karger; 2006:71–85 noma Study. COMS report no. 6. Am J Ophthalmol 1998; 125: 745–766
[14] Petersch B, Bogner J, Dieckmann K, Pötter R, Georg D. Automatic real-time [35] Castro JR, Char DH, Petti PL, et al. 15 years experience with helium ion radio-
surveillance of eye position and gating for stereotactic radiotherapy of uveal therapy for uveal melanoma. Int J Radiat Oncol Biol Phys 1997; 39: 989–996
melanoma. Med Phys 2004; 31: 3521–3527 [36] Char DH, Kroll SM, Castro J. Ten-year follow-up of helium ion therapy for
[15] Tokuuye K, Akine Y, Sumi M, Kagami Y, Ikeda H, Kaneko A. Fractionated ster- uveal melanoma. Am J Ophthalmol 1998; 125: 81–89
eotactic radiotherapy for choroidal melanoma. Radiother Oncol 1997; 43: [37] Alfonso A. Relationship of structural and functional measurements. In: Wein-
87–91 reb RN, Kitazawa Y, Kriegelstein GK, eds. Glaucoma in the 21st Century. Lon-
[16] Zehetmayer M, Menapace R, Kitz K, Ertl A. Suction attachment for stereotac- don, UK: Mosby International; 2000:57–65
tic radiosurgery of intraocular malignancies. Ophthalmologica 1994; 208: [38] Harwerth RS, Carter-Dawson L, Shen F, Smith EL III, Crawford ML. Ganglion
119–121 cell losses underlying visual field defects from experimental glaucoma. Invest
[17] Egan KM, Seddon JM, Glynn RJ, Gragoudas ES, Albert DM. Epidemiologic as- Ophthalmol Vis Sci 1999; 40: 2242–2250
pects of uveal melanoma. Surv Ophthalmol 1988; 32: 239–251 [39] Vladyka V, Liscák R, Subrt O, et al. Initial experience with gamma knife radio-
[18] Singh AD, Topham A. Incidence of uveal melanoma in the United States: surgery for advanced glaucoma. J Neurosurg 2000; 93 (Suppl 3): 180–183
1973–1997. Ophthalmology 2003; 110: 956–961 [40] Vladyka V, Liscák R, Simonová G, Pilbauer J, Hejduková I, Novácek L. Progress
[19] Manschot WA, van Strik R. Uveal melanoma: therapeutic consequences of in glaucoma treatment research: a nonrandomized prospective study of 102
doubling times and irradiation results; a review. Int Ophthalmol 1992; 16: patients with advanced refractory glaucoma treated by Leksell gamma knife
91–99 irradiation. J Neurosurg 2005; 102 (Suppl): 214–219
[20] Fakiris AJ, Lo SS, Henderson MA, et al. Gamma-knife-based stereotactic radio- [41] Bergink GJ, Deutman AF, van den Broek JE, van Daal WA, van der Maazen RM.
surgery for uveal melanoma. Stereotact Funct Neurosurg 2007; 85: 106–112 Radiation therapy for age-related subfoveal choroidal neovascular mem-
[21] Woodburn R, Danis R, Timmerman R, et al. Preliminary experience in the branes. A pilot study. Doc Ophthalmol 1995; 90: 67–74
treatment of choroidal melanoma with gamma knife radiosurgery. J Neuro- [42] Berson AM, Finger PT, Sherr DL, Emery R, Alfieri AA, Bosworth JL. Radiother-
surg 2000; 93 (Suppl 3): 177–179 apy for age-related macular degeneration: preliminary results of a potentially
[22] Zehetmayer M, Menapace R, Kitz K, Ertl A, Strenn K, Ruhswurm I. Stereotactic new treatment. Int J Radiat Oncol Biol Phys 1996; 36: 861–865
irradiation of uveal melanoma with the Leksell gamma unit. Front Radiat Ther [43] Valmaggia C, Ries G, Ballinari P. Radiotherapy for subfoveal choroidal neovas-
Oncol 1997; 30: 47–55 cularization in age-related macular degeneration: a randomized clinical trial.
[23] Kunaprayoon D, McKenzie J, Offeman S, et al. Stereotactic radiosurgery in the Am J Ophthalmol 2002; 133: 521–529
treatment of ocular melanoma. Int J Radiat Oncol Biol Phys 2012; 83 (Suppl [44] Finger PT, Berson A, Ng T, Szechter A. Ophthalmic plaque radiotherapy for
1): S284 age-related macular degeneration associated with subretinal neovasculariza-
[24] Toktas ZO, Bicer A, Demirci G, et al. Gamma knife stereotactic radiosurgery tion. Am J Ophthalmol 1999; 127: 170–177
yields good long-term outcomes for low-volume uveal melanomas without [45] Weinberger AW, Wolf S, Kube T, et al. [Radiation therapy of occult choroidal
intraocular complications. J Clin Neurosci 2010; 17: 441–445 neovascularisation (CNV) in age-related macular degeneration (AMD)]. Klin
[25] Wang Z, Nabhan M, Schild SE, et al. Charged particle radiation therapy for Monatsbl Augenheilkd 1999; 214: 96–99
uveal melanoma: a systematic review and meta-analysis. Int J Radiat Oncol [46] Chakravarthy U, Houston RF, Archer DB. Treatment of age-related subfoveal
Biol Phys 2013; 86: 18–26 neovascular membranes by teletherapy: a pilot study. Br J Ophthalmol 1993;
[26] Schirmer CM, Chan M, Mignano J, et al. Dose de-escalation with gamma knife 77: 265–273
radiosurgery in the treatment of choroidal melanoma. Int J Radiat Oncol Biol [47] Haas A, Papaefthymiou G, Langmann G, et al. Gamma knife treatment of sub-
Phys 2009; 75: 170–176 foveal, classic neovascularization in age-related macular degeneration: a pilot
[27] LENT SOMA tables. Radiother Oncol 1995; 35: 17–60 study. J Neurosurg 2000; 93 (Suppl 3): 172–176
[28] Egger E, Zografos L, Giotein G. Proton beam irradiation of choroidal melano- [48] Spaide RF, Guyer DR, McCormick B, et al. External beam radiation therapy for
ma: techniques and results. In: Sagerman RH, Alberti WE, eds. Radiotherapy choroidal neovascularization. Ophthalmology 1998; 105: 24–30
of Intraocular and Orbital Tumors. 2nd rev. ed. Berlin, Germany: Springer- [49] Kishan AU, Modjtahedi BS, Morse LS, Lee P. Radiation therapy for neovascular
Verlag; 2003:67–80 age-related macular degeneration. Int J Radiat Oncol Biol Phys 2013; 85:
[29] Dendale R, Lumbroso-Le Rouic L, Noel G, et al. Proton beam radiotherapy for 583–597
uveal melanoma: results of Curie Institut-Orsay proton therapy center (ICPO). [50] Henderson MA, Valluri S, Lo SS, et al. Gamma knife radiosurgery in the treat-
Int J Radiat Oncol Biol Phys 2006; 65: 780–787 ment of choroidal neovascularization (wet-type macular degeneration). Ster-
[30] Caujolle JP, Paoli V, Chamorey E, et al. Local recurrence after uveal melanoma eotact Funct Neurosurg 2007; 85: 11–17
proton beam therapy: recurrence types and prognostic consequences. Int J [51] Bloch RS, Gartner S. The incidence of ocular metastatic carcinoma. Arch Oph-
Radiat Oncol Biol Phys 2013; 85: 1218–1224 thalmol 1971; 85: 673–675
[31] Caujolle JP, Mammar H, Chamorey E, Pinon F, Herault J, Gastaud P. Proton
beam radiotherapy for uveal melanomas at nice teaching hospital: 16 years’
experience. Int J Radiat Oncol Biol Phys 2010; 78: 98–103

209
Stereotactic Radiosurgery for Pediatric Brain Tumors

26 Stereotactic Radiosurgery for Pediatric Brain Tumors


Christian C. Okoye, Ravi B. Patel, David B. Mansur, Alia Hdeib, Arjun Sahgal, Eric L. Chang, Mitchell Machtay, John H. Suh, Andrew E. Sloan, and
Simon S. Lo

26.2 Technical Issues Specific to


Key Points
Stereotactic Radiosurgery in
● Stereotactic radiosurgery is an emerging technology in the
treatment of pediatric brain tumors, particularly given the fa- Pediatric Patients
vorable dose-volume characteristics relative to external beam Unless a frameless radiosurgical system is used, rigid fixation is
radiation therapy. typically required in single-fraction SRS, thus allowing the mar-
● There are several technical issues specific to stereotactic ra- gin around the target volume to be minimized during treat-
diosurgery in pediatric patients that must be accounted for, ment planning. As opposed to adults, where rigid fixation of
particularly with regards to head frame placement, anesthe- the skull is readily achievable, younger children have thin
sia/sedation, and associated treatment times. skulls, which may not be completely fused. An alternative to
● The role of stereotactic radiosurgery in many pediatric insertion of pins into the outer skull table is the fabrication of a
brain tumors is poorly defined, as the literature consists al- cast or Aquaplast with piers where the pins can be inserted2
most exclusively of single-institution retrospective experi- (▶ Fig. 26.1). Due to their small body size, pediatric patients
ences. may also require body casts for additional neck support.
● The frequency and severity of normal tissue toxicities and Gamma Knife stereotactic radiosurgery (GKSRS) is typically a
complications of treatment are incompletely characterized, single-day procedure, consisting of image acquisition, complex
and caution should be exercised when utilizing these ap- treatment planning, and treatment delivery. Depending on a
proaches outside of the clinical trial setting. variety of treatment-related and logistical factors, including the
complexity of the planning, the treatment-planning system
used, hospital layout, and patient transportation, the time
required can vary significantly. In particular, the treatment time
26.1 Introduction can be prolonged in circumstances where older cobalt sources
are used within a GK unit or multiple small shots (isocenters)
Primary brain tumors constitute 20% of all pediatric cancers
are utilized.
and are the most common solid tumors in childhood.1,2 Sur-
Conscious sedation and/or general anesthesia may be
gical resection is usually the mainstay of treatment, and
required for younger children and those who are unable to
maximal safe resection is the goal in most cases. However,
withstand being immobile for the whole radiosurgical
conventional fractionated radiation therapy is often indicated
for high-grade or incompletely resected or unresectable
tumors.
Stereotactic radiosurgery (SRS) refers to a strategically
delivered high dose of radiation to a target volume using
megavoltage photons generated by a linear accelerator
(linac), gamma rays from a Gamma Knife (GK; Elekta Instru-
ments AB, Stockholm, Sweden) unit, or protons generated by
a cyclotron or synchrotron unit, 1,2 typically performed in one,
but up to five, session(s). Stereotactic radiosurgery has been
used extensively to treat intracranial tumors in adults and
has become one of the standard treatments for certain
tumors. In contrast, there are relatively limited data on the
use of SRS in the treatment of pediatric brain tumors. In this
population, there is an added emphasis on limiting the mor-
bidity of radiation-induced damage to the brain, which in
some patients may lead to neurocognitive, neurologic, and
neuroendocrine dysfunction. As a result of its favorable dosi-
metric characteristics, SRS represents a very attractive treat-
ment modality for some pediatric patients with brain
tumors. Through this modality, the dose and volume of nor-
mal brain irradiated can be reduced, and subsequently the Fig. 26.1 An Aquaplast was fabricated with piers where the pins can be
toxicities that are critically dependent on these two factors inserted for fixation. (From Witt TC, Lo SS, Timmerman RD. Successful
can minimized. The physics and radiobiology of SRS are cov- treatment of a skull base malignant rhabdoid tumor with surgery,
ered in other chapters of this book. In this chapter, we focus chemotherapy and Gamma Knife-based stereotactic radiosurgery in a
on the clinical applications and results of single-fraction SRS young child. Stereotact Funct Neurosurg. 2007;85:310–313. Used with
permission from Karger AG, Basel.)
in the treatment of pediatric brain tumors.

210
Stereotactic Radiosurgery for Pediatric Brain Tumors

procedure.3 To ensure the delivery of high-quality treatment, incompletely resected tumors. In the Pediatric Oncology Group
patients need to remain still and cooperative during head frame (POG) 9132 study, where radiation dose escalation to 69.6 Gy
placement, acquisition of computed tomography (CT) or mag- was achieved through hyperfractionation, the 19 patients who
netic resonance images (MRIs), and treatment delivery.1,2 Thus, underwent subtotal resection had superior survival outcomes
prolonged anesthesia of those children undergoing SRS can be compared with historic controls from the previous POG 8532
expected. When treatment planning is performed, the neuro- trial.5 Given SRS can deliver a single focused high dose of radia-
surgeon and radiation oncologist must strike a balance between tion to a target volume, it has been used as a boost therapy after
the time required in treatment planning and delivery against EBRT for these high-risk patients.
the extraconformality achieved with more complex planning. Investigators from St. Jude Children’s Research Hospital
During treatment delivery, to avoid unnecessary radiation (Memphis, TN) reported their early experience with this
exposure to support staff in the treatment area, patients are approach. Five pediatric patients with residual enhancing epen-
usually monitored by the anesthesiologist in the control area. A dymoma measuring < 3 cm received a boost with linac-based
new model of GK, the Perfexion (Elekta AB), allows for short- SRS after EBRT to doses of 50.4 to 55.8 Gy. 10 The SRS dose was
ened treatment and planning times by automating treatment 10 Gy (range 9–15 Gy) prescribed to the 80% isodose line, deliv-
delivery and adding inverse treatment-planning functionality, ered within the first 30 days after completing EBRT. With a
features that may be beneficial for pediatric patients receiving median follow-up time of 24 months, 80% survived with no evi-
SRS. dence of tumor progression, whereas complete responses were
A frameless robotically controlled radiosurgical system such achieved in three patients (60%). Radionecrosis occurred in one
as CyberKnife (Accuray, Inc., Sunnyvale, CA) can potentially (20%) of the five patients. In a pediatric SRS series from Harvard
solve some of the problems associated with frame-based radio- University (Boston, MA), 3 of the 28 patients with ependymoma
surgical systems. Other linear accelerator– (linac-)based sys- treated with SRS received it as a boost therapy after EBRT. 11
tems allow for frameless stereotactic delivery too. In particular, Two (67%) of the three patients survived without evidence of
given the image acquisition, treatment planning, and treatment disease progression at 30 and 62 months after radiosurgery. In
delivery that occur during separate sessions, the time required a series of nine patients treated with SRS for ependymoma from
for the patient to be sedated may be substantially reduced, and Washington University (St. Louis, MO), three were treated with
conscious sedation methods may be explored. The feasibility, SRS as a boost therapy after EBRT to doses of 45.0 to 53.6 Gy. 12
safety, and efficacy of the use of this system in the treatment of With follow-up times ranging from 56 to 91 months, the
brain tumors in pediatric patients, including very young chil- relapse-free survival rate was 100%. Two (67%) of the three
dren, have been demonstrated.4 patients developed complications (seizures in one patient and
facial nerve palsy in the other) after SRS. In the series from Indi-
ana University (Indianapolis, IN), two pediatric patients were
26.3 Treatment Options for treated with SRS as boost therapy after EBRT to doses of 54.0 to
55.8 Gy.13 Both patients remained progression-free at 40 and 65
Specific Tumors months, respectively. One of the two patients developed radio-
26.3.1 Ependymomas necrosis, which was controlled with hyperbaric oxygen and ste-
roid therapy.
Ependymomas account for 5 to 10% of all pediatric brain In summary, the combination of EBRT and boost with SRS is
tumors.5 Histologically, ependymomas are well-delineated, associated with excellent local control of gross residual disease
moderately cellular gliomas that have a sharp demarcation in ependymoma, and may result in long-term progression-free
from the surrounding brain parenchyma. Because of this char- survival (PFS). Late radiation complications may occur, particu-
acteristic, they are very well suited for SRS, where the pre- larly radionecrosis in the setting of dose escalation. Currently,
scribed isodose conforms highly to the target volume, and there there are no prospective data supporting this approach, and a
is a rapid falloff of the radiation dose beyond the target volume. prior POG phase II trial involving the use of SRS as a boost ther-
Although not specifically categorized by Larson et al, based on apy for pediatric patients with high-risk ependymoma was
their characteristics, ependymomas can be regarded as class IV closed, secondary to poor accrual. Future trials testing this
targets (tumor composed of early-responding tissue with no approach may be worthwhile to accurately delineate the risks
intermingling normal brain tissue).6 and benefits.
The most important factor that determines survival for
patients with ependymomas is the extent of surgical resec-
tion,5,7,8 and the presence of gross disease predicts a poor prog- 26.3.3 Salvage Therapy for Recurrent
nosis. The use of SRS in pediatric ependymoma is mainly in
three settings, namely, as a boost after external beam radiation
Disease
therapy (EBRT), as a salvage treatment of recurrent disease, and The recommended treatment for patients with recurrent epen-
less commonly, as the sole treatment. dymoma after prior surgery and postoperative EBRT is salvage
surgical resection. However, a complete resection may not be
safely achievable in some cases. Although chemotherapy may
26.3.2 Boost Therapy also be tried, tumor responses are typically suboptimal. Stereo-
In pediatric patients with ependymomas treated in the primary tactic radiosurgery provides an additional salvage option par-
setting, a radiation dose response has been demonstrated. 8,9 ticularly suited for patients with recurrent ependymoma in
This is particularly relevant in patients with high-risk, whom complete resection is not feasible.

211
Stereotactic Radiosurgery for Pediatric Brain Tumors

In an early series from the University of Pittsburgh (Pitts- adult and pediatric patients with 49 lesions were treated with
burgh, PA), seven pediatric patients with recurrent ependymo- GKSRS.16 Nineteen patients (73%) had received prior cranial
ma (grade not specified) were treated with GKSRS. The external beam radiotherapy to a median dose of 54 Gy, and a
reported recurrence rate was 100%, and the time to progression single patient developed an intracranial lesion after presenting
was 6 months.14 In a subsequent study from the same institu- with a spinal ependymoma. The median prescribed dose was
tion, 21 pediatric patients with recurrent or progressive epen- 18 Gy (range 12–24 Gy). After a median follow-up of 3.1 years,
dymoma (12 patients) or anaplastic ependymoma (9 patients) the 3-year local control for all lesions treated was 72%, whereas
were treated with GKSRS, to a median dose of 15 Gy (range 9– 38 and 27% of patients developed marginal and distant failures,
22 Gy).15 At a median follow-up of 27.6 months, local control respectively. Pathologically confirmed radiation necrosis was
was achieved in 72% of treated tumors, although distant intra- seen in two patients (7.7%). In the Indiana University series, five
cranial relapse occurred in 10 out of 21 patients (47%), and the patients (two children) with 10 recurrent ependymomas were
3-year distant failure rate after SRS was 80.3%. Ipsilateral facial treated with GKSRS.13 Out of the five patients who received SRS
paresis developed in a single patient (4.8%) after a marginal as salvage treatment, three (60%) were alive, two (40%) were
dose of 12 Gy and subsequently resolved after a short course of alive without recurrence, two (40%) developed distant failure,
steroids. Hodgson et al from Harvard University reported that and three (60%) had in-field control. The 3-year in-field control
among 28 pediatric patients treated with SRS for ependymoma for the 10 recurrent tumors was 62.5%. ▶ Fig. 26.2 shows a case
(3 treated with SRS as boost therapy after EBRT and the remain- example of SRS for treatment of resected recurrent anaplastic
ing treated for recurrent disease), the median PFS was 8.5 ependymoma.
months, and the 3-year local control was only 29%.11 Out of the In summary, children presenting with recurrent ependymo-
25 patients treated for recurrent disease, only 3 remained dis- mas not amenable to complete surgical resection have very few
ease-free. In a series from the Mayo Clinic (Rochester, MN), 26 effective treatment options. Local tumor progression is

Fig. 26.2 A 2-year-old child exhibited a localized


posterior fossa anaplastic ependymoma. The
patient underwent gross total resection and
postoperative radiotherapy to the operative bed
with a dose of 54 Gy in 30 fractions. He
developed recurrence in the posterior fossa (a)
and lumbar spine region and underwent gross
total resection of tumor in the posterior fossa (b)
and lumbar spine. To avoid pin insertions
associated with Gamma Knife–based stereotactic
radiosurgery in this very young child, CyberKnife
was used instead to deliver stereotactic radio-
surgery to the tumor bed and a dose of 12 Gy in
one fraction prescribed at 80% (c). There was no
evidence of disease after 18 months.

212
Stereotactic Radiosurgery for Pediatric Brain Tumors

uniformly fatal, and currently available chemotherapy has been (WHO grade I) represent the most indolent subtype of low-
used with limited success. Stereotactic radiosurgery provides a grade astrocytomas, with 10-year survival rates of over 95% in
minimally invasive, localized treatment modality, with the lim- the pediatric population.17 Diffuse astrocytomas (WHO grade II)
ited available data suggesting that some patients may achieve carry a slightly worse prognosis in the pediatric population,
objective responses and prolonged local control. However, mar- with 10-year survival rates of approximately 80%.17 The main-
ginal and distant central nervous system (CNS) relapses remain stay of treatment is maximal safe surgical resection, if achiev-
significant issues; thus, more effective chemotherapy treat- able. If a complete resection is not achieved, chemotherapy is
ments need to be developed. often given in young children to delay the need for immediate
postoperative radiation therapy due to concerns of collateral
damage it may cause to the developing brain. However, if gross
26.3.4 Stereotactic Radiosurgery as the total resection or subtotal resection can be achieved, observa-
Sole Postoperative Treatment tion may be appropriate. In the recurrent setting, treatment
options may be more limited.
Stereotactic radiosurgery is seldom used as the sole, postopera-
As described by Larson et al, low-grade astrocytomas or glio-
tive intervention for gross disease in ependymoma because
mas are classified as category III targets, which are defined as
most patients usually undergo EBRT. Rare exceptions are lim-
early-responding targets intermingled with late-responding
ited to patients in whom the morbidity of EBRT has been
normal brain parenchyma.6 It is more common to have normal
deemed excessive; there is a scarcity of data in the literature on
brain parenchyma intermingled with the target tissue in WHO
this approach. In a series from Indiana University, a 16-month-
grade II astrocytomas than those which are WHO grade I. Based
old child who had partial response to chemotherapy alone for a
on these characteristics, low-grade astrocytomas are not
subtotally resected cerebellopontine angle ependymoma
regarded as ideal targets for SRS given the large amount of nor-
received SRS as the sole radiotherapeutic treatment, with the
mal brain tissue that would need to be treated to encompass
goal of delaying EBRT. The ependymoma progressed after 13
the entirety of known disease.6 Stereotactic radiosurgery has
months, and salvage treatment with further chemotherapy and
been used in the treatment of low-grade astrocytoma in chil-
conformal radiation therapy was given.13
dren in select clinical scenarios, but the data on its safety and
Given the lack of robust data from the literature documenting
efficacy remain limited.
its efficacy and safety in the up-front, postoperative setting, SRS
In one of the largest experiences with SRS in pediatric low-
should not be offered in this fashion outside of a clinical trial
grade astrocytomas, investigators from the University of Pitts-
setting. ▶ Table 26.1 summarizes the treatment outcomes of
burgh described 37 patients (25 pediatric patients) with recur-
selected series.
rent or unresectable pilocytic astrocytoma treated with SRS. 18
Eighteen patients had brainstem tumors. The prescribed dose
26.4 Low-Grade Astrocytomas was 15 Gy. At a median follow-up of 28 months, 10 had com-
plete response, 8 had partial response, 7 had stable findings,
World Health Organization (WHO) grade I and II astrocytomas and 12 had delayed tumor progression on imaging studies,
are collectively known as low-grade astrocytomas, which occur resulting in a crude survival rate of 89%. There was no treat-
in both adult and pediatric patients. Pilocytic astrocytomas ment-related mortality. The researchers also reported their

Table 26.1 Summary of treatment outcomes of stereotactic radiosurgery for ependymoma in children
Study N Histology Marginal dose (Gy) Setting of treatment Local control (%)

Aggarwal et al10 5 Ependymoma 10 Boost 80

Grabb et al14 7 Ependymoma 11–20a Salvage 0


(grade not specified)

Kano et al15 21 (32 lesions) Ependymoma (12 pa- 15 Salvage 72%


tients), Anaplastic
ependymoma (9 pa-
tients)

Hodgson et al11 28 Ependymoma 12.5 Salvage and boost 29b

3 Not specified Boost 67

Stafford et al73 12 (adults and chil- Ependymoma 18 Salvage 68


dren)

Lo et al13 5 (2 children) Ependymomac 12–20 Salvage 60d

2 12–14 Boost 100


aDose range for the whole group of 25 children (including those with other histologies).
bLocal control for all 29 patients.
cOne patient had anaplastic ependymoma.
dLocal control for all five patients.

213
Stereotactic Radiosurgery for Pediatric Brain Tumors

treatment outcomes of SRS for WHO grade II fibrillary astrocy- who were treated with GKSRS.22 Out of the 51 patients, 12 had
toma.19 Twelve patients (four children) with progressive disease WHO grade I astrocytomas around the optic pathway or hypo-
were treated with SRS to a median dose of 16 Gy and a volume thalamus, whereas the remaining patients had WHO grade II
ranging from 1.2 to 45.1 mL. Four of the 12 patients had tumors astrocytomas. The mean age for patients with grade I astrocyto-
in the brainstem. At a median follow-up of 52 months, all mas was 9.8 years. The mean prescribed doses for grade I and II
patients were alive. Five patients had tumor shrinkage (one tumors were 12.5 and 15.7 Gy, respectively, with respective
complete and four partial responses), three had stable findings, tumor diameters of 2.54 and 2.37 cm. With a median follow-up
and four had delayed progression. The crude freedom from pro- period of 27.6 months, the response rates for grade I and II
gression (FFP) rate was 67%. astrocytomas were 50.0 and 46.2%, respectively.22 The corre-
Investigators from Taiwan also reported on their experience sponding tumor control rates were 91.7 and 87.2%, respec-
with 21 adult and pediatric patients (25 lesions) treated with tively.22 There was a significantly better response among
GKSRS for low-grade gliomas, predominantly as salvage treat- patients 10 years of age or older with grade I tumors and those
ment (72%), but also as primary treatment (19%) or boost after with a follow-up period of more than 24 months. Radiation-
EBRT (9%).20 The median peripheral tumor dose was 14.5 Gy induced edema occurred in 18 (35.3%) patients, cyst formation
(range 8–18 Gy). After a median follow up of 67 months, the or enlargement in 5 (9.8%), and transient tumor enlargement in
10-year PFS was 65% and complete remission rate was 17%, 3 (5.9%).22
with all treatment failures occurring within the radiation field. In a study from Spain, Barcia et al reported the treatment
Acute radiation effects (AREs) were graded as mild to moderate results of 16 patients with low-grade gliomas treated with SRS,
in 40% of patients, with most demonstrating minimal to no clin- 6 of whom had prior conventional radiation therapy.23 The
ical symptoms. Similar findings were demonstrated in a cohort mean dose given was 21.7 Gy. Complete response and partial
of 24 pediatric patients treated with GKSRS for low-grade glio- response or stabilization were observed in 50 and 31% of the
mas at the University of Virginia.21 The cohort consisted pri- tumors treated, respectively.23 Three patients with tumors
marily of 19 patients (79%) with biopsy-proven low-grade located in the brainstem died of tumor progression. Swedish
gliomas, whereas 1 patient (4%) had a WHO grade III lesion and investigators also reported favorable treatment outcomes in 19
4 patients (17%) did not undergo biopsies for presumed brain- patients (16 children) treated with SRS for pilocytic astrocyto-
stem gliomas. Lesions were treated to a median dose of 15 Gy ma.24 The median tumor volume and the median prescribed
(range 4–20 Gy), with a mean treatment tumor volume of dose were 2.2 mL and 10 Gy, respectively. Out of the 19 patients
2.4 cm3. At a median radiographic follow-up of 74 months, com- treated, the tumor shrinkage rate was 85% and 95% achieved
plete tumor responses were seen in five patients (21%). At last tumor control at a median follow-up of 4.7 years.24 Increased
follow-up, the PFS rate was 83%, with higher progression rates contrast enhancement and edema attributable to SRS were
seen in larger tumors. Increased edema after GK without tumor observed in 25% of the patients.24 ▶ Table 26.2 summarizes the
progression was seen in four patients (12.5%), although no treatment outcomes of selected series.
patients experienced radiographic radiation necrosis. In summary, despite the notion that low-grade astrocytomas
Investigators from Japan reported on another large cohort of are not ideal targets for SRS, data from various series have dem-
51 adult and pediatric patients with low-grade astrocytomas, onstrated good local control and acceptable toxicity profile.

Table 26.2 Summary of treatment outcomes of stereotactic radiosurgery for low-grade astrocytomas in children
Study N Histology Marginal dose (Gy) Setting of treatment Local control (%)

Eder et al34 12 Not specified 10–18 Primary or adjuvant 77

Hadjipanayis et al18 25 JPA 15 Primary or salvage 89a

Hadjipanayis et al19 12 (4 children) Well-circumscribed 16 Primary or salvage 67b


WHO grade II astrocy-
toma

Kida et al22 51 (adult and children) WHO grade I astrocy- 12.5 Not specified 91.7
toma WHO grade II 15.7 87.2
astrocytoma

Barcia et al23 16 Low-grade glioma 21.7 Primary or boost 81

Boëthius et al24 16 JPA 10 Adjuvant 94c

Wang et al20 21 WHO grade I and II 14.5 Primary, boost, or sal- 65


astrocytoma vage

Weintraub et al21 24 Who grade I/II astrocy- 15 Primary or salvage 83


toma (80%)

Abbreviations: JPA, juvenile pilocytic astrocytoma; WHO, World Health Organization.


aLocal control of 37 patients, including 12 adults.
bLocal control for all 12 patients.
cLocal control of 19 patients, including 3 adults.

214
Stereotactic Radiosurgery for Pediatric Brain Tumors

overall median survival time was 12 months after SRS. Other


series showed similar dismal outcomes. In a study from the
University of California, San Francisco, 11 children with high-
grade gliomas were treated with SRS. Out of the 14 tumors
treated, only 5 (36%) were controlled.29 In the study from the
University of Pittsburgh, 12 children with malignant glial
tumors (malignant astrocytoma or ependymoma) were treated
with GKSRS. Ten of the children had prior EBRT. Five of them
were alive 12 to 72 months after SRS, with three remaining dis-
ease-free.14 The remaining seven children died of their disease,
with a median survival of 6 months.
Based on the very limited data available, it is very difficult to
draw any meaningful conclusions regarding the role of SRS in
the management of high-grade gliomas in children, as the diag-
nosis portends a uniformly poor prognosis. Attempts have been
made by the Children’s Cancer Study Group (CCG) and the POG
to conduct prospective trials to test the use of SRS in recurrent
pediatric brain tumors, but they were closed because of poor
accrual. If data are extrapolated from adult literature, there
does not seem to be enough evidence to support the routine
Fig. 26.3 A 5-year-old patient with a WHO grade I (pilocytic) use of SRS as the sole treatment or as boost therapy after EBRT
astrocytoma in the posterior fossa treated with Gamma Knife in children with high-grade gliomas. Radiation Therapy Oncol-
radiosurgery to a prescribed marginal dose of 12 Gy.
ogy Group (RTOG) has conducted a phase III randomized trial
(RTOG 9305) comparing no boost and SRS boost after EBRT and
carmustine (BCNU) chemotherapy for adult patients with glio-
▶ Fig. 26.3 shows the treatment plan of a WHO grade I astrocy- blastoma multiforme, but has not resulted in any improvement
toma treated with SRS. It appears that SRS is especially useful in in survival outcomes.28 However, SRS may have a role in the
cases where there is a prior history of EBRT and complete surgi- recurrent setting when there is a history of prior EBRT, when
cal resection is not possible. Moreover SRS has shown promise the patient is not responding to chemotherapy, or when com-
in the primary setting when the tumor location precludes com- plete surgical resection is not achievable. An evidence-based
plete resection. However, for these same reasons, practitioners review by the American Society for Therapeutic Radiology and
should be cautious regarding possible toxicity given the lack of Oncology (ASTRO) arrived at the same conclusions.30
prospective data, and strict adherence to the available data on
normal tissue tolerance for SRS should be observed. Currently,
there are not enough data to support the routine use of SRS in
children with newly diagnosed low-grade astrocytoma; this is
26.6 Medulloblastomas
best tested in a clinical trial setting. Medulloblastomas constitute 15 to 20% of all childhood primary
brain tumors.9 Secondary to the high propensity for spinal axis
dissemination, craniospinal irradiation followed by a posterior
26.5 High-Grade Gliomas fossa boost with or without chemotherapy after maximal safe
High-grade gliomas constitute 15% of primary brain tumors in resection of the tumor is the standard therapy. Like other
children, and usually carry a dismal prognosis despite the malignant brain tumors, a radiation dose response has been
improvement in treatments in the past two decades.2 They demonstrated in medulloblastomas.9 Stereotactic radiosurgery
have a propensity to spread microscopically beyond the extent can potentially be used as boost therapy to escalate the radia-
of tumor demonstrated on diagnostic imaging, but the main tion dose to residual disease after EBRT. It can also be used as
pattern of failure is still within 2 cm of the original tumor. 25 In salvage therapy for children who develop recurrence after radi-
adults, prospective trials and studies have been done using ation therapy.
hyperfractionation or addition of a boost with brachytherapy. 26, There are very scarce data on the use of SRS for the treatment
27 Because of its favorable dosimetric characteristics, SRS has of children with medulloblastomas. Two series came from Har-
also been used as a boost therapy for dose escalation in adults vard University. In the earlier series, 11 children with recurrent
with high-grade gliomas,28 as well as in the recurrent setting and 3 children with newly diagnosed medulloblastomas were
after prior EBRT. Given the similar poor prognosis in children, treated with SRS to a median dose of 12 Gy as salvage treatment
SRS has been used in children with high-grade gliomas as pri- and as boost therapy, respectively.31 The three patients who
mary or adjuvant treatment, boost therapy, and salvage treat- received SRS as boost therapy were alive without evidence of
ment for recurrent disease. However, data regarding the use of recurrence at a median follow-up interval of 27 months.31 Out
SRS in these settings are very limited. In a series from Harvard of the 11 children with recurrent disease, 6 developed distant
University, 18 patients with glioblastoma multiforme or ana- failure and eventually died of their disease, but none of the 11
plastic astrocytoma received SRS.11 Out of the 18 patients patients failed locally. Their median survival time was 10
treated, only 4 of them, all of whom received SRS as boost ther- months.31 In their subsequent study with more patients
apy, survived, with progression after 50 to 119 months.11 The included and longer follow-up interval, Hodgson et al reported

215
Stereotactic Radiosurgery for Pediatric Brain Tumors

a median progression-free survival time of 11 months. The 3- Radiobiologically, meningiomas are category II targets (tumor
year local control rate was 57%.11 Again, the main pattern of composed of late-responding tissue with no intermingling nor-
failure was distant. In a smaller study by Woo et al, the reported mal brain tissue) and have a sharp demarcation from normal
local control rate of the four patients (two children) treated brain parenchyma.6 Although these characteristics lend them-
with SRS as boost therapy after craniospinal and posterior fossa selves well to SRS, the data on the use of SRS for the treatment
irradiation was 100%.32 The prescribed dose ranged from 4.5 to of children with meningiomas are very limited.
10.0 Gy. Finally, investigators from the United Kingdom Eder et al reported the treatment results of two pediatric
reported on their experience with three patients treated for patients treated with GKSRS for skull base meningiomas. Tumor
isolated intracranial relapses (one to three lesions) of medullo- shrinkage after treatment was demonstrated in both patients. 34
blastoma.33 At median prescribed doses of 15 to 25 Gy and fol- Im et al reported tumor control at 2.5 years in a 15-year-old pa-
low-up of between 2.5 and 4 years after SRS, two patients tient who underwent GKSRS after subtotal resection for a men-
demonstrated complete responses, whereas the third demon- ingioma.35 Other reports included both adult and pediatric
strated stable disease. No patients have developed out of field patients, and the results were collectively reported. In a series
failures. of 121 adult and pediatric patients treated with GKSRS, DiBiase
Based on the very limited data available, it is very difficult to et al reported a crude local control rate of 91.7%. 36
draw meaningful conclusions regarding the role of SRS in this Data from the large body of experience in adults and limited
disease. Given our knowledge that the amount of residual data in children suggest that SRS is a safe and efficacious treat-
tumor after surgical resection affects disease control, using SRS ment for meningiomas. It appears that similarly favorable out-
as boost therapy appears to be a sound approach for radiation comes in adults can be reproduced in children, although
dose escalation to residual tumor. However, at present, there is caution should be exercised in its routine practice given results
insufficient evidence to support this treatment approach as of a recent meta-analysis of pediatric meningiomas failing to
routine practice in children with residual medulloblastoma demonstrate any significant relapse-free survival benefit with
after surgical resection, and these high-risk patients should up-front radiation therapy (almost exclusively EBRT).37 The typ-
continue to be enrolled on available clinical trials. In the recur- ical prescribed dose for meningiomas in adults is 12 to 18 Gy,
rent setting, in which treatment options are usually much more and similar guidelines should apply to children. ▶ Fig. 26.4
limited, SRS can provide a noninvasive option for ablative local shows the treatment plan of a prepontine meningioma treated
therapy. Based on the limited data from the literature, SRS may with SRS.
provide reasonable local control for recurrent medulloblasto-
ma. However, it does not address the risk of distant failure,
which is addressed with chemotherapy. 26.8 Vestibular Schwannomas
Vestibular schwannomas (VSs) are typically slow-growing
26.7 Meningiomas benign tumors that usually occur in adults. In children, VSs are
usually associated with neurofibromatosis type II (NF2).9 Treat-
Meningiomas are rare in children and account for 1 to 3% of all ment options include observation, surgery, SRS, and fractio-
childhood intracranial tumors.9 The standard treatment is com- nated radiation therapy. Vestibular schwannomas are regarded
plete surgical extirpation. If a gross total resection is achieved as category II targets, and they have very sharp margins of
and the pathology confirms a WHO grade I tumor, no further demarcation from the surrounding normal tissue.6 These char-
treatment is required. However, complete resection of meningi- acteristics render them excellent targets for SRS.
omas in difficult locations, such as the skull base, is not always Stereotactic radiosurgery has emerged as one of the standard
achievable even with modern microsurgical techniques. treatments for VSs. There is a large body of experience in the
In adults, SRS has become one of the standard treatments for use of SRS in adults, but the data on its use in pediatric patients
meningiomas. There are abundant data in the literature docu- are scarce.38,39 In the earlier adult series where the prescribed
menting the efficacy and safety of SRS in the treatment of men- marginal dose was 16 to 20 Gy, there was a substantial inci-
ingiomas in adults; this is discussed elsewhere in this book. dence of hearing loss and cranial nerve deficits.39,40 These

Fig. 26.4 A 16-year-old patient with a prepontine


meningioma treated with Gamma Knife radio-
surgery to a prescribed marginal dose of 12 Gy.

216
Stereotactic Radiosurgery for Pediatric Brain Tumors

results have prompted dose reduction to minimize those risks. transformation of NF2-associated VS has always been a concern
Utilization of reduced doses of 12 to 13 Gy in modern series has after SRS,44 there is no conclusive evidence in the literature sug-
resulted in a much higher rate of preservation of serviceable gesting that the rates are higher in those with NF2 when com-
hearing and also a lower incidence of trigeminal and facial pared with the very low rate reported in sporadic VSs.41 In
nerve injury from SRS.38,39 those refractory to other treatment, systemic therapy may be
In a series from the University of Pittsburgh, 62 adult and beneficial.45
pediatric patients with 74 VSs were treated with GKSRS for VS
associated with NF2. Patients with bilateral VSs were treated
with staged SRS. The median marginal dose was 14 Gy. Thirty-
two tumors were associated with complete deafness, whereas
26.9 Craniopharyngiomas
the remaining tumors were associated with some hearing func- Craniopharyngiomas are benign brain tumors that commonly
tion (26 of them associated with serviceable hearing). The local occur in children. They are commonly characterized as being
control rates were 85, 85, and 81%, respectively, at 5, 10, and 15 purely solid, purely cystic, or mixed solid and cystic. If feasible,
years.41 The rates of preservation of serviceable hearing were maximal safe resection is the gold standard treatment. How-
73, 59, and 48% at 1, 2, and 5 years, respectively, if MRI-based ever, complete surgical resection is not always achievable, and
image-guided treatment planning and a prescribed dose of < 14 radiation therapy is often offered in the postoperative or sal-
Gy were used.41 Other studies including both adults and chil- vage settings to improve local control.2 Occasionally, primary
dren treated with SRS for VS associated with NF2 showed simi- EBRT or intralesional brachytherapy is offered.
larly favorable tumor control rates, but worse hearing Similar to other extra-axial benign tumors in childhood, cra-
preservation rates.42,43 ▶ Table 26.3 summarizes the treatment niopharyngiomas have sharp margins of demarcation from the
outcomes of selected series. surrounding structures2 and based on the description by Larson
In children, VS is usually associated with NF2, and special et al should be regarded as category II targets.6 These character-
considerations have to be taken in treatment recommenda- istics lend themselves well to SRS, allowing for a rapid falloff of
tions. Preservation of serviceable hearing is of particular impor- radiation dose beyond the target volume and potentially lead-
tance in children because hearing deficits can result in speech ing to reduced toxicity. However, the typically close proximity
and learning problems. Compared with sporadic VS, a higher of the optic apparatus and hypothalamus with the tumor may
risk of treatment-related complications is observed in NF2- not allow for the differential dose delivery to the target and at
associated VS.41 Furthermore, patients with the latter may risk tissue, thus posing a challenge in the use of SRS for
develop multiple CNS tumors, and it has been postulated that treatment.9
radiation might predispose NF2 patients who have loss of one There is a moderate amount of data from the literature on
allele of a tumor suppressor gene to late secondary malignant the use of SRS for craniopharyngiomas in adults and children.
transformation. Therefore, the goals of therapy are shifted In a study from Sweden, the treatment outcomes of 21 patients
toward improving the therapeutic ratio and minimizing treat- (11 children) with 22 craniopharyngiomas were reported. The
ment-related morbidity rather than complete tumor ablation at prescribed dose ranged from 3 to 25 Gy, and mean tumor vol-
all costs. In those with asymptomatic NF2-associated VSs, close ume was 7.8 mL. Some patients who had craniopharyngiomas
observation may also be the most reasonable option. Treatment with a cystic component received SRS for the solid component
should be pursued, however, when there is progressive hearing and intralesional brachytherapy for the cystic component. With
loss or evidence of progressive tumor growth in symptomatic a mean follow-up time of 7.5 years (range 0.5–29.0 years), 5
lesions. Stereotactic radiosurgery provides a minimally invasive, tumors shrank in size, 3 were unchanged, and 14 enlarged.46
effective method to control those tumors. Although malignant Eleven (85%) of 13 tumors that received a dose < 6 Gy to the

Table 26.3 Summary of treatment outcomes of stereotactic radiosurgery for vestibular schwannoma associated with neurofibromatosis type II in chil-
dren
Study Age range (y) No. of patients/ Dose (Gy) Tumor control (%) Hearing preserva- Complications (%)
tumors tion (%)

Kida et al42 7–71 20/20 13 (10–15) 100 33.3 CN VII: 20%

Rowe et al43 11–59 96/122 Mostly 10–16, with 79 40 CN VII: 8%


30 tumors receiv- CN V: 2%
ing 17.5–25.0

Phi et al74 13–55 30/36 8–14 66 33 CN VII: 5.6%


CN V: 2.8%

Meijer et al75 11–70 7/7 10–12 100 54 CN VII: 0%

Sharma et al76 10–56 30/54 10–15 67.3 67 CN VII: 3.1%

Mathieu et al41 11–79 60/72a 14 (11–20) 88 53 CN VII: 8%


CN V: 4%

Abbreviation: CN, cranial nerve.


aTwo patients were lost to follow-up.

217
Stereotactic Radiosurgery for Pediatric Brain Tumors

margin enlarged compared with only 3 (33%) of 9 tumors that was 77.5 and 100%, respectively. In patients with mixed tumors,
received > 6 Gy.46 Nine (82%) of 11 tumors in children ultimately the 5-year control rate for complete tumor coverage and solid
enlarged after SRS, compared with 5 (50%) of 10 in adults. Eight component coverage only was 64.3 and 51.9%, respectively. In
patients developed visual deterioration, which was attributed patients with normal visual function before SRS, two patients
to tumor enlargement in all but one case.46 Four patients devel- (8%) had worsening vision, whereas in patients with visual defi-
oped pituitary deficiencies. In a study from Taiwan, Chung et al cits before SRS, two patients (11%) and five patients (26%) had
treated 31 patients (9 children younger than 16 years) who had improvements and worsening of their visual function, respec-
craniopharyngioma with SRS. Ten tumors were purely cystic, 5 tively. All three patients with normal baseline endocrine func-
purely solid, and 16 mixed cystic and solid. Two patients had tion prior to SRS did not develop any endocrine abnormalities
EBRT. Although the tumor marginal doses ranged from 9.5 to afterwards, whereas two patients with anterior hypopituitar-
16.0 Gy (mean 12.2 Gy), the dose delivered to the optic appara- ism (13%) showed changes in their function after SRS (one
tus was limited to 8 Gy (range 7.2–12.5 Gy). Three patients improved, one worsened). The 28 patients with panhypopitui-
underwent stereotactic aspiration to decompress the cystic tarism prior to treatment remained so after treatment.
component before SRS. The mean target volume was 8.94 mL. In a report from Pakistan, 35 patients (17 children) were
The tumor response and tumor control rates were 64.6 and treated with SRS for craniopharyngiomas after one of several
87.2%, respectively, and the complete response rate was previous treatments, including 11 patients receiving microsur-
32.7%.47 Tumors that were purely cystic or solid had better local gical procedures, 12 patients receiving cyst fenestration/aspira-
control than tumors with mixed components, and smaller tion, and 2 patients receiving EBRT.52 A mean lesion volume of
tumors had better tumor control than larger ones. Three 12 cm3 (range 1–33.3 cm3) was treated to a mean prescription
patients developed cystic enlargement of their tumor that com- dose of 11.5 Gy (range 8–14 Gy). At a mean follow–up of 22
pressed the adjacent structures at 5, 8, and 17 months after months, the tumor response rate (complete or partial response)
SRS, respectively.47 They all required further intervention for and control rate after GKSRS were 77.1 and 88.5%, respectively.
decompression.47 Subsequent surgical intervention was required in several
In an analysis from the University of Virginia (Charlottesville, patients, including four patients undergoing stereotactic cyst
VA), Xu et al reviewed their experience with 37 patients (17 aspiration, two requiring ventriculoperitoneal shunting, and
children) undergoing 39 GKSRS procedures for patients with two requiring Ommaya reservoir placement. Among those pre-
craniopharyngioma.48 Although a few patients underwent SRS senting with visual impairment, seven patients (50%) had
as primary treatment (4 patients were clinically diagnosed and improvement in symptoms, whereas one patient (7%) had wor-
2 patents received biopsies only), an additional 28 patents sening acuity despite decreased tumor size.
underwent previous surgery, and 2 underwent prior cyst aspi- The largest body of experience on SRS for craniopharyngioma
ration. Only the solid portions of tumor were targeted with to date was reported by a group in Japan. Kobayashi et al
GKSRS, with a median tumor volume of 1.6 cm3 (range 0.1– reported the treatment outcomes of 107 patients (38 patients <
18.6 cm3) and prescribed to a median marginal dose of 14.5 Gy 15 years) who had craniopharyngiomas treated with GKSRS.
(range 6–25 Gy). Cystic portions of tumor received either aspi- Among those patients, 13 had prior external beam radiother-
ration or phosphorus-32 treatment. At a median follow-up of apy. Ninety-eight patients had follow-up information. The
50 months, the 5 year in-field PFS rate was 67.0%. In a multi- tumor diameter and volume were 1.88 cm and 3.5 mL, respec-
variate analysis, factors relating to longer PFS included absence tively. The prescribed dose was 11.5 Gy. With a median follow-
of visual field defects at the time of SRS, tumor volume ≤ 1.6 up of 65.5 months, the tumor control rate was 79.6%, with a
cm3, and marginal dose > 14.5 Gy. Mokry et al from Austria complete response rate of 19.4%.53 The 5- and 10-year survival
reported on 23 patients (11 children < 15 years) who had cra- rates were 94.1 and 91%, respectively.53 The progression-free
niopharyngiomas treated with SRS. Ten patients underwent survival rates were 60.8 and 53.8%, respectively.53 Among the
cyst aspiration and intracystic instillation of bleomycin prior to 91 patients with evaluable information on neurologic and
SRS. The mean dose was 10.8 Gy (range 8–15 Gy). Tumor control endocrinologic symptoms, symptomatic deterioration occurred
was 56%.49 Five patients with multicystic residual or recurrent in 16.5% of the patients.53
tumors developed tumor progression. In an analysis limited to cystic lesions, Park et al retrospec-
Another study by Amendola et al reported on the outcomes tively reviewed outcomes of 27 pediatric and adult patients
of 14 patients (12 children) who had craniopharyngiomas with craniopharyngioma treated with GKSRS, of whom 14
treated with GKSRS. The mean dose was 14 Gy, whereas the patients had undergone previous subtotal resection and 13
maximum doses to the optic apparatus and brainstem were patients undergone neuroendoscopic aspiration.54 Baseline
limited to 8 and 14 Gy, respectively. One patient had prior EBRT. characteristics showed similar preintervention tumor size
A PFS rate of 100% was achieved at follow-up times ranging (p = 0.962) and mean tumor volume at the time of SRS
from 6 to 86 months.50 In the series from the University of Pitts- (p = 0.896). At a median follow up of 32 months, a higher num-
burgh, 46 patients (15 patients < 14 years old) received GKSRS ber of patients recurred in the neuroendoscopic group when
for craniopharyngiomas, of whom 3 patients were treated in compared with the subtotal resection group (54% vs. 14%,
the primary setting and 43 patients were treated for recurrent/ p = 0.046), although several hormone levels, including growth
residual disease.51 The median marginal dose was 13.0 Gy hormone (p = 0.001), follicle-stimulating hormone (p = 0.025),
(range 9.0–20.0 Gy), the median tumor volume was 1.0 cm 3, and adrenocorticotropic hormone (p = 0.011), were reduced 1
and the median follow-up was 32 months. Tumor control varied year postoperatively in the subtotal resection group.
according to tumor type and SRS coverage. In solid and cystic ▶ Table 26.4 summarizes the treatment outcomes of selected
tumor types, the 5-year tumor control with complete coverage series.

218
Stereotactic Radiosurgery for Pediatric Brain Tumors

Table 26.4 Summary of treatment outcomes of stereotactic radiosurgery for craniopharyngioma in children
Study N Dose (Gy) Other treatment received Local control (%)

Ulfarsson et al46 21 (11 children) 3–25 Intralesional brachytherapy in < 6 Gy: 15%a 6 Gy: 67%
some patients with cystic Children: 18%
component

Chung et al47 31 (9 children < 16 y) 9.5–16.0 3 children had stereotactic 87.2


aspiration (CR: 32.7)

Mokry49 23 (11 children < 15 y) 8–15 10 patients with cystic com- 56


ponent received cystic aspira-
tion and intracystic bleomycin

Amendola et al50 14 (12 children) 14 1 patient had prior EBRT 100%

Niranjan et al51 46 with 51 tumors (15 12.5–20.0 3 patients had only biopsy; 43 Solid component: 91.6%
patients < 13 y) patients had residual/recurrent Solid and cystic: 68%
tumor after surgery; 2 patients
had prior EBRT; 11 patients
had prior cyst aspiration and
phosphorus-32 administration

Xu et al48 37 (17 children) 6–25 28 patients had prior surgery; 5 y: 67.0


3 patients had biopsies only;
2 had up-front stereotactic
cyst aspiration; and 4 patients
underwent GKSRS as primary
treatment

Saleem et al52 35 patients (17 children) 8.0–14.0 2 patients had fractionated 88.5
EBRT; 11 patients had micro-
surgical resection; 12 patients
cyst fenestration/aspiration

Eder et al34 7 children 8.0–12.0 4 patients had prior resection; 86


3 patients had cystic drainage
with intracystic bleomycin

Kobayashi et al53 107 (38 children < 15 y)a 11.5 13 had prior EBRT 79.6

Abbreviations: CR, complete response; EBRT, external beam radiation therapy; Gamma Knife stereotactic radiosurgery.
aOnly98 patients had follow-up.

Data in the literature show reasonable local control with the low if the maximum dose to the anterior visual pathway was
use of SRS for craniopharyngiomas as salvage therapy in the limited to ≤ 12 Gy in one fraction.57 Data in the literature sug-
setting of recurrent disease; however, there may also be a role gest that 12 Gy might be an adequate dose to achieve local
for SRS as primary treatment or as an adjunctive treatment to control of craniopharyngioma46,47,50,53; thus, highly conformal
surgery, either alone or in combination with intralesional bra- plans may be able to minimize the risk of optic neuropathy
chytherapy (for cystic lesions or components).46–54 The close while maximizing tumor control. For GKSRS, this can be
proximity of the optic apparatus to craniopharyngiomas in achieved using smaller shots and plugging/blank sectors. For
many cases can create technical challenges in treatment plan- CyberKnife and other linac-based systems, a dose constraint
ning, as the treatment dose may be limited in the region of should be set on the optic apparatus when using inverse plan-
tumor very close to or abutting the optic apparatus. 2 Typically, ning. Another treatment-related factor that may lead to mor-
the dose constraint for the optic apparatus is set at 8 Gy, based bidity includes cystic enlargement of craniopharyngiomas
on a seminal study by Tishler et al from Harvard University.55 after SRS. This can result in compression of the optic appara-
However, a separate analysis by Stafford et al from the Mayo tus, leading to rapid visual deterioration.47 This possibility has
Clinic showed that optic neuropathy may not be observed even to be kept in mind when a child develops visual deterioration
with a maximum point dose of 10 to 12 Gy to the optic appara- after SRS.
tus.56 The risk of optic neuropathy was 1.7% for patients receiv-
ing < 8 Gy, 1.8% for patients receiving between 8 and 10 Gy, and
6.9% for patients receiving > 12 Gy.56 This dataset suggests that
the marginal dose can be safely increased to 10 to 12 Gy even
26.10 Pituitary Adenomas
when the tumor is abutting the optic apparatus, although these Pituitary adenomas are a heterogeneous group of tumors aris-
constraints may also be volume dependent. In a recent update ing from the pituitary, and are relatively rare in children.2 As in
from Mayo Clinic with mean clinical and radiographic follow- adults, they are classified as either secretory or nonsecretory.
up times of 83 and 123 months, respectively, it was confirmed Maximal safe resection is the mainstay of treatment, especially
that the risk of radiation-induced optic neuropathy was very when the tumor is encroaching on the optic pathway. However,

219
Stereotactic Radiosurgery for Pediatric Brain Tumors

complete surgical resection may not always be achieved, and in dose to the optic apparatus is typically limited to 8 Gy, a suffi-
cases of secretory pituitary adenomas, persistent hypersecre- cient distance between the optic apparatus and the pituitary
tion may still occur despite complete surgical resection. In these tumor is needed to avoid injury to the optic apparatus.
situations, postoperative radiation therapy may be offered for In these situations, several options are available to providers.
tumor or endocrine control. Dosimetrically, different maneuvers can be used to avoid the
There is a fair amount of literature on the use of SRS for pitui- optic apparatus, such as plugging and adjustment of the gamma
tary adenomas in adults.58 However, data on the use of SRS for angle (to align with the long axis of the optic apparatus).58 If
pituitary adenoma in children are much more limited. In a the tumor is far enough (3–5 mm) from the optic pathway, SRS
study from the Karolinska Institute in Sweden, Thorén et al may be offered, either as definitive treatment or as an salvage
reported excellent outcomes in eight children with Cushing dis- treatment after subtotal resection.2 Alternatively, other modal-
ease treated with GKSRS. The observation time ranged from ities of treatment may be more appropriate. If there is concern
2.60 to 6.75 years. Among the eight patients treated, seven for tumor-related mass effect, transsphenoidal resection should
showed complete endocrine remission.59 All patients exhibited be offered because significant shrinkage of the tumor after SRS
deficiencies of growth hormones after SRS.59 In a study from is not expected. External beam radiation therapy can also be
Japan, Kobayashi et al also reported excellent results in terms of safely offered when optic pathway encroachment is an issue,
endocrine response in two children with Cushing disease given the wider therapeutic window and lower probability of
treated with GKSRS.60 Castinetti et al from France reported the complications provided by fractionation.2 However, it is not
results of 40 patients (4 patients ≤ 18 years) with Cushing dis- always preferable, as some data suggest SRS may produce a
ease treated with GKSRS after surgery. Out of the four children quicker normalization of hypersecretion from secretory pitui-
treated, two achieved normalization of their cortisol levels at tary adenomas.2,58
an average of 12 months.61 The median dose given was 25 Gy at
the 50% isodose line. Both pediatric patients who did not
achieve an endocrine cure were on ketoconazole at the time of
SRS.61 In the study from Harvard University, 18 patients (includ-
26.11 Pineal Tumors
ing adults and children) with pituitary adenomas were treated Pineal tumors represent a wide spectrum of histologies and are
with linac-based SRS. The prescribed dose ranged from 10 Gy at relatively rare. Common histologies include germ cell and pine-
the 85% isodose line to 15 Gy at the 65% isodose line. Although al parenchymal tumors.2 Primitive neuroectodermal tumor
the tumor control rate was 100%, the rate of normalization of (PNET) can also occur in the pineal gland.2 Specific treatment
hormonal activity was only 33%.62 The 3-year freedom from depends highly on histology, but is dependent on the complex
CNS adverse effects rate was 72.2%.62 In the largest published anatomy and adjacent organs at risk. The pineal gland is located
series to date, Sheehan et al reviewed the University of Virginia immediately adjacent to the tectal plate of the midbrain, where
experience of 418 pediatric and adult patients undergoing the superior colliculi are located. Larger pineal tumors can also
GKSRS for pituitary adenoma with either persistent functioning involve the interstitial nuclei of Cajal, which control downward
or radiological evidence of growth.63 Typical tumor doses gaze. Overall, the data in the literature on the use of SRS for
ranged from12 to 18 Gy for nonfunctioning adenomas, and 18 pineal tumors are limited.
to 30 Gy for functioning adenomas (overall range 9–30 Gy). At a In an early series from France, 11 patients (adult and chil-
median follow-up of 31 months, 90.7% of lesions demonstrated dren) with pineal tumors of various histologies, including pine-
radiographic control. Marginal dose significantly correlated ocytoma, tectal astrocytoma, germinoma, pineoblastoma, and
with improved control (p = 0.02), whereas cessation of antise- meningioma, were treated with SRS. The marginal doses ranged
cretory medications was not. Endocrine remission rates in from 12 to 20 Gy. None of the 11 patients received EBRT. At a
patients with acromegaly, Cushing disease, prolactinoma, and median follow-up of 12.3 months (range 2–34 months), all
Nelson syndrome were 53, 54, 26, and 20%, respectively. New tumors responded to SRS.64 No significant complications were
hormonal deficiencies were seen in 24.4% of all patients, and observed. In another series from France, 13 patients (5 children)
factors found to significantly correlate with such deficiencies were treated with GKSRS for pineal parenchymal tumors.
were suppressive medications at the time of GK (p < 0.001), pri- Among the five children treated, three had pineocytomas and
or craniotomy (p = 0.027), and larger tumor volumes (p = 0.007). two had pineoblastomas. None of them received EBRT. The cen-
Limited data in the literature on children suggest that the tral doses ranged from 24 to 40 Gy. Out of the five pediatric
treatment outcomes with SRS are similar to those in adults. 59–63 patients treated, one with pineocytoma was lost to follow-up.
Typically, the tumor control rates are uniformly excellent, Both patients with pineoblastoma had either complete res-
whereas the endocrine control rates are lower. The adverse ponse or 90% partial response, but one developed brain meta-
effect of antisecretory medications on endocrine normalization stasis at 23 months, and one died at 10 months.65 For the
is also demonstrated in children.61 One major challenge in the remaining two patients with pineocytoma, one achieved a com-
use of SRS in the treatment of pituitary adenomas is the prox- plete response at 72 months and one a partial response after
imity of the target volume to the optic pathway.9 Unlike SRS for SRS at 12 months.65
craniopharyngiomas, a much higher prescribed dose to the tar- In the study from the University of Pittsburgh, Hasegawa et al
get volume is needed in secretory pituitary adenomas for endo- reported the results of 16 patients (adults and children) with
crine cure.58 As in adults, if SRS is used in children with pineal parenchymal tumors treated with GKSRS as the primary
secretory pituitary adenomas, a dose of at least 25 to 30 Gy is or an adjuvant treatment after surgery. There were six patients
required if the dose to the optic apparatus permits. For nonse- in the pediatric age group. Three of them had pineocytoma, two
cretory tumors, a lower dose (15–20 Gy) is required. Given the had pineoblastoma, and one had mixed pineal parenchymal

220
Stereotactic Radiosurgery for Pediatric Brain Tumors

tumor. The prescribed dose ranged from 14 to 16 Gy. The two subtype, with the 5-year PFS for pineal parenchymal tumors,
patients with pineoblastoma also received craniospinal irradia- astrocytomas, ependymomas, and those without histological
tion. The local control was 100% for the 14 patients with imag- diagnosis being 54, 100, 67, and 81%, respectively. Worse out-
ing follow-up.66 Both pediatric patients with pineoblastoma comes were seen among those with higher initial tumor grades
died 18 and 47 months after SRS, whereas the other four pedia- (p = 0.04), previous radiotherapy (p = 0.002), and radiological
tric patients survived.66 Complications were not observed in evidence of necrosis (p = 0.03). Transient diplopia was noted in
the six pediatric patients. In another study from the University one patient (2.2%), which resolved within 1 year.
of Pittsburgh, four patients (aged 9, 18, 18, and 21 years) with The variety of tumor histologies and lack of good outcome
pineal nongerminomatous germ cell tumors were treated with data in both the adult and pediatric literature on the use of SRS
GKSRS. All patients received chemotherapy and fractionated for the treatment of pineal tumors renders it very difficult to
radiotherapy, and SRS was used as a boost. The marginal dose draw any meaningful conclusions regarding its role in these
for the SRS boost ranged from 12 to 16 Gy. At a mean follow-up tumors. The most important factor in determining the treat-
time of 25 months, local control was obtained in three of the ment strategy is histology, which governs the biological behav-
four patients treated.67 ior. Tumors such as germ cell tumors and pineoblastomas have
In a study from Japan, Kobayashi et al reported the outcomes a high propensity for neural axis dissemination and require
of 30 patients (both adult and pediatric) with pineal and related more than local treatment of the primary tumor alone.2 Based
tumors treated with GKSRS. Out of the 30 patients treated in on the limited literature available and the knowledge of the
this study, 19 received previous radiotherapy. There were 21 patterns of failure for germ cell tumors and pineoblastomas, it
pineal tumors, including 4 germinomas, 7 malignant germ cell is probably appropriate to consider SRS as a boost therapy in
tumors, 2 syncytiotrophoblastic giant cell tumors, 3 pineocyto- some cases. Stereotactic radiosurgery can also be used as a sal-
mas, and 2 pineoblastomas. Pineal tumors were treated to a vage therapy for recurrent germ cell tumors and pineoblasto-
mean dose of 15.7 Gy, whereas pineocytomas and pineoblasto- mas. For pineocytomas, which have a low propensity for neural
mas were treated to a mean dose of 16.6 Gy. The complete res- axis dissemination, SRS appears to be an appropriate treatment
ponse rate was 26.7%, and the overall response rate was modality for selected patients.
73.3%.68 The local control in germinomas and pineocytomas In situations where SRS may be considered, highly conformal
was 100%, whereas pineoblastomas and malignant germ cell treatment plans are desirable to minimize the risk of damage to
tumors responded poorly to SRS.68 In another series from Japan, surrounding critical structures. This may be achieved by using
three patients with pineal germinomas were treated with smaller shots if the GK is used or by using an intensity-modu-
GKSRS to a dose of 10 to 12 Gy, followed by whole ventricular lated radiosurgical system with micro-multileaf collimation if a
irradiation of 24 Gy. All patients had solitary pineal tumors linac-based system is used. There are no well-defined guide-
measuring < 3 cm. Complete response was obtained in all three lines regarding the dosing for SRS for pineal tumors, but 12 to
patients.69 16 Gy appears to be a reasonable dose range based on the lim-
Amendola et al from the Miami Neuroscience Center in Flori- ited data in the literature.
da reported their outcomes of 20 patients (14 patients in the
pediatric age group) with pineal tumors of various histologies,
including 13 germ cell tumors, 2 pineoblastomas, 2 low-grade 26.12 Complications
gliomas, 1 primitive neuroectodermal tumor, 1 teratoma, and 1
Specific complications of treatment are outlined above, as they
pineocytoma, treated with SRS. The median dose was 11 Gy
relate to each individual disease process. Long-term risks of
(range 8–20 Gy), and the mean target volume was 3.1 mL. Five
radiation-induced neoplasia are also a concern in pediatric
patients underwent chemotherapy, and four received adjuvant
patients who have many years at risk for this rare, but late-term
EBRT. Seventeen (85%) patients were alive after treatment, with
side effect.
a median follow-up of 30.4 months.70 Lekovic et al from the
Overall, SRS remains an attractive modality in the treatment
Barrow Neurological Institute (Phoenix, AZ) reported their
of pediatric intracranial tumors, with its ability to give high
experience with 17 patients (2 pediatric patients) with pineal
doses of radiation to focal areas and spare significant dose to
tumors treated with GKSRS. One of the pediatric patients had
the surrounding normal brain. Given the limited body of litera-
malignant teratoma, which was treated with a dose of 13 Gy,
ture, however, it remains unknown whether dose-volume con-
with a partial response at 15 months; the other had a nonger-
straints of normal tissues can be extrapolated from the adult
minomatous germ cell tumor, which was treated with a dose of
experience. In the absence of clear data, caution should be exer-
15 Gy, with a complete response maintained at 73 months. 71
cised in utilizing these techniques outside of the clinical trial
Both patients survived without complications.71
setting.
Yianni et al reported on outcomes of 44 consecutive pediatric
and adult patients undergoing 50 radiosurgical procedures for a
variety of pineal lesions, including 11 pineal parenchymal
tumors, 6 astrocytomas, 3 ependymomas, 2 papillary endothe-
26.13 Summary
lial tumors, 2 germ cell tumors, and 20 patients without a prior The dosimetric characteristics, namely, high conformality and
histological diagnosis.72 Lesions were treated to a marginal dose rapid dose falloff, of SRS render it an attractive treatment for
of 18.0 Gy (range 10–30 Gy), with a mean tumor volume of pediatric patients with brain tumors in various settings.
2.7 cm3. At a mean follow-up of 62.5 months, the 1-, 5-, 10-, Although treatment outcomes from prospective trials are lack-
and 20- PFS after SRS was 93, 77, 67, and 67%, respectively. ing in the literature, experience with the use of SRS for pediatric
Some variability in tumor control was noted by histological brain tumors accumulated at various major radiosurgical

221
Stereotactic Radiosurgery for Pediatric Brain Tumors

centers across the globe have provided valuable data that can [22] Kida Y, Kobayashi T, Mori Y. Gamma knife radiosurgery for low-grade astro-
cytomas: results of long-term follow up. J Neurosurg 2000; 93 (Suppl 3): 42–
guide neurosurgeons and radiation oncologists in their treat-
46
ment decisions. With the accumulation of more data and longer [23] Barcia JA, Barcia-Salorio JL, Ferrer C, Ferrer E, Algás R, Hernández G. Stereo-
follow-up, it is hoped that more information will be available tactic radiosurgery of deeply seated low grade gliomas. Acta Neurochir Suppl
with which to make even better management decisions. Pro- (Wien) 1994; 62: 58–61
spective trials testing the role of SRS in pediatric brain tumors [24] Boëthius J, Ulfarsson E, Rähn T, Lippittz B. Gamma knife radiosurgery for pilo-
cytic astrocytomas. J Neurosurg 2002; 97 (Suppl): 677–680
are desperately needed and given the rarity of these pediatric
[25] Hochberg F, Grossman SA, Mikkelsen T, Glantz M, Fisher JD, Piantadosi S,
brain tumors, those trials should be conducted in a cooperative NABTT CNS Consortium. Lack of efficacy of 9-aminocamptothecin in adults
group, multi-institutional, or consortium setting. with newly diagnosed glioblastoma multiforme and recurrent high-grade as-
trocytoma. Neuro-oncol 2000; 2: 29–33
[26] Selker RG, Shapiro WR, Burger P, et al. Brain Tumor Cooperative Group. The

References Brain Tumor Cooperative Group NIH Trial 87–01: a randomized comparison
of surgery, external radiotherapy, and carmustine versus surgery, interstitial
[1] Suh JH, Barnett GH. Stereotactic radiosurgery for brain tumors in pediatric radiotherapy boost, external radiation therapy, and carmustine. Neurosur-
patients. Technol Cancer Res Treat 2003; 2: 141–146 gery 2002; 51: 343–355, discussion 355–357
[2] Lo SS, Fakiris AJ, Abdulrahman R, et al. Role of stereotactic radiosurgery and [27] Laperriere NJ, Leung PM, McKenzie S, et al. Randomized study of brachyther-
fractionated stereotactic radiotherapy in pediatric brain tumors. Expert Rev apy in the initial management of patients with malignant astrocytoma. Int J
Neurother 2008; 8: 121–132 Radiat Oncol Biol Phys 1998; 41: 1005–1011
[3] Bauman GS, Brett CM, Ciricillo SF, et al. Anesthesia for pediatric stereotactic [28] Souhami L, Seiferheld W, Brachman D, et al. Randomized comparison of ster-
radiosurgery. Anesthesiology 1998; 89: 255–257 eotactic radiosurgery followed by conventional radiotherapy with carmustine
[4] Giller CA, Berger BD, Pistenmaa DA, et al. Robotically guided radiosurgery for to conventional radiotherapy with carmustine for patients with glioblastoma
children. Pediatr Blood Cancer 2005; 45: 304–310 multiforme: report of Radiation Therapy Oncology Group 93–05 protocol. Int
[5] Lo SS, Chang EL, Sloan AE. Role of stereotactic radiosurgery and fractionated J Radiat Oncol Biol Phys 2004; 60: 853–860
stereotactic radiotherapy in the management of intracranial ependymoma. [29] Baumann GS, Wara WM, Larson DA, et al. Gamma knife radiosurgery in chil-
Expert Rev Neurother 2006; 6: 501–507 dren. Pediatr Neurosurg 1996; 24: 193–201
[6] Larson DA, Flickinger JC, Loeffler JS. The radiobiology of radiosurgery. Int J Ra- [30] Tsao MN, Mehta MP, Whelan TJ, et al. The American Society for Therapeutic
diat Oncol Biol Phys 1993; 25: 557–561 Radiology and Oncology (ASTRO) evidence-based review of the role of radio-
[7] Pollack IF, Gerszten PC, Martinez AJ, et al. Intracranial ependymomas of child- surgery for malignant glioma. Int J Radiat Oncol Biol Phys 2005; 63: 47–55
hood: long-term outcome and prognostic factors. Neurosurgery 1995; 37: [31] Patrice SJ, Tarbell NJ, Goumnerova LC, Shrieve DC, Black PM, Loeffler JS. Re-
655–666, discussion 666–667 sults of radiosurgery in the management of recurrent and residual medullo-
[8] Healey EA, Barnes PD, Kupsky WJ, et al. The prognostic significance of post- blastoma. Pediatr Neurosurg 1995; 22: 197–203
operative residual tumor in ependymoma. Neurosurgery 1991; 28: 666–671, [32] Woo C, Stea B, Lulu B, Hamilton A, Cassady JR. The use of stereotactic radio-
discussion 671–672 surgical boost in the treatment of medulloblastomas. Int J Radiat Oncol Biol
[9] Marks JE, Adler SJ. A comparative study of ependymomas by site of origin. Int Phys 1997; 37: 761–764
J Radiat Oncol Biol Phys 1982; 8: 37–43 [33] King D, Connolly D, Zaki H, Lee V, Yeomanson D. Successful treatment of
[10] Aggarwal R, Yeung D, Kumar P, Muhlbauer M, Kun LE. Efficacy and feasibility metastatic relapse of medulloblastoma in childhood with single session ster-
of stereotactic radiosurgery in the primary management of unfavorable pe- eotactic radiosurgery: a report of 3 cases. J Pediatr Hematol Oncol
diatric ependymoma. Radiother Oncol 1997; 43: 269–273 [34] Eder HG, Leber KA, Eustacchio S, Pendl G. The role of gamma knife radiosur-
[11] Hodgson DC, Goumnerova LC, Loeffler JS, et al. Radiosurgery in the manage- gery in children. Childs Nerv Syst 2001; 17: 341–346, discussion 347
ment of pediatric brain tumors. Int J Radiat Oncol Biol Phys 2001; 50: 929– [35] Im SH, Wang KC, Kim SK, et al. Childhood meningioma: unusual location,
935 atypical radiological findings, and favorable treatment outcome. Childs Nerv
[12] Mansur DB, Drzymala RE, Rich KM, Klein EE, Simpson JR. The efficacy of ster- Syst 2001; 17: 656–662
eotactic radiosurgery in the management of intracranial ependymoma. J [36] DiBiase SJ, Kwok Y, Yovino S, et al. Factors predicting local tumor control after
Neurooncol 2004; 66: 187–190 gamma knife stereotactic radiosurgery for benign intracranial meningiomas.
[13] Lo SS, Abdulrahman R, Desrosiers PM, et al. The role of Gamma Knife Radio- Int J Radiat Oncol Biol Phys 2004; 60: 1515–1519
surgery in the management of unresectable gross disease or gross residual [37] Kotecha RS, Pascoe EM, Rushing EJ, et al. Meningiomas in children and ado-
disease after surgery in ependymoma. J Neurooncol 2006; 79: 51–56 lescents: a meta-analysis of individual patient data. Lancet Oncol 2011; 12:
[14] Grabb PA, Lunsford LD, Albright AL, Kondziolka D, Flickinger JC. Stereotactic 1229–1239
radiosurgery for glial neoplasms of childhood. Neurosurgery 1996; 38: 696– [38] Flickinger JC, Kondziolka D, Niranjan A, Maitz A, Voynov G, Lunsford LD.
701, discussion 701–702 Acoustic neuroma radiosurgery with marginal tumor doses of 12 to 13 Gy. Int
[15] Kano H, Yang HC, Kondziolka D, et al. Stereotactic radiosurgery for pediatric J Radiat Oncol Biol Phys 2004; 60: 225–230
recurrent intracranial ependymomas. J Neurosurg Pediatr 2010; 6: 417–423 [39] Lunsford LD, Niranjan A, Flickinger JC, Maitz A, Kondziolka D. Radiosurgery of
[16] Stauder MC, Ni Laack N, Ahmed KA, Link MJ, Schomberg PJ, Pollock BE. Ster- vestibular schwannomas: summary of experience in 829 cases. J Neurosurg
eotactic radiosurgery for patients with recurrent intracranial ependymomas. 2005; 102 (Suppl): 195–199
J Neurooncol 2012; 108: 507–512 [40] Kondziolka D, Lunsford LD, McLaughlin MR, Flickinger JC. Long-term out-
[17] Ostrom QT, Gittleman H, Farah P, et al. CBTRUS statistical report: primary comes after radiosurgery for acoustic neuromas. N Engl J Med 1998; 339:
brain and central nervous system tumors diagnosed in the United States in 1426–1433
2006–2010. Neuro-oncol 2013; 15 (Suppl 2): ii1–ii56 [41] Mathieu D, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for
[18] Hadjipanayis CG, Kondziolka D, Gardner P, et al. Stereotactic radiosurgery for vestibular schwannomas in patients with neurofibromatosis type 2: an anal-
pilocytic astrocytomas when multimodal therapy is necessary. J Neurosurg ysis of tumor control, complications, and hearing preservation rates. Neuro-
2002; 97: 56–64 surgery 2007; 60: 460–468, discussion 468–470
[19] Hadjipanayis CG, Niranjan A, Tyler-Kabara E, Kondziolka D, Flickinger JC, [42] Kida Y, Kobayashi T, Tanaka T, Mori Y. Radiosurgery for bilateral neurinomas
Lunsford LD. Stereotactic radiosurgery for well-circumscribed fibrillary grade associated with neurofibromatosis type 2. Surg Neurol 2000; 53: 383–389,
II astrocytomas: an initial experience. Stereotact Funct Neurosurg 2002; 79: discussion 389–390
13–24 [43] Rowe JG, Radatz MW, Walton L, Soanes T, Rodgers J, Kemeny AA. Clinical ex-
[20] Wang LW, Shiau CY, Chung WY, et al. Gamma Knife surgery for low-grade as- perience with gamma knife stereotactic radiosurgery in the management of
trocytomas: evaluation of long-term outcome based on a 10-year experience. vestibular schwannomas secondary to type 2 neurofibromatosis. J Neurol
J Neurosurg 2006; 105 (Suppl): 127–132 Neurosurg Psychiatry 2003; 74: 1288–1293
[21] Weintraub D, Yen CP, Xu Z, Savage J, Williams B, Sheehan J. Gamma knife sur- [44] Plowman PN, Evans DG. Stereotactic radiosurgery XI. Acoustic neuroma ther-
gery of pediatric gliomas. J Neurosurg Pediatr 2012; 10: 471–477 apy and radiation oncogenesis. Br J Neurosurg 2000; 14: 93–95

222
Stereotactic Radiosurgery for Pediatric Brain Tumors

[45] Plotkin SR, Stemmer-Rachamimov AO, Barker FG II, et al. Hearing improve- [61] Castinetti F, Nagai M, Dufour H, et al. Gamma knife radiosurgery is a success-
ment after bevacizumab in patients with neurofibromatosis type 2. N Engl J ful adjunctive treatment in Cushing’s disease. Eur J Endocrinol 2007; 156:
Med 2009; 361: 358–367 91–98
[46] Ulfarsson E, Lindquist C, Roberts M, et al. Gamma knife radiosurgery for cra- [62] Mitsumori M, Shrieve DC, Alexander E III, et al. Initial clinical results of LI-
niopharyngiomas: long-term results in the first Swedish patients. J Neuro- NAC-based stereotactic radiosurgery and stereotactic radiotherapy for pitui-
surg 2002; 97 (Suppl): 613–622 tary adenomas. Int J Radiat Oncol Biol Phys 1998; 42: 573–580
[47] Chung WY, Pan DH, Shiau CY, Guo WY, Wang LW. Gamma knife radiosurgery [63] Sheehan JP, Pouratian N, Steiner L, Laws ER, Vance ML. Gamma Knife surgery
for craniopharyngiomas. J Neurosurg 2000; 93 (Suppl 3): 47–56 for pituitary adenomas: factors related to radiological and endocrine out-
[48] Xu Z, Yen CP, Schlesinger D, Sheehan J. Outcomes of Gamma Knife surgery for comes. J Neurosurg 2011; 114: 303–309
craniopharyngiomas. J Neurooncol 2011; 104: 305–313 [64] Manera L, Régis J, Chinot O, et al. Pineal region tumors: the role of stereotactic
[49] Mokry M. Craniopharyngiomas: a six year experience with Gamma Knife ra- radiosurgery. Stereotact Funct Neurosurg 1996; 66 (Suppl 1): 164–173
diosurgery. Stereotact Funct Neurosurg 1999; 72 (Suppl 1): 140–149 [65] Reyns N, Hayashi M, Chinot O, et al. The role of Gamma Knife radiosurgery in
[50] Amendola BE, Wolf A, Coy SR, Amendola MA. Role of radiosurgery in cranio- the treatment of pineal parenchymal tumours. Acta Neurochir (Wien) 2006;
pharyngiomas: a preliminary report. Med Pediatr Oncol 2003; 41: 123–127 148: 5–11, discussion 11
[51] Niranjan A, Kano H, Mathieu D, Kondziolka D, Flickinger JC, Lunsford LD. Ra- [66] Hasegawa T, Kondziolka D, Hadjipanayis CG, Flickinger JC, Lunsford LD. The
diosurgery for craniopharyngioma. Int J Radiat Oncol Biol Phys 2010; 78: 64– role of radiosurgery for the treatment of pineal parenchymal tumors. Neuro-
71 surgery 2002; 51: 880–889
[52] Saleem MA, Hashim AS, Rashid A, Ali M. Role of gamma knife radiosurgery in [67] Hasegawa T, Kondziolka D, Hadjipanayis CG, Flickinger JC, Lunsford LD. Ster-
multimodality management of craniopharyngioma. Acta Neurochir Suppl eotactic radiosurgery for CNS nongerminomatous germ cell tumors. Report of
(Wien) 2013; 116: 55–60 four cases. Pediatr Neurosurg 2003; 38: 329–333
[53] Kobayashi T, Kida Y, Mori Y, Hasegawa T. Long-term results of gamma knife [68] Kobayashi T, Kida Y, Mori Y. Stereotactic gamma radiosurgery for pineal and
surgery for the treatment of craniopharyngioma in 98 consecutive cases. J related tumors. J Neurooncol 2001; 54: 301–309
Neurosurg 2005; 103 (Suppl): 482–488 [69] Endo H, Kumabe T, Jokura H, Tominaga T. Stereotactic radiosurgery followed
[54] Park YS, Chang JH, Park YG, Kim DS. Recurrence rates after neuroendoscopic by whole ventricular irradiation for primary intracranial germinoma of the
fenestration and Gamma Knife surgery in comparison with subtotal resection pineal region. Minim Invasive Neurosurg 2005; 48: 186–190
and Gamma Knife surgery for the treatment of cystic craniopharyngiomas. J [70] Amendola BE, Wolf A, Coy SR, Amendola MA, Eber D. Pineal tumors: analysis
Neurosurg 2011; 114: 1360–1368 of treatment results in 20 patients. J Neurosurg 2005; 102 (Suppl): 175–179
[55] Tishler RB, Loeffler JS, Lunsford LD, et al. Tolerance of cranial nerves of the [71] Lekovic GP, Gonzalez LF, Shetter AG, et al. Role of Gamma Knife surgery in the
cavernous sinus to radiosurgery. Int J Radiat Oncol Biol Phys 1993; 27: 215– management of pineal region tumors. Neurosurg Focus 2007; 23: E12
221 [72] Yianni J, Rowe J, Khandanpour N, et al. Stereotactic radiosurgery for pineal
[56] Stafford SL, Pollock BE, Leavitt JA, et al. A study on the radiation tolerance of tumours. Br J Neurosurg 2012; 26: 361–366
the optic nerves and chiasm after stereotactic radiosurgery. Int J Radiat Oncol [73] Stafford SL, Pollock BE, Foote RL, Gorman DA, Nelson DF, Schomberg PJ. Ster-
Biol Phys 2003; 55: 1177–1181 eotactic radiosurgery for recurrent ependymoma. Cancer 2000; 88: 870–875
[57] Leavitt JA, Stafford SL, Link MJ, Pollock BE. Long-term evaluation of radiation- [74] Phi JH, Kim DG, Chung HT, Lee J, Paek SH, Jung HW. Radiosurgical treatment
induced optic neuropathy after single-fraction stereotactic radiosurgery. Int J of vestibular schwannomas in patients with neurofibromatosis type 2: tumor
Radiat Oncol Biol Phys 2013; 87: 524–527 control and hearing preservation. Cancer 2009; 115: 390–398
[58] Witt TC. Stereotactic radiosurgery for pituitary tumors. Neurosurg Focus [75] Meijer OW, Vandertop WP, Lagerwaard FJ, Slotman BJ. Linear accelerator-
2003; 14: E10 based stereotactic radiosurgery for bilateral vestibular schwannomas in pa-
[59] Thorén M, Rähn T, Hallengren B, et al. Treatment of Cushing’s disease in child- tients with neurofibromatosis type 2. Neurosurgery 2008; 62 (Suppl): A37–
hood and adolescence by stereotactic pituitary irradiation. Acta Paediatr A42, discussion A42–A43
Scand 1986; 75: 388–395 [76] Sharma MS, Singh R, Kale SS, Agrawal D, Sharma BS, Mahapatra AK. Tumor
[60] Kobayashi T, Kida Y, Mori Y. Gamma knife radiosurgery in the treatment of control and hearing preservation after Gamma Knife radiosurgery for vestib-
Cushing disease: long-term results. J Neurosurg 2002; 97 (Suppl): 422–428 ular schwannomas in neurofibromatosis type 2. J Neurooncol 2010; 98: 265–
270

223
27 Radiosurgery for Glial Tumors 226
Part VII
28 Stereotactic Radiosurgery for the
Malignant Tumor Indications Management of One to Four Brain
Metastases 235

29 Stereotactic Radiosurgery for the


Management of Five or More Brain
Metastases 243

30 Repeat Radiosurgery for Brain


Metastases 251

VII
Radiosurgery for Glial Tumors

27 Radiosurgery for Glial Tumors


Jason Lee Schroeder, John H. Suh, Michael A. Vogelbaum, and Gene H. Barnett

11,12,13Although patient survival appeared to improve in the


Key Points earlier phase II trials, no survival advantage was borne out in
two subsequent North American phase III trials of brachyther-
● Glial tumors remain a vexing problem with relatively limited apy boosts, suggesting that much of the apparent benefit seen
advances in treatment that have translated into increased in the phase II trials was due to selection bias that chose more
survival over the past several decades. favorable patients in the brachytherapy groups compared with
● RTOG 93–05, the only prospective, randomized trial investi- the control groups.10,12,14
gating the addition of radiosurgery to conventional therapy As the evaluation and debate regarding the use of brachy-
reported no survival advantage from adding an up-front ra- therapy boosts initially for recurrent disease and then subse-
diosurgical boost to conventional therapy for the initial treat- quently for up-front treatment of malignant gliomas has run its
ment of glioblastoma. course, a new era opened to the parallel debate regarding radio-
● Despite the results of RTOG 93–05, a variety of investigators surgical boosts for the same groups of patients. Data initially
have continued to explore and report on the potential role of collected in retrospective studies on patients with recurrent
stereotactic radiosurgery in treating glial tumors—particu- gliomas were used to help support interest in using radiosur-
larly for specific subsets of patients or when other treatment gery in the up-front setting for malignant glioma. Over approx-
options have been exhausted. imately two decades, a variety of level III to level I evidence has
been generated (retrospective and prospective studies of treat-
ment for both recurrent and newly diagnosed disease) that
27.1 Introduction authors have used to support or refute the advancement of
using radiosurgery in the treatment of malignant glioma. How-
Although the physical properties of metastases are often con- ever, over that time only one prospective randomized trial eval-
sidered ideal for treatment with stereotactic radiosurgery (SRS) uating the use of radiosurgery boost in the up-front setting for
—typically small size at presentation, relatively spherical shape, malignant glioma has been completed, and as for brachyther-
and histological and radiographic delineation from normal sur- apy, the reported outcome does not show a survival benefit for
rounding brain tissue,1,2,3,4 the properties of gliomas are nearly the addition of a targeted boost to conventional radiation ther-
the complete opposite4 and often may not be considered to apy and chemotherapy.15 Critics and defenders of this study
appropriately fit within the constraints of common criteria for abound and variations on methodology have given rise to new
treatment with stereotactic radiosurgery: well-circumscribed reports of SRS efficacy for newly diagnosed as well as recur-
lesions, < 4 cm in size, without subependymal spread, and not rent/progressive malignant glioma, albeit at lower levels of evi-
adjacent to the optic chiasm or brainstem.5 Depending on the dence. As a result, the relative degree of excitement for or
histological subtype and grade of glial tumor, treatment meth- disappointment in the potential role of radiosurgery in the
ods traditionally consist of surgical resection or debulking, radi- treatment of malignant gliomas has swayed depending on the
ation therapy, and/or chemotherapy. specific articles used to support or refute one’s individual posi-
The Brain Tumor Study Group (BTSG) scientifically estab- tion. In this chapter, we review the potential role of radiosur-
lished the role of radiotherapy for high-grade gliomas; BTSG gery in both low-grade and high-grade glioma treatment.
6901 was a randomized trial that showed superior survival for
malignant glioma of 14 weeks with supportive care alone ver-
sus 36 weeks with radiotherapy.6 The same group later showed 27.2 Treatment Options
the superiority of radiotherapy over chemotherapy in the Brain
Tumor Cooperative Group (BTCG) 7201 study where survival 27.2.1 Pilocytic Astrocytoma and Optic
after radiation (with or without chemotherapy) was superior to Nerve Tumors
chemotherapy alone.7 In fact, with few exceptions (molecular
profiling particularly for anaplastic oligodendroglioma 8 and a Pilocytic astrocytoma (PA) is a tumor of childhood and adoles-
paradigm change in the primary chemotherapeutic agent for cence that often has the capacity for surgical cure, assuming the
glioblastoma9 [GBM]), the prognosis for treatment of gliomas anatomical location is favorable. Unfortunately, these tumors
has not markedly improved over the past several decades.6,7,8,9 also occur in surgically untreatable areas (optic nerves and
With limited improvement in the prognosis for gliomas, chiasm) or in regions where complete resection is not feasible.
interest emerged in potentially utilizing “more” radiation—in Pilocytic astrocytomas can also behave more aggressively and
the form of radiosurgery or fractionated stereotactic radiother- may defy surgical cure due to early recurrence or metastasis
apy—in an effort to improve outcomes for these patients. within the central nervous system (CNS). Consequently, radio-
Enticed by results of focal reirradiation for recurrent disease surgery may be an appealing option for at least some of these
and concerns about higher dosage of conventional radiotherapy tumors in an effort to provide durable local control while also
on normal brain toxicity, more regional approaches utilizing trying to minimize long-term morbidity from repeat attempts
radiation “boosts” have been applied to newly diagnosed at surgical resection or potential radiation toxicity in children.
tumors. Initially, these were delivered via brachytherapy There remain relatively few focused reports on the successful
implants using temporary or permanent radioactive sources.10, use of radiosurgery to treat these lesions.

226
Radiosurgery for Glial Tumors

Proust et al reported a case of a deep hemispheric PA that report complete response with near disappearance of the
was treated with 21 Gy to the 70% isodose line (IDL) using sin- tumors at last radiographic follow-up. Neither patient was
gle-session radiosurgery.16 Subsequent enlargement of the reported to develop any endocrine dysfunction from treatment,
tumor cyst regressed after applying intracavitary treatment but vision outcome was not reported.
with rhenium. The reported outcome at 3-years posttreatment Newer, larger series have also been reported and include
was tumor stability in an asymptomatic patient. Around the some limited to pediatric pilocytic astrocytoma,20 whereas
same time, Somaza et al reported their experience treating nine others include multiple low-grade histologies21 or both pedia-
pediatric patients with growing, deep-seated pilocytic astrocy- tric and adult patients with PA.22 Kano et al reported on out-
tomas.17 This group of patients represented diverse presenta- comes in 50 pediatric patients with pilocytic astrocytoma
tions, including two patients with new diagnosis via biopsy treated at a single institution.20 Patient presentations were var-
versus prior incomplete resections (seven patients) and failed ied, including prior failures of fractionated radiation therapy
fractionated radiotherapy (two patients). After radiosurgery, all with or without chemotherapy. At a median follow-up of 55.5
tumors had growth arrest and five showed definite regression. months, the progression-free survival (PFS) for the entire
There were no treatment toxicities reported and long-term cohort ranged between 91.7% (at 1 year) and 70.8% (at 5 years).
results (favorable or unfavorable) were not addressed. Weintraub et al reported a histologically mixed series (with
Lim and Leem reported two cases, one pediatric (PA) and one variable presentations, i.e., unresectable as well as residual or
adult (fibrillary astrocytoma), of optic pathway gliomas treated recurrent tumors) that included 15 pilocytic astrocytomas. 21
with single-fraction radiosurgery (12 Gy to the 40% IDL and The authors concluded that radiosurgery can provide good clin-
14.4 Gy to the 40% IDL, respectively).18 Follow-up imaging for ical control of residual or recurrent pediatric gliomas, while
each case showed a decrease in tumor size and both patients cautioning that worse outcomes were associated with larger
had improvement of vision. ▶ Fig. 27.1 shows a similar case tumor volumes at the time of treatment. The most recent of
treated by our team of a 41-month-old boy with biopsy-proven these series, reported by Hallemeier et al, consists of recurrent
pilocytic astrocytoma of the optic nerve and chiasm. Radiosur- or unresectable PAs in both children and adults.22 Compared
gery was completed (12 Gy to the 48% IDL) and subsequent with the Kano series, the median age and median target volume
imaging showed initial tumor shrinkage. The patient did not in this series are both higher. Although the median treatment
experience any improvement in his preoperative blindness and dose was similar in these two series (14.5 Gy in Kano et al vs.
ultimately experienced leptomeningeal dissemination (at 4.5 15 Gy in Hallemeier et al), the reported PFS in the Hallemeier
years posttreatment) and expired 7.5 years after treatment. series is lower at 65% (1 year) and 17% (5 year), possibly reflect-
More recently, Liang et al reported on two cases of optic path- ing important differences in the patient populations and/or
way glioma treated with single-session radiosurgery.19 Both tumor characteristics between the two studies.
tumors were confirmed to be PA, one of the optic chiasm Stereotactic radiosurgery for gliomas within the optic appa-
(treated with 11 Gy to the 50% IDL) and the other of the right ratus risks worsening of vision from the treatment itself, partic-
optic nerve (treated with 15 Gy to the 50% IDL). The authors ularly when the optic nerve receives more than 8 Gy in a single

Fig. 27.1 (a) A 41-month-old boy with biopsy-proven pilocytic astrocytoma of the optic nerve and chiasm. The tumor was treated with stereotactic
radiosurgery using 12 Gy to the 48% isodose line. (b) At 8-month posttreatment, there was reduction in the tumor size.

227
Radiosurgery for Glial Tumors

session. Trying to reduce this risk, Debus et al used fractionated treated with conventional radiotherapy. Three cases were
stereotactic radiotherapy (FSRT) delivering 1.8 Gy daily for a brainstem gliomas, none of which responded to the treatment
total of 54 Gy to treat 10 patients with optic nerve gliomas. 23 and the three patients succumbed to their disease.
Nine of the 10 tumors showed response to treatment, with In contrast, Kihlström et al reported on successful radiosurgi-
three complete remissions associated with vision improvement. cal treatment for low-grade glial tumors within the tectum. 30
This series was later updated by Combs et al and reported PFS Of seven total cases, six tumors responded to treatment by
of 92% (at 3 years) and 72% (at 5 years).24 showing either regression or growth arrest. Doses administered
Taken together, these reports suggest that radiosurgery or ranged from 14 to 35 Gy. The authors concluded that treatment
multifraction SRS may be an effective treatment for inoperable was safe and effective if the treatment dose did not exceed
or recurrent PAs, even when those tumors are situated in func- 14 Gy (due to development of radiation necrosis, edema, and
tionally critical areas. Surgical resection probably remains the permanent deficits in the two cases that used doses between
treatment of choice for accessible lesions. 30 and 35 Gy).
Park et al reported on a series of 25 patients treated for either
newly diagnosed or progressive low-grade astrocytomas.31 Six-
27.2.2 Low-Grade Gliomas teen of these patients were treated for newly diagnosed tumors,
Fibrillary low-grade astrocytomas tend to be genetically unsta- and those treated for progression had been previously treated
ble tumors that typically evolve into more malignant forms over with surgery, radiation therapy, or a combination of both. Their
time. Extent of resection has been cited as a favorable survival reported PFS for the entire cohort was 91.3% at 1 year, but sub-
factor for these lesions; however, this assertion remains contro- sequently decreased to 54.1% at 5 years and 37.1% at 10 years.
versial as tumor cells likely exist beyond the radiographic boun- Factors associated with better response were smaller tumor tar-
daries of these lesions. Despite significant improvements in the get volumes, higher margin treating dose, and absence of con-
results of surgery due to surgical navigation,25 awake cranioto- trast enhancement on the pretreatment imaging studies.
my,26 and resection control through the use of intraoperative A few cases of low-grade, nonenhancing tumors developing
magnetic resonance imaging (iMRI),27 complete resection of contrast enhancement several months after SRS treatment have
these tumors may be limited by poor surgical access or by loca- been seen at our institution. ▶ Fig. 27.2 provides an illustrative
tion of the lesions within a functionally critical area of the case in which a 34-year-old woman was diagnosed with World
brain. Some of these inoperable lesions may lend themselves to Health Organization (WHO) grade II astrocytoma of the right
radiosurgical treatment. thalamus by stereotactic biopsy. The 6-mm tumor was treated
Plowman reported radiographic control of two discrete and with radiosurgery using 18 Gy to the 77% IDL. At 6-month post-
spherical low-grade gliomas treated with radiosurgery at 2- treatment, the patient developed headaches and her follow-up
and 5-year posttreatment.28 The tumor observed over a longer imaging showed avid enhancement of the lesion with peritu-
period of time showed slow regression during the later stages moral edema. A subsequent fluorodeoxyglucose (FDG) positron
of imaging follow-up. Barcia et al presented 16 cases of deeply emission tomography (PET) scan documented lesion hypome-
seated low-grade glioma treated with radiosurgery.29 The mean tabolism despite the development of contrast enhancement.
dose delivered was 22 Gy; 13 of 16 lesions showed regression, 8 Ultimately, the patient required a cerebrospinal fluid (CSF)
with complete response. Six of the 16 cases had previously been diversion procedure to treat hydrocephalus. The patient’s

Fig. 27.2 (a) A 34-year-old woman with biopsy-proven WHO grade II astrocytoma of the right thalamus. The 6-mm lesion was treated with SRS using
18 Gy to the 77% isodose line. (b) At 6-month posttreatment, the tumor developed postcontrast enhancement on magnetic resonance imaging and
the patient required steroids to treat peritumoral edema. (c) At 4-year poststereotactic radiosurgery the tumor remained stable in size, but continued
to show contrast enhancement.

228
Radiosurgery for Glial Tumors

follow-up imaging continued to show enhancement for years; In 1995, Hall et al reported on the results of SRS in 35
her overall survival (OS) was 12 years from the time of patients with malignant glioma—75% GBM—that had failed con-
radiosurgery. ventional treatment.37 The actuarial survival was 8 months
from radiosurgery, with the vast majority of patients (85%)
dying from local failure. Nearly a third of these patients
27.2.3 High-Grade Gliomas required reoperation after radiosurgery, with the rate of
High-grade astrocytomas (WHO grades III and IV) constitute radionecrosis found to be 14%. The authors concluded that
the most common primary intra-axial brain tumors in adults. SRS was a useful treatment, with lower morbidity than bra-
As described in the introduction, the value of radiotherapy as a chytherapy, for recurrent malignant glioma. Similarly, median
component of management for these lesions was originally OS was 9 months after SRS in a group of 62 patients with
established in the late 1970s. Further efforts to enhance tumor recurrent GBM treated by van Kampen et al, with half the
control and to minimize radiation-induced toxicity by deliver- patients showing improved quality of life (neurologic find-
ing a boost to the tumor and its immediate surroundings rest ings, frequency of seizure, and steroid dose) at 4.5 months
on the observation that tumor recurrence or failure most com- after radiosurgery.38 Our institution reported a median PFS of
monly occurs locally—with greater than 80% of tumors recur- 4.7 months in 22 patients with recurrent GBMs (median vol-
ring within 2 cm of the enhancing edge of the primary lesion ume of 9.9 cm3) when treated with 15 Gy delivered to the
and half recurring within 1 cm.32,33 Consequently, whole-brain (median) 60% IDL.39
radiotherapy does not provide superior survival or local control Larson et al reported a mixed series of 189 patients harboring
over partial field treatment of these lesions. primary and recurrent gliomas, including tumors of all histolog-
Foundational studies in radiotherapy have shown that, up to ical grades and found that survival correlated strongly with five
a point, higher doses of radiation provide an increase in survival patient or tumor factors: lower pathological grade, younger age,
compared with lower doses.34,35 As a result, standard radiother- better performance status, smaller tumor volume, and unifocal
apy of these tumors, as defined by the Radiation Therapy Oncol- tumor.40 Chamberlain et al reported another series of 20
ogy Group (RTOG), uses the “shrinking field” method of patients with mixed glioma histologies, although all were
treating the region of edema (defined on T2-weighted MRI) treated with radiosurgery at the time of recurrence.41 Despite
plus 2 cm with 4600 centigray (cGy) in 23 fractions of 200 cGy using a median treatment dose of 17 Gy, seven patients devel-
each, and a cone-down field, determined by the region of en- oped early radiation complications (one of whom died) and an
hancement (or resection bed) on postcontrast T1-weighted MRI eighth patient developed a late radiation complication. The
plus 2 cm, treated with an additional 1400 cGy in 7 fractions of authors rated the efficacy of this treatment strategy as “mod-
200 cGy each. est” and the toxicity as “acceptable.” In an effort to define
Building on these practices, the notion that even higher acceptable doses for radiosurgical treatments, the RTOG con-
“boosts” of radiation limited to the tumor and its immediate ducted a phase I/II dose-escalation trial of SRS (RTOG 90–05) in
surroundings evolved. The development of image-guided patients with recurrent malignant gliomas and metastases. 4
stereotactic techniques allowed for precise targeting of struc- Maximum treatment doses were derived based on acceptable
tures that could be visualized on neuroimaging studies, levels of toxicity (▶ Table 27.1). Higher toxicity was found in
including computed tomography (CT), MRI, and PET. The maximum tumor diameters ≥ 21 mm and those treated with lin-
premise then being that adding focal stereotactic radiation ear accelerators (linacs) compared with the Gamma Knife (GK).
treatment—via brachytherapy (temporary or permanent) 10–14 Additionally, SRS treatment using a linac versus GK had 2.84
or radiosurgery—would allow for escalation of radiation dose times greater risk for local progression. This has not been corro-
to the target while maintaining a sharp dose falloff that borated by other studies.42,43
would spare the adjacent normal brain tissue. Sheehan Radiosurgery for recurrent malignant glioma has been com-
astutely points out that reasonable arguments can be made pared with both alternative therapies and additional therapies,
both for and against using radiosurgery in the treatment of but never in a randomized trial. Shrieve et al retrospectively
high-grade gliomas.36 On one hand, the majority of treatment compared the efficacy and toxicity of radiosurgery with brachy-
failures from standard therapy occur within 2 cm of the origi- therapy for treatment of recurrent malignant gliomas. 44
nal lesion,32,33 supporting the potential role of additional Although survival after SRS was nearly as good as for iodine-
“local” therapy; however, on the other hand, malignant glio- 125 (I-125) brachytherapy, the rate of reoperation was substan-
mas are highly infiltrative lesions with tumor cells beyond tially less. In 2002, Larson et al reported results from a phase II
the apparent radiographic lesion borders, making it appear
illogical to expect lasting success from the use of a precise,
focal treatment.36 Table 27.1 Maximum radiosurgical treatment dose for acceptable toxic-
ity in recurrent malignant tumors

Radiosurgery for Recurrent Malignant Glioma Maximum dimension ≤ 20 mm 21–30 mm 31–40 mm

A number of prospective and retrospective series have been Treatment dose 2400 cGy 1800 cGy 1500 cGy
reported regarding the potential efficacy of radiosurgery in the Source: Results of RTOG 90–05. (Reprinted from Shaw E, Scott C,
setting of recurrent glioma. However, to date there are no Souhami L, et al. Single dose radiosurgical treatment of recurrent
randomized trials comparing radiosurgery in this group of previously irradiated primary brain tumors and brain metastases: final
patients with alternative therapies, including surgery, chemo- report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys.
therapy, reirradiation (fractionated), or best supportive care.5 2000;47:291–298 with permission from Elsevier.)

229
Radiosurgery for Glial Tumors

study of radiosurgery with marimastat in the treatment of failure from SRS is the norm. Currently, it remains unclear if
recurrent malignant glioma.45 The reported PFS after treatment SRS is the best treatment for particular patient subgroups with
was 15 weeks for GBM with a corresponding median OS of 38 recurrent malignant glioma, particularly at doses that have lim-
weeks, which was not improved compared with historical con- ited toxicity. Nevertheless, SRS should be considered a reason-
trols. Skeie et al reported a retrospective series of GBM patients able option in selected cases of focal recurrence, particularly if
treated for disease recurrence with either GKSRS or reopera- other treatment options have been exhausted.
tion.46 The authors reported longer median survival after
retreatment in patients who received SRS compared with re-
Radiosurgery for Newly Diagnosed Malignant
operation only and a lower rate of complications associated
with SRS compared with reoperation. Skeie et al conclude that
Glioma
SRS may be an appropriate alternative to reoperation for In general, treatments that show some benefit at tumor recur-
patients with small-volume GBM recurrence, while also rence are thought to potentially provide even greater benefit
acknowledging the potential weaknesses of their retrospective when delivered as part of initial treatment. Because of this,
study—possible selection bias and nonuniform treatment radiosurgery as a part of up-front treatment has been used at
modalities among the treatment groups over time. multiple centers for some time. The publication of results from
Fractionation of a stereotactically guided radiotherapy treat- RTOG 93–05 in 2004, the only randomized prospective trial
ment may reduce normal tissue toxicity. Therefore, Cho et al comparing up-front SRS followed by conventional therapy
compared the results of single-session radiosurgery (17 Gy to (radiotherapy and carmustine) to conventional therapy alone
the 50% IDL, median) with FSRT (37.5 Gy in 15 fractions to the for patients with GBM, put a damper on this practice at many
85% IDL, median) in a group of 71 patients with recurrent institutions. RTOG 93–05 reported no advantage with radiosur-
malignant glioma.47 Despite prognostic factors (age, perform- gery boost in treating newly diagnosed GBM (< 4 cm in diame-
ance status, tumor size) being more favorable in the SRS group, ter) in terms of survival (median OS of 13.6 months for patients
survival was longer in the FSRT cohort (11 vs. 12 months, treated with conventional therapy vs. 13.5 months in the radio-
respectively) and complications were lower (30% vs. 8%, respec- surgery followed by conventional therapy arm; p = 0.5711).15
tively). These results suggested that FSRT may be a more appro- However, a variety of criticisms have been leveled against the
priate choice in this setting, especially for large tumors or those RTOG 93–05 trial design, most typically (1) few centers have
in critical areas. In other reports, median survival of 7 months used radiosurgery boost prior to conventional radiotherapy as
was achieved in 88 patients with recurrent GBM when Taxol it was in 93–05,36,50,51 and (2) carmustine is no longer standard
(120 mg/m2) was used as a radiation sensitizer for multisession first-line chemotherapy for GBM.9,36,51 As a result, other authors
radiosurgery (24 Gy in four sessions, mean).48,49 Patients with have continued to publish on the potential utility of SRS as part
tumor volume less than 30 cm3 survived significantly longer of the overall up-front treatment for high-grade gliomas.
than those with larger tumors. One of the earliest reports of radiosurgery as a boost for
Although none of these series is free from potential selection malignant gliomas was from the Joint Center by Loeffler et al. 52
bias, there do appear to be some objective responses to SRS in Twenty-three GBM and 14 anaplastic astrocytoma patients
the setting of recurrent malignant glioma. In ▶ Fig. 27.3, we underwent radiosurgery of enhancing tissue 2 to 4 weeks after
illustrate the case of a 63-year-old woman with recurrent GBM. completion of conventional radiotherapy (5940 cGy, max).
This tumor was treated with GKSRS using 18 Gy to the 50% IDL. Median SRS minimum dose was 12 Gy (range 10–20 Gy) and
Tumor regression was achieved; she survived 23 months after maximum dose was 15 Gy (range 12.5–25.0 Gy). Tumor volume
radiosurgery. Despite these observations, the literature suggests ranged from 1.2 to 72 cm3. Only 24% of patients had died at 19-
that these treatments are ultimately palliative in nature, as local month median follow-up. This, as well as subsequent studies53,

Fig. 27.3 (a) A 63-year-old woman with recurrent


glioblastoma. The tumor was treated with Gam-
ma Knife stereotactic radiosurgery using 18 Gy to
the 50% isodose line. (b) At 9-month postster-
eotactic radiosurgery treatment, the tumor
shows regression on follow-up magnetic reso-
nance imaging.

230
Radiosurgery for Glial Tumors

54,55 concluded that SRS was a potentially useful adjunct in the diagnosed GBM treated between 2002 and 2007. Temozolo-
management of these lesions. The Joint Center for Radiation mide (TMZ) was not given to the initial 54% of patients because
Therapy (Boston, MA), University of Wisconsin–Madison, and the phase III trial data establishing its use had not yet been pub-
University of Florida–Gainesville, pooled their data and found lished. The majority of patients included in the Einstein report
that median survival was 24 months among 115 GBM patients underwent subtotal surgical resection (27/35) and a small per-
treated with radiosurgery as part of initial management. 56 centage (4/35) had biopsy as initial surgical management. After
Additionally, Alexander et al found results similar to those pre- a pathological diagnosis of GBM, MRS was undertaken to identi-
viously seen with brachytherapy in terms of both benefit and fy areas within the T2 MRI abnormality believed to be at high
toxicity.57 Interpretation of these studies has been cautious risk for harboring residual tumor or to be at high risk for tumor
because they were nonrandomized, raising the possibility for recurrence. Patients were treated with GKSRS prior to confor-
significant skewing of the results due to potential selection mal radiotherapy to “minimize the impact of post-radiation
bias.5,15,58,59 Several of these early studies attempted to mini- changes on the MRS analysis.”63 The treatment paradigm pro-
mize the influence of selection bias on their outcomes by inter- duced a median OS of 15.8 months for the entire cohort. The
preting their results according to RTOG classifications of median OS stratified by RTOG RPA classification was greater
outcome. Sarkaria et al found the median survival difficult to than 22 months for RPA class 3 patients (75% were still alive at
assess, but the 2-year survival for GBM patients to be signifi- the time of their report), 18.7 months for RPA class 4, 12.5
cantly better than predicted,56 whereas Mehta et al showed sig- months for RPA class 5, and 3.9 months for RPA class 6—with
nificant improvement over expected outcome for both median statistically significant differences between the respective sur-
and 2-year survival.60 vival times. Comparing the groups treated with or without TMZ
Biswas et al retrospectively analyzed their experience using showed a difference in median OS of 20.8 months (for patients
SRS to treat GBM in an effort to better define what conditions with concurrent TMZ) versus 11 months (for those not receiv-
influence its efficacy.61 Their study included 33 patients treated ing TMZ). The authors reported 9 of 35 patients experiencing
between 2000 and 2007, all of whom had pathological confir- grade 3 or 4 toxicity. These included one patient with post-SRS
mation of GBM. Eighteen of these patients were treated with stroke (patient on warfarin) and eight patients requiring reope-
SRS at tumor recurrence; 15 were treated up-front after com- ration (five patients with recurrent tumor and three with
pletion of standard fractionated radiotherapy. The median OS symptomatic radiation necrosis). The authors concluded that
after initial diagnosis was 16.9 months and median survival OS is improved in patients treated with MRS-targeted SRS com-
after SRS was 6.7 months. There was no statistical difference in pared with historical controls.
median OS between the groups treated with SRS at recurrence Finally, Pannullo et al conducted a meta-analysis of current
versus those treated as part of their initial treatment paradigm. therapeutic applications of radiosurgery in neuro-oncology. 64
Univariate analysis showed a difference in time to progression As part of this meta-analysis, the authors screened studies eval-
for patients treated with up-front radiosurgery boost compared uating the use of radiosurgery for GBM. Eleven studies were
with those receiving SRS at tumor recurrence (6 months vs. 3.4 included in their meta-analysis (which comprised about 450
months). No acute toxicity greater than grade 2 was reported. total GBM patients)—nine studies were graded as providing
The authors concluded that radiosurgery is well tolerated as a class III evidence and two were graded as class II evidence: one
part of GBM management and that further studies are needed case-control study and one prospective randomized study. Six
to more adequately define patients likely to respond to SRS. of the studies were case series reporting on local experience
Pouratian et al retrospectively analyzed their experience with with adjuvant or salvage SRS in the treatment of GBM. The oth-
SRS for patients with GBM.51 Their report included 48 patients er five studies compared SRS with a different, designated com-
with histologically confirmed GBM, where SRS was part of the parison treatment group. The ranges for median survival were
initial treatment paradigm (22 patients) or SRS was utilized at similar whether SRS was given as adjunctive initial treatment
tumor progression (26 patients). The patients treated at tumor (9.5–25 months) or as salvage therapy at progression/recur-
progression had longer median OS than those treated on initial rence (10.2–26 months). The overall complication rate for SRS
presentation, 17.4 versus 15.1 months, respectively. In their in GBM treatment was calculated as 11.4%.
multivariate analysis, RTOG recursive partitioning analysis Although the impact of SRS on a typical patient with high-
(RPA) class 3 patients fared better than other patients, as did grade glioma remains unknown, some patients with malignant
those patients with more extensive surgical resections and glioma treated with SRS do have prolonged survival. ▶ Fig. 27.4
those not dependent on steroids. Similarly, Villavicencio et al shows the case of a 36-year-old woman with a biopsy-proven
reported their experience with SRS for treatment of GBM GBM of the right temporal stem. This was treated with linear
patients.62 This retrospective study of 46 patients included 20 accelerator (linac)-based SRS using 25 Gy to the 80% IDL and
patients treated at the time of initial diagnosis and 26 treated at 59.4 Gy in 33 fractions of conventional radiotherapy. The pa-
tumor progression. Median OS was better for patients treated at tient remains alive today 22 years after SRS, with no evidence
progression rather than as part of the initial treatment para- of tumor recurrence or progression.
digm, 21 versus 11.5 months, respectively, leading the authors
to conclude that SRS should be reserved for patients whose
tumors recur or progress after conventional therapy.
In 2012 Einstein et al reported on their phase II prospective
27.3 Complications
trial utilizing magnetic resonance spectroscopy (MRS) to define Despite the numerous series and individual anecdotal cases
and target high-risk tumor volumes for subsequent treatment presented above, it is clear that radiosurgery for gliomas may
with SRS.63 The study included 35 patients with newly be associated with toxicity, especially with high prescription

231
Radiosurgery for Glial Tumors

Fig. 27.4 (a) A 36-year-old woman with biopsy-


proven glioblastoma of the right temporal stem.
The tumor was treated with linear accelerator–
based radiosurgery using 25 Gy to the 80%
isodose line immediately preceded by 59.4 Gy of
conventional radiotherapy delivered in 33 frac-
tions. (b) The patient remains alive today 22 years
post stereotactic radiosurgery treatment without
evidence of tumor recurrence or progression.

doses or when large lesions are treated. Potential complications 29 (13.8%).60 Additionally, the rate of reoperation in patients
from SRS for gliomas are related to two factors: the radiosurgi- treated with SRS as part of an initial treatment algorithm was
cal treatment itself and our current interpretation of the avail- reported to vary from 19 to 33%. The studies reviewed regard-
able literature as it pertains to this type of treatment for glial ing use of SRS treatment at the time of glioma progression or
lesions. Determining whether complications from treatment recurrence showed similar potential toxicities, ultimately lead-
have occurred may also be difficult. Simply distinguishing ing to the conclusion that although there may be some evidence
recurrent tumor from posttreatment radiation necrosis may for improved survival and local control with SRS as salvage glio-
require special imaging studies (such as MRS, PET, or nuclear ma treatment, this may be counter-balanced by possible
medicine spectroscopy) or even biopsy.65–70 toxicity.5
With regard to complications from the radiosurgery itself,
these are similar to the complications seen with cranial SRS for
other malignant or nonmalignant pathologies. Some of these 27.4 Summary
include the potential for tumor recurrence despite treatment,
Prior to the report of RTOG 93–05, many providers hoped this
the potential for local radionecrosis as a result of treatment,
trial would provide definitive proof either for or against the use
and the potential for radiation injury to adjacent critical neuro-
of radiosurgery as at least a component in the initial treatment
vascular structures (optic pathways, brainstem, etc.). In con-
of patients with GBM. As reviewed above, RTOG 93–05 reported
trast, some potential complications from SRS in glioma
no outcome advantage and no clear quality-of-life or cognitive
treatment are related to the quality of and our current under-
function advantage to adding radiosurgery before conventional
standing of the available salient literature (which is relatively
radiation therapy and carmustine therapy in the treatment of
limited). In regard to the depth and quality of the current litera-
GBM. However, after the 93–05 report was published a number
ture, there has been a single prospective randomized study and
of important criticisms of the study have been made.36,50,51
a small variety of other class II and III studies used to argue for
As a result, questions regarding the role for radiosurgery in
and against the use of SRS in the treatment of gliomas. This
the up-front or salvage treatment of glial tumors remain insuffi-
gives rise to the possibility that any reported positive treatment
ciently answered. Many different “camps” remain with regard
outcomes may be inappropriately overemphasized and the
to the “most acceptable” interpretation of the current literature
potential for significant treatment related toxicities may be
supporting using or withholding radiosurgery in the treatment
underappreciated.
of glial tumors. Further prospective randomized trials to answer
RTOG 93–05 reported four grade 3 late toxicities in the SRS
the remaining questions surrounding the role of SRS in the
study arm—three neurologic and one classified as “other” (all of
treatment of gliomas are not likely to be forthcoming in the
these patients received 15 Gy SRS boost)—compared with a sin-
near future. For the time being, decisions to accept or reject the
gle late grade 3 toxicity in the radiation therapy study arm. For
use of SRS treatment in these challenging cases will most fre-
the same study, the rate and type of late grade 1 and 2 toxicities
quently be made at the individual clinician level or possibly the
were similar between the SRS and radiation therapy arms.
individual institutional level based on personal experience and
Additionally in RTOG 93–05, for all patients who underwent
influenced by the imperfect literature that has been generated
salvage surgery there were more with necrosis only identified
thus far.
in the salvage pathology specimen from the SRS arm (7/28)
than from the radiation therapy arm (3/31).15
Tsao et al reported on summary data regarding complications References
from SRS treatment in both newly diagnosed and recurrent glial
[1] Buatti JM, Friedman WA, Bova FJ, Mendenhall WM. Treatment selection fac-
tumors. The articles reviewed in Tsao’s report that address the
tors for stereotactic radiosurgery of intracranial metastases. Int J Radiat Oncol
use of radiosurgery in newly diagnosed malignant glioma Biol Phys 1995; 32: 1161–1166
report toxicities ranging from “no significant acute or late toxic- [2] Coffey RJ, Flickinger JC, Bissonette DJ, Lunsford LD. Radiosurgery for solitary
ities” to edema and radiation necrosis.5 The reported rates of brain metastases using the cobalt-60 gamma unit: methods and results in 24
radiation necrosis ranged from 2 out of 37 (5.4%) 52 to 4 out of patients. Int J Radiat Oncol Biol Phys 1991; 20: 1287–1295

232
Radiosurgery for Glial Tumors

[3] Kondziolka D, Patel A, Lunsford LD, Kassam A, Flickinger JC. Stereotactic ra- [25] Barnett GH, Kormos DW, Steiner CP, Weisenberger J. Use of a frameless, arm-
diosurgery plus whole brain radiotherapy versus radiotherapy alone for pa- less stereotactic wand for brain tumor localization with two-dimensional and
tients with multiple brain metastases. Int J Radiat Oncol Biol Phys 1999; 45: three-dimensional neuroimaging. Neurosurgery 1993; 33: 674–678
427–434 [26] Taylor MD, Bernstein M. Awake craniotomy with brain mapping as the rou-
[4] Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treatment of tine surgical approach to treating patients with supratentorial intraaxial tu-
recurrent previously irradiated primary brain tumors and brain metasta- mors: a prospective trial of 200 cases. J Neurosurg 1999; 90: 35–41
ses: final report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys [27] Black PM, Alexander E III, Martin C, et al. Craniotomy for tumor treatment in
2000; 47: 291–298 an intraoperative magnetic resonance imaging unit. Neurosurgery 1999; 45:
[5] Tsao MN, Mehta MP, Whelan TJ, et al. The American Society for Therapeu- 423–431, discussion 431–433
tic Radiology and Oncology (ASTRO) evidence-based review of the role of [28] Plowman PN. Stereotactic multiple arc radiotherapy. V: primary treatment of
radiosurgery for malignant glioma. Int J Radiat Oncol Biol Phys 2005; 63: discrete low grade glioma. Br J Neurosurg 1997; 11: 331–334
47–55 [29] Barcia JA, Barcia-Salorio JL, Ferrer C, Ferrer E, Algás R, Hernández G. Stereo-
[6] Walker MD, Alexander E Jr, Hunt WE, et al. Evaluation of BCNU and/or radio- tactic radiosurgery of deeply seated low grade gliomas. Acta Neurochir Suppl
therapy in the treatment of anaplastic gliomas. A cooperative clinical trial. J (Wien) 1994; 62: 58–61
Neurosurg 1978; 49: 333–343 [30] Kihlström L, Lindquist C, Lindquist M, Karlsson B. Stereotactic radiosurgery
[7] Walker MD, Green SB, Byar DP, et al. Randomized comparisons of radiother- for tectal low-grade gliomas. Acta Neurochir Suppl (Wien) 1994; 62: 55–57
apy and nitrosoureas for the treatment of malignant glioma after surgery. N [31] Park KJ, Kano H, Kondziolka D, Niranjan A, Flickinger JC, Lunsford LD. Early or
Engl J Med 1980; 303: 1323–1329 delayed radiosurgery for WHO grade II astrocytomas. J Neurooncol 2011;
[8] Cairncross JG, Ueki K, Zlatescu MC, et al. Specific genetic predictors of chemo- 103: 523–532
therapeutic response and survival in patients with anaplastic oligodendro- [32] Hochberg FH, Pruitt A. Assumptions in the radiotherapy of glioblastoma. Neu-
gliomas. J Natl Cancer Inst 1998; 90: 1473–1479 rology 1980; 30: 907–911
[9] Stupp R, Mason WP, van den Bent MJ, et al. European Organisation for Re- [33] Wallner KE, Galicich JH, Krol G, Arbit E, Malkin MG. Patterns of failure follow-
search and Treatment of Cancer . Brain Tumor and Radiotherapy Groups. Na- ing treatment for glioblastoma multiforme and anaplastic astrocytoma. Int J
tional Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus con- Radiat Oncol Biol Phys 1989; 16: 1405–1409
comitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; [34] Bleehen NM, Stenning SP, The Medical Research Council Brain Tumour Work-
352: 987–996 ing Party. A Medical Research Council trial of two radiotherapy doses in the
[10] Florell RC, Macdonald DR, Irish WD, et al. Selection bias, survival, and brachy- treatment of grades 3 and 4 astrocytoma. Br J Cancer 1991; 64: 769–774
therapy for glioma. J Neurosurg 1992; 76: 179–183 [35] Walker MD, Strike TA, Sheline GE. An analysis of dose-effect relationship in
[11] Gutin PH, Prados MD, Phillips TL, et al. External irradiation followed by an in- the radiotherapy of malignant gliomas. Int J Radiat Oncol Biol Phys 1979; 5:
terstitial high activity iodine-125 implant “boost” in the initial treatment of 1725–1731
malignant gliomas: NCOG study 6G-82–2. Int J Radiat Oncol Biol Phys 1991; [36] Sheehan J. Stereotactic radiosurgery for glioblastoma—time to revisit this ap-
21: 601–606 proach. World Neurosurg 2012; 78: 592–593
[12] Laperriere NJ, Leung PM, McKenzie S, et al. Randomized study of brachyther- [37] Hall WA, Djalilian HR, Sperduto PW, et al. Stereotactic radiosurgery for recur-
apy in the initial management of patients with malignant astrocytoma. Int J rent malignant gliomas. J Clin Oncol 1995; 13: 1642–1648
Radiat Oncol Biol Phys 1998; 41: 1005–1011 [38] van Kampen M, Engenhart-Cabillic R, Debus J, Fuss M, Rhein B, Wannen-
[13] Prados MD, Gutin PH, Phillips TL, et al. Highly anaplastic astrocytoma: a re- macher M. [The radiosurgery of glioblastoma multiforme in cases of recur-
view of 357 patients treated between 1977 and 1989. Int J Radiat Oncol Biol rence. The Heidelberg experiences compared to the literature]. Strahlenther
Phys 1992; 23: 3–8 Onkol 1998; 174: 19–24
[14] Selker RG, Shapiro WR, Burger P, et al. Brain Tumor Cooperative Group. The [39] Park J, Suh J, Barnett G, et al. Survival after stereotactic radiosurgery for re-
Brain Tumor Cooperative Group NIH Trial 87–01: a randomized comparison current glioblastoma multiforme. J Radiosurgery 2000; 3: 169–175
of surgery, external radiotherapy, and carmustine versus surgery, interstitial [40] Larson DA, Gutin PH, McDermott M, et al. Gamma knife for glioma: selection
radiotherapy boost, external radiation therapy, and carmustine. Neurosur- factors and survival. Int J Radiat Oncol Biol Phys 1996; 36: 1045–1053
gery 2002; 51: 343–355, discussion 355–357 [41] Chamberlain MC, Barba D, Kormanik P, Shea WM. Stereotactic radiosurgery
[15] Souhami L, Seiferheld W, Brachman D, et al. Randomized comparison of ster- for recurrent gliomas. Cancer 1994; 74: 1342–1347
eotactic radiosurgery followed by conventional radiotherapy with carmustine [42] Sanghavi SN, Miranpuri SS, Chappell R, et al. Radiosurgery for patients with
to conventional radiotherapy with carmustine for patients with glioblastoma brain metastases: a multi-institutional analysis, stratified by the RTOG recur-
multiforme: report of Radiation Therapy Oncology Group 93–05 protocol. Int sive partitioning analysis method. Int J Radiat Oncol Biol Phys 2001; 51: 426–
J Radiat Oncol Biol Phys 2004; 60: 853–860 434
[16] Proust F, Coche-Dequeant B, Carpentier P, et al. [Combination treatment for [43] Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy
pilocytic astrocytoma: stereotaxic radiosurgery and endocavitary radiother- with or without stereotactic radiosurgery boost for patients with one to three
apy]. Neurochirurgie 1998; 44: 50–54 brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet
[17] Somaza SC, Kondziolka D, Lunsford LD, Flickinger JC, Bissonette DJ, Albright 2004; 363: 1665–1672
AL. Early outcomes after stereotactic radiosurgery for growing pilocytic as- [44] Shrieve DC, Alexander E III, Wen PY, et al. Comparison of stereotactic radio-
trocytomas in children. Pediatr Neurosurg 1996; 25: 109–115 surgery and brachytherapy in the treatment of recurrent glioblastoma multi-
[18] Lim YJ, Leem W. Two cases of Gamma Knife radiosurgery for low-grade optic forme. Neurosurgery 1995; 36: 275–282, discussion 282–284
chiasm glioma. Stereotact Funct Neurosurg 1996; 66 (Suppl 1): 174–183 [45] Larson DA, Prados M, Lamborn KR, et al. Phase II study of high central dose
[19] Liang CL, Lu K, Liliang PC, Chen HJ. Gamma Knife surgery for optic glioma. Re- Gamma Knife radiosurgery and marimastat in patients with recurrent malig-
port of 2 cases. J Neurosurg 2010; 113 (Suppl): 44–47 nant glioma. Int J Radiat Oncol Biol Phys 2002; 54: 1397–1404
[20] Kano H, Niranjan A, Kondziolka D, et al. Stereotactic radiosurgery for pilocytic [46] Skeie BS, Enger PO, Brøgger J, et al. γ knife surgery versus reoperation for re-
astrocytomas part 2: outcomes in pediatric patients. J Neurooncol 2009; 95: current glioblastoma multiforme. World Neurosurg 2012; 78: 658–669
219–229 [47] Cho KH, Hall WA, Gerbi BJ, Higgins PD, McGuire WA, Clark HB. Single dose
[21] Weintraub D, Yen CP, Xu Z, Savage J, Williams B, Sheehan J. Gamma knife sur- versus fractionated stereotactic radiotherapy for recurrent high-grade glio-
gery of pediatric gliomas. J Neurosurg Pediatr 2012; 10: 471–477 mas. Int J Radiat Oncol Biol Phys 1999; 45: 1133–1141
[22] Hallemeier CL, Pollock BE, Schomberg PJ, Link MJ, Brown PD, Stafford SL. Ster- [48] Lederman G, Arbit E, Odaimi M, Lombardi E, Wrzolek M, Wronski M. Fractio-
eotactic radiosurgery for recurrent or unresectable pilocytic astrocytoma. Int nated stereotactic radiosurgery and concurrent taxol in recurrent glioblasto-
J Radiat Oncol Biol Phys 2012; 83: 107–112 ma multiforme: a preliminary report. Int J Radiat Oncol Biol Phys 1998; 40:
[23] Debus J, Kocagoncu KO, Hoss A, Wenz F, Wannenmacher M. Fractionated ster- 661–666
eotactic radiotherapy (FSRT) for optic glioma. Int J Radiat Oncol Biol Phys [49] Lederman G, Wronski M, Arbit E, et al. Treatment of recurrent glioblastoma
1999; 44: 243–248 multiforme using fractionated stereotactic radiosurgery and concurrent pa-
[24] Combs SE, Schulz-Ertner D, Moschos D, Thilmann C, Huber PE, Debus J. clitaxel. Am J Clin Oncol 2000; 23: 155–159
Fractionated stereotactic radiotherapy of optic pathway gliomas: toler- [50] Kondziolka D, Lunsford LD, Flickinger JC. In regard to Dr. Souhami et al. (Int J
ance and long-term outcome. Int J Radiat Oncol Biol Phys 2005; 62: Radiat Oncol Biol Phys 2004;60:853–860). Int J Radiat Oncol Biol Phys 2005;
814–819 62: 614–615, author reply 615–616

233
Radiosurgery for Glial Tumors

[51] Pouratian N, Crowley RW, Sherman JH, Jagannathan J, Sheehan JP. Gamma [61] Biswas T, Okunieff P, Schell MC, et al. Stereotactic radiosurgery for glioblasto-
Knife radiosurgery after radiation therapy as an adjunctive treatment for glio- ma: retrospective analysis. Radiat Oncol 2009; 4: 11
blastoma. J Neurooncol 2009; 94: 409–418 [62] Villavicencio AT, Burneikiene S, Romanelli P, et al. Survival following stereo-
[52] Loeffler JS, Alexander E III, Shea WM, et al. Radiosurgery as part of the initial tactic radiosurgery for newly diagnosed and recurrent glioblastoma multi-
management of patients with malignant gliomas. J Clin Oncol 1992; 10: forme: a multicenter experience. Neurosurg Rev 2009; 32: 417–424
1379–1385 [63] Einstein DB, Wessels B, Bangert B, et al. Phase II trial of radiosurgery to
[53] Gannett D, Stea B, Lulu B, Adair T, Verdi C, Hamilton A. Stereotactic radiosur- magnetic resonance spectroscopy-defined high-risk tumor volumes in pa-
gery as an adjunct to surgery and external beam radiotherapy in the treat- tients with glioblastoma multiforme. Int J Radiat Oncol Biol Phys 2012;
ment of patients with malignant gliomas. Int J Radiat Oncol Biol Phys 1995; 84: 668–674
33: 461–468 [64] Pannullo SC, Fraser JF, Moliterno J, Cobb W, Stieg PE. Stereotactic radiosur-
[54] Kondziolka D, Flickinger JC, Bissonette DJ, Bozik M, Lunsford LD. Survival ben- gery: a meta-analysis of current therapeutic applications in neuro-oncologic
efit of stereotactic radiosurgery for patients with malignant glial neoplasms. disease. J Neurooncol 2011; 103: 1–17
Neurosurgery 1997; 41: 776–783, discussion 783–785 [65] Chao ST, Suh JH, Raja S, Lee SY, Barnett G. The sensitivity and specificity
[55] Shrieve DC, Alexander E III, Black PM, et al. Treatment of patients with pri- of FDG PET in distinguishing recurrent brain tumor from radionecrosis in
mary glioblastoma multiforme with standard postoperative radiotherapy and patients treated with stereotactic radiosurgery. Int J Cancer 2001; 96:
radiosurgical boost: prognostic factors and long-term outcome. J Neurosurg 191–197
1999; 90: 72–77 [66] Graves EE, Nelson SJ, Vigneron DB, et al. A preliminary study of the prognostic
[56] Sarkaria JN, Mehta MP, Loeffler JS, et al. Radiation Therapy Oncology Group. value of proton magnetic resonance spectroscopic imaging in gamma knife
Radiosurgery in the initial management of malignant gliomas: survival com- radiosurgery of recurrent malignant gliomas. Neurosurgery 2000; 46: 319–
parison with the RTOG recursive partitioning analysis. Int J Radiat Oncol Biol 326, discussion 326–328
Phys 1995; 32: 931–941 [67] Kline JL, Noto RB, Glantz M. Single-photon emission CT in the evaluation
[57] Alexander E III, Loeffler JS. Radiosurgery for primary malignant brain tumors. of recurrent brain tumor in patients treated with gamma knife radiosur-
Semin Surg Oncol 1998; 14: 43–52 gery or conventional radiation therapy. AJNR Am J Neuroradiol 1996; 17:
[58] Irish WD, Macdonald DR, Cairncross JG. Measuring bias in uncontrolled brain 1681–1686
tumor trials—to randomize or not to randomize? Can J Neurol Sci 1997; 24: [68] Schlemmer HP, Bachert P, Herfarth KK, Zuna I, Debus J, van Kaick G. Proton
307–312 MR spectroscopic evaluation of suspicious brain lesions after stereotactic ra-
[59] Loeffler JS, Shrieve DC, Alexander E III. Radiosurgery for glioblastoma multi- diotherapy. AJNR Am J Neuroradiol 2001; 22: 1316–1324
forme: the importance of selection criteria. Int J Radiat Oncol Biol Phys 1994; [69] Schwartz RB, Hsu L, Kacher DF, et al. Intraoperative dynamic MRI: localization
30: 731–733 of sites of brain tumor recurrence after high-dose radiotherapy. J Magn Reson
[60] Mehta MP, Masciopinto J, Rozental J, et al. Stereotactic radiosurgery for glio- Imaging 1998; 8: 1085–1089
blastoma multiforme: report of a prospective study evaluating prognostic [70] Chao ST, Ahluwalia MS, Barnett GH, et al. Challenges with the diagnosis and
factors and analyzing long-term survival advantage. Int J Radiat Oncol Biol treatment of cerebral radiation necrosis. Int J Radiat Oncol Biol Phys 2013;
Phys 1994; 30: 541–549 87: 449–457

234
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

28 Stereotactic Radiosurgery for the Management of One


to Four Brain Metastases
Sten Myrehaug, Simon S. Lo, Siavash Jabbari, Lijun Ma, Sunit Das, Aliaksandr Karotki, Eric L. Chang, and Arjun Sahgal

performance status, age, and stability of extracranial disease,


Key Points placing patients into three RPA groups with correspondent
median survival times of 7 months, 3 to 6 months, and approx-
● Stereotactic radiosurgery plays a dominant role in the ma- imately 2 months. Given the heterogeneous nature of brain
nagement of patients with one to four brain metastases < 3 metastasis presentation and biological characteristics, the
cm in size, and an alternative to surgery for tumors not re- Graded Prognostic Assessment (GPA) and diagnosis-specific
quiring urgent resection. GPA (DS-GPA) scoring systems have been subsequently devel-
● Despite improvements in intracranial control, the addition of oped,2 incorporating more disease-specific parameters such as
whole-brain radiotherapy to stereotactic radiosurgery does the number of metastases and in some cases molecular profil-
not improve overall survival and is associated with worsened ing. As a result, we have an improved ability to tailor individu-
neurocognitive and quality-of-life outcomes. alized treatment plans for patients, selecting when appropriate
● Postoperative single fraction or hypofractionated radiosur- focal SRS and/or surgery, and aim to maximize local control
gery provides excellent local control while sparing radiation rather than to simply temporize brain control with WBRT.
exposure to the normal brain.
● Radiation technologies have evolved such that most modern
units are able to perform radiosurgery. This is increasing its
availability beyond specialized tertiary care centers; more-
28.3 Role of Surgical Resection
over, the technology has made advances such that we can In patients with solitary metastasis, good performance status,
perform frameless image-guided radiosurgery. patients and controlled extracranial disease, surgery followed by
WBRT can improve OS, local control, median time to death from
neurologic cause, and preservation of performance status as
28.1 Introduction compared with WBRT alone. Patchell et al3 established the role
of surgical resection followed by WBRT versus WBRT alone, in
An estimated 20 to 40% of cancer patients will develop brain
the management of solitary brain metastasis, with improvement
metastases during the course of their illness, more commonly
in survival (10 vs. 4.2 months), recurrence at original site of dis-
in patients with lung carcinoma, colorectal carcinoma, breast
ease (20% vs. 50%), time to recurrence, median time to death
carcinoma, renal cell carcinoma, or melanoma. Solitary brain
from neurologic cause, and preservation of performance status
metastases are seen in 40 to 50% of presentations, with multi-
(8.7 vs. 1.8 months). Importantly, the role of surgical resection
focal or leptomeningeal disease occurring in the remainder.
for multiple brain metastases is not as defined, and the Patchell
Brain metastases are a significant cause of morbidity and mor-
et al study is specific to solitary metastases. Neurosurgical prin-
tality in cancer patients. Patients with untreated brain metastasis
ciples are such that surgery is performed for patients even in
have a poor prognosis, with a life expectancy of 1 to 2 months,
the presence of multiple metastases for large brain metastasis
which can be improved slightly with the use of corticosteroid
(> 3 cm), those causing significant edema, and/or those causing
therapy. Whole-brain radiotherapy (WBRT) has been utilized for
neurologic symptoms, as removing the tumor is the fastest way
over 60 years, and has yielded modest gains with respect to
to yield clinical improvement in a deteriorating patient.
improvements in neurologic symptoms, intracranial disease con-
Recommendation: Patients with solitary metastases should
trol, and overall survival (OS). As therapeutic and technical advan-
have a neurosurgical consult. Any patient with a brain metasta-
ces in cancer treatment and medical imaging have evolved, earlier
sis that causes life-threatening signs or symptoms should have
identification of central nervous system (CNS) disease has resulted
a surgical consultation regardless of the number of metastases.
with a greater proportion of patients presenting with a limited
number of brain metastases (i.e., one to four metastases), and the
ubiquitous use of WBRT has been questioned. This has led to the
development of more aggressive approaches to the treatment of 28.4 Postoperative Management
intracranial disease such as stereotactic radiosurgery (SRS). In the
The routine use of WBRT was then questioned in patients with
contemporary management of patients presenting with one to
solitary metastasis undergoing surgery, and resulted in a
four metastases, SRS has a dominant role in treating these
randomized trial evaluating surgery alone to surgery plus
patients and must be discussed with the appropriate patient.
WBRT in good performance study patients with controlled
extracranial disease. Patchell et al reported that phase III study, 4
28.2 Patient Selection and postoperative WBRT was found to significantly reduce the
rate of local recurrence (10% vs. 46%), distant brain failures
The Radiation Therapy Oncology Group (RTOG) recursive parti- (14% vs. 37%), and neurologic deaths (14% vs. 44%). However, OS
tioning analysis (RPA) has been the most commonly used prog- and duration of functional independence outcomes were simi-
nostic scale and treatment decision-making tool for patients lar between the two cohorts. This study ultimately concluded
with brain metastases.1 This scale assesses patients based upon that postoperative WBRT was the standard of care.

235
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

A more recent randomized trial by Kocher et al evaluated the and improved QoL scores at 6-week posttherapy. Although oth-
role of WBRT versus observation following either surgery or er retrospective series have confirmed this finding,13,14,15 a con-
SRS.5 In the surgical cohort (160 patients), postoperative WBRT trary report was published by the MD Anderson group
decreased intracranial disease progression at 24 months (31% vs. (Houston, TX) in 1996, who matched 13 patients treated with
54%, respectively) and the risk of neurologic death (25% vs. 43%), SRS with 62 treated with surgical resection alone and found
while not impacting overall survival or preservation of perform- that local recurrence was superior in the surgical cohort (21%
ance status. Hence nearly a decade later, the yield of postopera- vs. 8%, respectively).16
tive WBRT was similar to the original Patchell et al trial.4 Recommendation: Surgery should be recommended in gener-
Concern has been raised over the association of WBRT with al if tissue diagnosis is required, for solitary lesions that are sur-
neurotoxicity and subsequent declines in quality-of-life (QoL) gically resectable, and those metastases > 3 to 4 cm or causing
domains such as fatigue6 and cognitive impairment7 (discussed considerable edema and/or mass effect (in particular when in
in a later section). As a result, postoperative cavity radiation the posterior fossa). Stereotactic radiosurgery is an appropriate
with the use of single-fraction or hypofractionated SRS has alternative for patients with small, multiple and/or deep
emerged and increasingly being practiced as a standard of care. lesions, or for patients not suitable for surgical resection.
A review by Roberge and Souhami summarized outcomes on
492 postoperative patients treated with SRS or hypofractio-
nated SRS to the resection cavity as reported in essentially ret-
28.5.1 Stereotactic Radiosurgery with
rospective and few prospective studies.8 Crude local control or without Whole-Brain Radiotherapy
(LC) was 79%, and the risk of radiation necrosis was estimated
With the recognition that local control following conventional
at approximately 5%. In 2014, the Memorial Sloan Kettering
WBRT alone is suboptimal,17 and treatment advances allowing
Cancer Center (New York, NY) published their phase II study9
for focal radiation, the next phase of trials emerged evaluating
evaluating single-fraction cavity radiosurgery (mean dose 1800
the role of SRS as a boost to WBRT and then as an alternative to
cGy), and concluded that significantly greater local control rates
WBRT. The randomized trials are summarized in ▶ Table 28.1.
could be achieved than with surgery alone. Al-Omair et al fur-
ther demonstrated that even in patients with significant prior
intracranial therapy demonstrating treatment resistance, sal- WBRT versus WBRT plus SRS
vage postoperative hypofractionated stereotactic radiotherapy An early study of 27 patients with two to four brain metastases
(HSRT) can provide excellent local control with an acceptable was published in 1999 by Kondziolka et al, in which patients
toxicity profile.10 Current trends in practice suggest that focal were randomized to WBRT alone versus WBRT plus SRS.18 Local
cavity radiation will replace WBRT, with a confirmatory failure was 100% at 1 year in patients treated with WBRT alone,
randomized study underway.11 and only 8% progressed in those boosted with SRS. A statisti-
Recommendation: There is sufficient literature, albeit uncon- cally nonsignificant OS benefit was observed in the SRS group
trolled, and a sensible therapeutic rationale to recommend focal (median survival (MS) 7.5 vs. 11 months). This study is contro-
radiation to the surgical cavity as an alternative option to versial, as the high local failure rate in the WBRT alone cohort
WBRT. However, whether single-fraction SRS or HSRT is opti- may reflect selection bias or lack of sufficient sample size.
mal remains to be determined. If available, patient enrollment The larger and more definitive study was subsequently
in a randomized trial comparing postoperative WBRT and SRS reported in 2004. RTOG 95–08 reported by Andrews et al 19
is preferable. randomized 331 patients to WBRT with or without SRS. Stereo-
tactic radiosurgery was shown to improve local control, Karnof-
sky Performance Score (KPS) steroid dependency, and for
28.5 Stereotactic Radiosurgery as patients with a single brain metastasis an improvement in OS
an Alternative to Surgery (6.5 vs. 4.9 months for WBRT alone). In addition, a trend to sup-
port a median survival advantage for RPA class I patients (11.6
There are no reported randomized trials directly comparing vs. 9.6 months), lung histology (5.9 vs. 3.9 months), and tumor
surgical resection to SRS. Given the lack of randomized data, we size > 2 cm (6.5 vs. 5.3 months) was observed in subgroup
primarily rely on retrospective series to guide treatment deci- analysis.
sion making. Due to the nature of these studies, results and Tsao et al reported a meta-analysis based on these two trials
interpretation are variable and conflicting. The study by Mua- using the published outcomes20 and concluded that although
cevic et al attempted to randomize patients with a single there was no difference in OS, with a hazard ratio (HR) of 1.63
resectable brain metastasis measuring ≤ 3 cm to surgery plus (95% confidence interval [CI] 0.72–3.69, p = 0.24), LC favored
WBRT versus SRS alone.12 This study was closed early due to WBRT plus SRS with a HR of 2.88 (95% CI 1.63–5.08, p = 0.003).
poor accrual; however, based on the results of 64 patients the Recommendation: If WBRT is offered to patients with one to
authors reported similar OS (median, 9.5 vs. 10.8 months, four brain metastases, SRS should also be considered in those
p = 0.8), neurologic death rates (29% vs. 11%, p = 0.3), and local with good KPS.
control (82% vs. 96%, p = 0.06) in the two groups. As expected,
the patients randomized to SRS alone were observed to have
more distant brain recurrences, although this difference was no SRS versus WBRT plus SRS
longer observed when accounting for salvage SRS. Stereotactic Three randomized studies have been reported evaluating SRS
radiosurgery alone was also associated with a shorter hospital- alone to WBRT plus SRS in patients with up to three to four
ization, decreased corticosteroid use, decreased neurotoxicity, metastases.5,7,21 Aoyama et al published the first study based on

236
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

Table 28.1 Summary of the phase III randomized controlled studies evaluating SRS and WBRT
Study Randomization % of single metastasis Primary end point Local control Survival

Kondziolka et al, SRS + WBRT (n = 13) 0% Local control 92% 11 mo


199918 WBRT (n = 14) 0% 7.5 mo

Andrews et al, 200419 WBRT + SRS (n = 164) 56% Overall survival 82%
WBRT (n = 167) 56% 71%

Aoyama et al, 200621 SRS (n = 67) 49% Tumor recurrence 72.5% 28.4%
WBRT + SRS 48% 88.7% 38.5%

Kocher et al, 20115 SRS (n = 100) 68% Functional independ- 69% 10.9 mo
WBRT + SRS (n = 99) 66% ence 81% 10.7 mo

Chang et al, 20097 SRS (n = 30) 60% Neurocognition 67% 63%


WBRT + SRS (n = 28) 54% 100% 21%

Abbreviations: SRS, stereotactic radiosurgery; WBRT, whole-brain radiation therapy.

132 patients randomized to SRS alone versus WBRT plus SRS 21 p < 0.00001).20 Given how the EORTC reported the OS data,
for patients with one to four metastases. Whole-brain radio- meta-analyses of the Aoyama and Chang studies could only be
therapy plus SRS was found to reduce the risk of developing performed. Individually, the Aoyama et al study reported no dif-
new brain metastases (63.7% vs. 41.5%, respectively) and ference in survival, whereas the Chang et al study reported a
improve the 1-year local control rate (72.5% vs. 88.7%). How- survival advantage favoring SRS alone. A meta-analysis con-
ever, no difference in OS (8.0 vs. 7.5 months) was identified. cluded no difference in OS with a HR of 0.98 (95% CI 0.71–1.35,
Neurocognitive functioning were measured using the Mini- p = 0.88),20 suggesting that SRS alone does not adversely impact
Mental Status Examination (MMSE) and no significant differen- survival, but the larger EORTC study could not be included,
ces were observed. However, it is well recognized that the which would have increased the power of the analysis. There-
MMSE is suboptimal for determining neurocognitive functions fore, as yet the potential for either treatment to influence OS is
most likely to be damaged by radiation.22 not known.
To study neurocognitive changes associated with WBRT, Recommendation: If the aim is to preserve neurocognition
Chang et al reported a trial evaluating 58 patients with one to and QoL, then SRS alone is the preferred therapeutic option.
three metastases randomized to SRS alone versus WBRT plus The drawback of SRS alone is a greater need for salvage proce-
SRS. The primary end point was neurocognitive changes at 4 dures, as the risk of distant brain relapse is greater, and to a
months using the Hopkins Verbal Learning Test (HVLT), as lesser extent local failure risk is also greater. If the patient is
opposed to the MMSE.7 Worse neurocognitive function with more concerned about overall brain control and less concerned
respect to memory at 4 months following treatment in the about radiation-induced neurocognitive deficits, then WBRT
WBRT plus SRS cohort was reported. This impairment in mem- plus SRS is the preferred strategy. In either case, close imaging-
ory was observed despite better local and distant brain tumor based follow-up is required to provide salvage therapy as
control in that cohort. needed given that the risk of distant brain relapse is still on the
In 2010, Kocher et al published the results of the European order of 30% despite WBRT.
Organisation for Research and Treatment of Cancer (EORTC)
22952–26001 study, in which 359 patients with up to three
metastases were randomized following either surgical resection 28.6 Radiation Planning
or SRS to observation versus adjuvant WBRT.5 The primary end
point was time to deterioration of performance status defined 28.6.1 Immobilization Systems
as a World Health Organization (WHO) status > 2. The addition Traditionally, SRS has required rigid head immobilization with
of WBRT, despite reducing the risk of local and distant relapse an invasive head frame; this frame also afforded the reference
and rate of neurologic death (28% vs. 44%), was not associated three-dimensional (3D) coordinate system to localize the target.
with improvements with respect to maintaining a good per- This has been the standard since the development of stereotaxis
formance status (10.0 vs. 9.5 months, p = 0.71) or OS (median, by Leksell in the 1950s. Despite the robust immobilization and
10.9 vs. 10.7 months, p = 0.89). A QoL analysis was also localization capabilities of the invasive head frame, there are
reported, and a detriment in QoL was observed in the WBRT significant drawbacks, including the requirement for neurosur-
arm despite gains in local control, distant brain control, and gical support, making it impractical for smaller oncology cen-
equivalent probability of maintaining a good performance ters to adopt SRS, the negative experience with head frame
status.23 placement perceived by patients, the small risk of bleeding and
Tsao et al performed a meta-analysis on the outcomes of local infection from pin insertions for head frame placement, and the
control and distant brain control based on the published data limitation of treatment to a single fraction (SRS) in most cases.
from these three trials.20 They concluded that the addition of Advances in head frame technology and the development of
WBRT improved local control and distant brain control vs. SRS on-board image guidance have allowed the development of
alone. The HR for local control was 2.61 (95% CI 1.68–4.06, “frameless” SRS. The same level of precision and accuracy can
p < 0.0001) and for distant brain control 2.15 (95% CI 1.55–2.99, now be achieved without the use of an invasive frame. Image

237
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

guidance is a key component to accurate delivery and can include the larger treatment volume space, making it easier to
include stereoscopic X-ray or on-board computed tomography treat tumors along the base of skull or at extreme lateral loca-
(CT)-based systems.24 Stereoscopic X-ray systems are based on tions within the cranium, and a single tungsten collimator hel-
real-time orthogonal images that are processed to yield spatial met now embedded into the unit and computer controlled. The
location of the target to be tracked based on bone or a visible design is based on multiple rings of collimating holes drilled
fiducial.24 The process of stereoscopic X-ray image guidance is directly into the one tungsten cast, creating nominal beam sizes
based on the X-rays taken before treatment in reference to the of 16, 8, and 4 mm in diameter at the isocenter. An eight-sliding
digitally reconstructed radiographs (DRRs) taken at the time of sector design allows each sector to slide along the outside sur-
simulation to ensure accurate patient positioning, and then sev- face of the tungsten collimator and align with the different
eral times per minute during the beam delivery to ensure that rings of prefabricated holes to allow for 192 individual 16-, 8-,
the position remains stable. When deviations in position are or 4-mm-diameter focused beams. Individual sectors are
detected, the linear accelerator (linac) position is adjusted, or blocked, so that delivered shots can be a composite of any of the
the patient position is adjusted via couch shifts, or both. The three beam diameters or zero within any of the eight sectors.
robotic arm is the core technology fundamental to CyberKnife- The latest innovations include an integrated relocatable head
based radiation delivery. frame that allows for fractionated SRS and the integration of
Online CT-based image guidance has almost completely taken image guidance with an on-board CBCT unit,28,29 allowing for
the form of cone-beam CT units (CBCT).24 The key advantage is frameless SRS and multiple fraction treatments.
the acquisition of high-quality 3D images such that the bony The CyberKnife (Accuray Inc., Sunnyvale, CA) is based on a
anatomy or tumor itself can be matched to the fan-beam-based minilinac mounted onto a robotic arm that is capable of moving
treatment-planning CT, and directly verified at the unit console. the linac in all 6 degrees of freedom. Near-real-time stereo-
The disadvantage to CBCT image guidance is the time required scopic image guidance and automated robotic beam adjustment
for image acquisition such that the treatment beam has to be system allows for high-precision treatment delivery.30 Recent
stopped, making real-time image guidance generally not possi- advances include the introduction of the IRIS collimator system,
ble. Multiple CBCTs during delivery are not practical due to pro- which is designed to improve efficiency of the treatment proc-
hibitive treatment times; so robust immobilization systems ess, as prior models required manual exchange the circular col-
play a more important role in maintaining high precision when limator if different diameter beams are required; a higher-
relying on CBCT alone for patient or beam alignment, rather output linac, again with the aim to reduce treatment time; and
than with stereoscopic imaging systems. This is in contrast to a new more sophisticated treatment-planning system.31
alignments with stereoscopic imaging systems where rigid A linac-based SRS was first developed by adding a tertiary
immobilization may be less critical; however, they are based on collimator onto an existing linac head to yield small circular
planar imaging information rather than based on the 3D imag- beams with a minimum diameter of 0.5 to 1 cm.24 Modifications
ing information of CBCT. to the treatment couch and the in-room laser system were
Infrared (IR) camera guidance is gaining acceptance.25,26 This required to ensure submillimeter precision in delivery. The
technology relies on reflective fiducial markers, placed on the treatment-planning strategy was based on delivering multiple
patient’s body or immobilization device, which are tracked con- beam arcs with the focal point being the linac isocenter. As the
tinuously by in-room infrared cameras during treatment. The technology improved and SRS was increasingly being applied to
markers provide real-time spatial feedback of the patient’s posi- tumors, one of the first dedicated linac systems built for intra-
tion, which is relayed to the beam delivery system and regis- cranial SRS was the Novalis (Brainlab, Munich, Germany). This
tered with the original stereotactic coordinates. If a deviation unit was also one of the first to integrate a stereoscopic X-ray
beyond preset thresholds is detected, treatment delivery is system into the delivery process to ensure precision and ulti-
automatically halted to allow for adjustments and patient repo- mately frameless SRS.
sitioning as required.26 The key issue with the use of reflective Modern linac systems are now developed with the intention
fiducial markers is robust registration and correlation between of delivering high-precision radiation. The technology has
the shifts in the markers versus the actual target shifts inside evolved to incorporate intensity-modulated radiotherapy
the patient body. Because of this, IR camera guidance is primar- (IMRT), and volume-modulated arc therapy (VMAT), which is
ily used for monitoring or assist in initial treatment setups possible due to the development of multileaf collimators
rather than being used as a primary target tracking or beam (MLCs). Now radiosurgery can be delivered over several
directing tool. minutes as opposed to hours. The precision in delivery is
ensured by image-guidance systems and robotic couch technol-
ogy. Further improvement in treatment efficiency has focused
28.6.2 Treatment Delivery Systems on maximizing machine output. Flattening filter-free techni-
The Gamma Knife unit is the hallmark SRS technology invented ques have become available that allow for higher mu/min out-
by Lars Leksell. The current unit is a self-contained unit deliver- put yielding delivery of radiosurgery in a few minutes. There is
ing 192 cobalt-based photon beams to an intracranial target, a point of caution, as the biological effects with these unprece-
with the patient immobilized using an invasive head frame.24,27 dented dose rates remain unclear, with potential for increased
The Gamma Knife has evolved to its latest model, the Leksell biological potency to affect both the tumor and the adjacent
Gamma Knife Perfexion (Elekta Instruments AB, Stockholm, normal tissues. As more clinical data accumulate, more knowl-
Sweden). This unit allows for great versatility in treatment edge about the safety profile of this technology will be gained.
planning and delivery, and this is most relevant to the treat- Although there is no doubt that add-on collimators to an
ment of multiple brain metastases. Key features of the Perfexion existing linac requires stringent quality assurance measures

238
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

to avoid catastrophic errors in delivery, intensity-modulated The historic dogma of single-fraction SRS has begun to shift,
radiosurgery using a MLC has its own drawbacks as well. In as frameless systems have allowed for HSRT, utilizing three to
particular, for lesions measuring < 1 cm, inherent uncertain- five fractions to deliver ablative radiation doses. This practice
ties in small-field dosimetry and beam-shaping concerns due began for the treatment of large lesions (> 4 cm), lesions associ-
to finite leaf widths limit the use of MLC for accurate dose ated with significant mass effect or for tumors adjacent to crit-
delivery. Tertiary collimators are still recommended for the ical organs-at-risk. The concept is that fractionation allows for
treatment of small targets. Another point of caution in using greater repair of the normal brain tissue, and hence allows for
MLC-based linac SRS is in the treatment of multiple targets. It dose escalation with a similar risk profile. Hypofractionated
has been shown that as the number of targets to be treated stereotactic radiotherapy is increasingly being practiced with
in a single session increases, so does the integral brain dose. 32 various fractionation schemes adopted. As yet there are no
This can become an issue in dose prescription, as in order to randomized trials completed to support superior efficacy and
maintain the same risk of radiation necrosis, using a periph- toxicity profiles in support of HSRT as compared with single-
eral isodose dose volume such as the 12-Gy volume as a sur- fraction SRS. Noteworthy series include that from Fahrig et al 37
rogate, one may have to reduce the dose prescribed, which who reported on 150 patients with 228 brain metastases
may compromise local control.33 Furthermore, the toxic effect treated with three different hypofractionated radiation doses:
of this low-dose spillage to the brain doses is unknown 5 × 6 to 7 Gy, 10 × 4 Gy, and 7 × 5 Gy. For tumors > 3 cm, although
because the threshold dose for neurocognitive damage is not 10 × 4 Gy was associated with no toxicity, improvements in res-
well defined, although this may not be an issue for solitary ponse rates were seen using the 5 × 6 to 7 Gy and 7 × 5 Gy pre-
targets.34 scriptions. The suitability of this range of dose fractionation was
also described by Märtens et al,38 who found improved local
control at doses with a biological equivalent dose (BED) > 35 Gy,
28.6.3 Dose Prescription and by the group at Emory39 (Atlanta, GA) who reviewed their
The maximum tolerated dose of single-fraction SRS was estab- experience using five different fractionation schedules, and giv-
lished according to the RTOG 90–05 dose-escalation SRS trial. en equivalent local control, favored the high BED and single-
In that trial, 156 patients with recurrent tumors < 4 cm in maxi- fraction equivalent dose (SFED) of the 6 Gy × 5 scheme. There is
mal diameter were treated with size as the primary stratifica- one series—comparing in a retrospective fashion—single-frac-
tion, and grade III–V neurotoxicity being the stopping end tion SRS to 36 Gy in six fractions HSRT.40 Despite being utilized
point. This study determined that the maximum tolerable doses for larger tumors or tumors in critical locations, hypofractio-
for lesions < 20, 21–30, and 31–40 mm were 24, 18, and 15 Gy, nated treatment provided equivalent progression-free survival
respectively.35 Further dose refinement was suggested by She- at 6 months and 1 year, but was associated with a favorable tox-
hata et al,36 who observed that for patients with metastases < icity profile (5% vs. 17%, p = 0.05). Further work in a prospective
20 mm in diameter treated with WBRT, local control improve- randomized fashion is required to establish the optimal dose
ments were not seen with doses > 20 Gy, with worsening neuro- for HSRT, 6 Gy × 5 is the lead author’s routine practice.
toxicity at the expense of further dose escalation. As such, ▶ Fig. 28.1 illustrates a complex case where an intact large le-
20 Gy has been established as a standard dose for small lesions sion and surgical cavity were treated at the same time focally
(≤ 2 cm). with 6 Gy x 5 fractions.

Fig. 28.1 Hypofractionated stereotactic radiotherapy (HSRT) for large intact brain metastases. A complex case with an intact large lesion that is deep
planned for HSRT with 6 Gy × 5 fractions at the maximum diameter 2.2 cm. The smaller lesion (arrow) will be treated with stereotactic radiosurgery; it
is too small (0.6 cm) for multileaf collimator-based linear accelerator HSRT.

239
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

Fig. 28.2 Patient treatment algorithm for patients presenting with one to four metastases. Option of therapy: *Authors’ preferred option; #Resection
of symptomatic lesion(s) followed by SRS to remaining metastases; †HSRT to large lesion and SRS to remaining metastases; +HSRT to postoperative
cavity; ^For Karnofsky Performance Score (KPS) > 70 suggest WBRT or best supportive care. SRS, stereotactic radiosurgery; HSRT, hypofractionated
radiation therapy; OBS, observation; WBRT, whole-brain radiation therapy.

28.7 Complications selection. There can be serious side effects with this medication,
including a greater risk of wound complications should surgery
28.7.1 Radiation Necrosis be required urgently while on the drug.44,45,46

Radiation necrosis is a serious side effect associated with intra-


cranial SRS. Symptomatic radiation necrosis rates are typically 28.7.2 Neurocognitive Dysfunction and
in the order of 5%.36 However, factors such as the treatment
dose, volume, and location can modify the risk profile.41 Diag-
the Effect of WBRT
nosing radiation necrosis from tumor recurrence is a major As mentioned previously, Chang et al reported on the pri-
challenge given similar radiological appearances. The gold mary end point of neurocognition changes at 4 months in
standard is biopsy, although noninvasive modalities such as patients randomized to SRS alone versus WBRT plus SRS. 7
perfusion magnetic resonance imaging (MRI), C-methionine The study concluded superior outcomes for memory func-
positron emission tomography (PET), and MR spectroscopy, tioning with SRS alone. Although there were several limita-
have been investigated as alternatives to tissue confirmation, tions in this study that could have also contributed to the
but are not sensitive or specific enough to be reliable for defini- poorer neurocognitive outcomes in the WBRT group, such as
tive clinical diagnosis.42 Management options include observa- greater extracranial and intracranial disease burden and
tion if asymptomatic, steroids for symptomatic lesions, surgical more deaths at the 4-month interval, this study was the first
resection, and now bevacizumab.43 A small randomized study to provide randomized evidence that WBRT independently
showed significant improvements in patients with radiation and adversely impacts neurocognition. The findings of the
necrosis treated with four cycles of bevacizumab; however, trial by Chang et al were corroborated by the recent RTOG
further research is required before we understand patient 0214 trial, which questioned the role of prophylactic cranial

240
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

WBRT (PC-WBRT) in patients with non–small cell lung can- [2] Sperduto PW, Kased N, Roberge D, et al. Summary report on the graded prog-
nostic assessment: an accurate and facile diagnosis-specific tool to estimate
cer.47 Based on HVLT assessments, PC-WBRT was shown to
survival for patients with brain metastases. J Clin Oncol 2012; 30: 419–425
adversely impact memory both at 6 months and 1 year [3] Patchell RA, Tibbs PA, Walsh JW, et al. A randomized trial of surgery in the
despite a reduction in the risk of development of brain meta- treatment of single metastases to the brain. N Engl J Med 1990; 322: 494–
stases in the PC-WBRT group. From the EORTC study that uti- 500
lized the validated EORTC Quality of Life Questionnaire C30 [4] Patchell RA, Tibbs PA, Regine WF, et al. Postoperative radiotherapy in the
treatment of single metastases to the brain: a randomized trial. JAMA 1998;
and the EORTC QLQ Brain Cancer Module, we learned that
280: 1485–1489
quality of life (QoL) was impaired with the use of WBRT in [5] Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant whole-brain radiotherapy
patients following either surgery or SRS. 23 Although many versus observation after radiosurgery or surgical resection of one to three
QoL measures in patients who received WBRT normalized, cerebral metastases: results of the EORTC 22952–26001 study. J Clin Oncol
2011; 29: 134–141
the cognitive decline domain remained clinically worse up to
[6] Pulenzas N, Khan L, Tsao M, et al. Fatigue scores in patients with brain meta-
1-year posttreatment.
stases receiving whole brain radiotherapy. Support Care Cancer 2014; 22:
Attempts to mitigate the deleterious effect of WBRT include 1757–1763
hippocampus sparing, which has been shown in some clinical [7] Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain
models to decrease the risk of memory loss. Early reports of the metastases treated with radiosurgery or radiosurgery plus whole-brain ir-
radiation: a randomised controlled trial. Lancet Oncol 2009; 10: 1037–
RTOG 0933 observed a decreased rate in memory loss at 4
1044
months compared with historical controls.48 These data show [8] Roberge D, Souhami L. Tumor bed radiosurgery following resection of brain
that sparing a key center within the brain associated with mem- metastases: a review. Technol Cancer Res Treat 2010; 9: 597–602
ory can yield clinical improvements. Memantine was another [9] Brennan C, Yang TJ, Hilden P, et al. A phase 2 trial of stereotactic radiosurgery
strategy tested to reduce the cognitive dysfunction associated boost after surgical resection for brain metastases. Int J Radiat Oncol Biol Phys
2014; 88: 130–136
with WBRT.49 The rate of cognitive failure, defined as the
[10] Al-Omair A, Soliman H, Xu W, et al. Hypofractionated stereotactic radiother-
change in the Hopkins Verbal Learning Test—Revised for apy in five daily fractions for post-operative surgical cavities in brain meta-
Delayed Recall at 24-week posttherapy, trended toward a signif- stases patients with and without prior whole brain radiation. Technol Cancer
icant improvement with the use of memantine compared with Res Treat 2013; 12: 493–499
[11] Roberge D, Parney I, Brown PD. Radiosurgery to the postoperative surgical
placebo (median decline, 0 vs. −0.9, p = 0.059), with a probabil-
cavity: who needs evidence? Int J Radiat Oncol Biol Phys 2012; 83: 486–493
ity of cognitive function failure at 24 weeks being 53.8% in the [12] Muacevic A, Wowra B, Siefert A, Tonn JC, Steiger HJ, Kreth FW. Microsurgery
memantine arm and 64.9% in the placebo group. These data plus whole brain irradiation versus Gamma Knife surgery alone for treatment
illustrate that although some risk to cognition can be mitigated, of single metastases to the brain: a randomized controlled multicentre phase
the use of WBRT yields significant risk of memory dysfunction. III trial. J Neurooncol 2008; 87: 299–307
[13] Muacevic A, Kreth FW, Horstmann GA, et al. Surgery and radiotherapy com-
The best way to reduce the potential for neurocognitive side
pared with gamma knife radiosurgery in the treatment of solitary cerebral
effects is to minimize the exposure of normal brain parenchy- metastases of small diameter. J Neurosurg 1999; 91: 35–43
ma to radiation; this was the underlying rationale for omission [14] O’Neill BP, Iturria NJ, Link MJ, Pollock BE, Ballman KV, O’Fallon JR. A compari-
of WBRT totally in the three phase III randomized trials. son of surgical resection and stereotactic radiosurgery in the treatment of
solitary brain metastases. Int J Radiat Oncol Biol Phys 2003; 55: 1169–1176
[15] Schöggl A, Kitz K, Reddy M, et al. Defining the role of stereotactic radiosur-
28.8 Conclusion gery versus microsurgery in the treatment of single brain metastases. Acta
Neurochir (Wien) 2000; 142: 621–626
The impact of increasingly effective systemic and targeted [16] Bindal AK, Bindal RK, Hess KR, et al. Surgery versus radiosurgery in the treat-
therapies has made intracranial control increasingly important ment of brain metastasis. J Neurosurg 1996; 84: 748–754
[17] Nieder C, Nestle U, Walter K, Niewald M, Schnabel K. Dose/effect relation-
for patients with metastatic disease. Stereotactic radiosurgery
ships for brain metastases. J Cancer Res Clin Oncol 1998; 124: 346–350
has evolved as a standard of care in the up-front management [18] Kondziolka D, Patel A, Lunsford LD, Kassam A, Flickinger JC. Stereotactic ra-
of brain metastases as compared with WBRT alone, and as com- diosurgery plus whole brain radiotherapy versus radiotherapy alone for pa-
pared with WBRT plus SRS due to the increasing evidence of tients with multiple brain metastases. Int J Radiat Oncol Biol Phys 1999; 45:
serious toxicities with WBRT. For postoperative patients, there 427–434
[19] Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy
is emerging data on the use of SRS, yielding promising results:
with or without stereotactic radiosurgery boost for patients with one to three
It may potentially become the preferred option for this group brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet
of patients. Currently, the N107C trial (NCI-2011–02676, 2004; 363: 1665–1672
NCT01372774), a randomized phase III trial comparing WBRT [20] Tsao M, Xu W, Sahgal A. A meta-analysis evaluating stereotactic radiosurgery,
and SRS in postoperative patients with one to four brain meta- whole-brain radiotherapy, or both for patients presenting with a limited
number of brain metastases. Cancer 2012; 118: 2486–2493
stases, is enrolling patients, and the results of the trial will bet-
[21] Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosurgery plus whole-
ter define the role of SRS in this setting. Ultimately, WBRT will brain radiation therapy vs stereotactic radiosurgery alone for treatment of
be reserved for leptomeningeal disease, brain metastases aris- brain metastases: a randomized controlled trial. JAMA 2006; 295: 2483–
ing from histologies such as small cell lung carcinoma and lym- 2491
[22] Meyers CA, Brown PD. Role and relevance of neurocognitive assessment in
phoma, or miliary dissemination such that SRS is not feasible.
clinical trials of patients with CNS tumors. J Clin Oncol 2006; 24: 1305–
Our proposed patient flow algorithm is found in ▶ Fig. 28.2 for 1309
patients with one to four metastases. [23] Soffietti R, Kocher M, Abacioglu UM, et al. A European Organisation for Re-
search and Treatment of Cancer phase III trial of adjuvant whole-brain radio-

References therapy versus observation in patients with one to three brain metastases
from solid tumors after surgical resection or radiosurgery: quality-of-life re-
sults. J Clin Oncol 2013; 31: 65–72
[1] Gaspar L, Scott C, Rotman M, et al. Recursive partitioning analysis (RPA) of
[24] Sahgal A, Ma L, Chang E, et al. Advances in technology for intracranial stereo-
prognostic factors in three Radiation Therapy Oncology Group (RTOG) brain
tactic radiosurgery. Technol Cancer Res Treat 2009; 8: 271–280
metastases trials. Int J Radiat Oncol Biol Phys 1997; 37: 745–751

241
Stereotactic Radiosurgery for the Management of One to Four Brain Metastases

[25] Li G, Ballangrud A, Kuo LC, et al. Motion monitoring for cranial frameless ster- [38] Märtens B, Janssen S, Werner M, et al. Hypofractionated stereotactic radio-
eotactic radiosurgery using video-based three-dimensional optical surface therapy of limited brain metastases: a single-centre individualized treatment
imaging. Med Phys 2011; 38: 3981–3994 approach. BMC Cancer 2012; 12: 497
[26] Ryken TC, Meeks SL, Pennington EC, et al. Initial clinical experience with [39] Eaton BR, Gebhardt B, Prabhu R, Shu HK, Curran WJ Jr, Crocker I. Hypofractio-
frameless stereotactic radiosurgery: analysis of accuracy and feasibility. Int J nated radiosurgery for intact or resected brain metastases: defining the opti-
Radiat Oncol Biol Phys 2001; 51: 1152–1158 mal dose and fractionation. Radiat Oncol 2013; 8: 135
[27] Sahgal A, Ma L, Tsao M, Larson DA. Gamma Knife radiosurgery. In: Vaezy S, [40] Kim YJ, Cho KH, Kim JY, et al. Single-dose versus fractionated stereotactic
Zderic V, eds. Image-Guided Therapy Systems. 1st ed. Norwood, MA: Artech radiotherapy for brain metastases. Int J Radiat Oncol Biol Phys 2011; 81:
House; 2009 483–489
[28] Ruschin M, Nayebi N, Carlsson P, et al. Performance of a novel reposition- [41] Flickinger JC, Kondziolka D, Lunsford LD, et al. Arteriovenous Malformation
ing head frame for gamma knife perfexion and image-guided linac-based Radiosurgery Study Group. Development of a model to predict permanent
intracranial stereotactic radiotherapy. Int J Radiat Oncol Biol Phys 2010; symptomatic postradiosurgery injury for arteriovenous malformation pa-
78: 306–313 tients. Int J Radiat Oncol Biol Phys 2000; 46: 1143–1148
[29] Ruschin M, Komljenovic PT, Ansell S, et al. Cone beam computed tomography [42] Chao ST, Ahluwalia MS, Barnett GH, et al. Challenges with the diagnosis and
image guidance system for a dedicated intracranial radiosurgery treatment treatment of cerebral radiation necrosis. Int J Radiat Oncol Biol Phys 2013;
unit. Int J Radiat Oncol Biol Phys 2013; 85: 243–250 87: 449–457
[30] Murphy MJ. Intrafraction geometric uncertainties in frameless image-guided [43] McPherson CM, Warnick RE. Results of contemporary surgical management
radiosurgery. Int J Radiat Oncol Biol Phys 2009; 73: 1364–1368 of radiation necrosis using frameless stereotaxis and intraoperative magnetic
[31] Dieterich S, Gibbs IC. The CyberKnife in clinical use: current roles, future ex- resonance imaging. J Neurooncol 2004; 68: 41–47
pectations. Front Radiat Ther Oncol 2011; 43: 181–194 [44] Gonzalez J, Kumar AJ, Conrad CA, Levin VA. Effect of bevacizumab on radia-
[32] Ma L, Petti P, Wang B, et al. Apparatus dependence of normal brain tissue tion necrosis of the brain. Int J Radiat Oncol Biol Phys 2007; 67: 323–326
dose in stereotactic radiosurgery for multiple brain metastases. J Neurosurg [45] Levin VA, Bidaut L, Hou P, et al. Randomized double-blind placebo-controlled
2011; 114: 1580–1584 trial of bevacizumab therapy for radiation necrosis of the central nervous sys-
[33] Sahgal A, Barani IJ, Novotny J Jr, et al. Prescription dose guideline based on tem. Int J Radiat Oncol Biol Phys 2011; 79: 1487–1495
physical criterion for multiple metastatic brain tumors treated with stereo- [46] Torcuator R, Zuniga R, Mohan YS, et al. Initial experience with bevacizumab
tactic radiosurgery. Int J Radiat Oncol Biol Phys 2010; 78: 605–608 treatment for biopsy confirmed cerebral radiation necrosis. J Neurooncol
[34] Ma L, Sahgal A, Descovich M, et al. Equivalence in dose fall-off for isocentric 2009; 94: 63–68
and nonisocentric intracranial treatment modalities and its impact on dose [47] Sun A, Bae K, Gore EM, et al. Phase III trial of prophylactic cranial irradiation
fractionation schemes. Int J Radiat Oncol Biol Phys 2010; 76: 943–948 compared with observation in patients with locally advanced non-small-cell
[35] Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treatment of lung cancer: neurocognitive and quality-of-life analysis. J Clin Oncol 2011;
recurrent previously irradiated primary brain tumors and brain metasta- 29: 279–286
ses: final report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys [48] Gondi V, Mehta MP, Pugh S, et al. Memory preservation with conformal
2000; 47: 291–298 avoidance of the hippocampus during whole brain radiation therapy for pa-
[36] Shehata MK, Young B, Reid B, et al. Stereotatic radiosurgery of 468 brain tients with brain metastases: primary endpoint results of RTOG 0933. Int J
metastases < or =2 cm: implications for SRS dose and whole brain radiation Radiat Oncol Biol Phys 2013
therapy. Int J Radiat Oncol Biol Phys 2004; 59: 87–93 [49] Brown PD, Pugh S, Laack NN, et al. Radiation Therapy Oncology Group
[37] Fahrig A, Ganslandt O, Lambrecht U, et al. Hypofractionated stereotactic ra- (RTOG). Memantine for the prevention of cognitive dysfunction in patients
diotherapy for brain metastases—results from three different dose concepts. receiving whole-brain radiotherapy: a randomized, double-blind, placebo-
Strahlenther Onkol 2007; 183: 625–630 controlled trial. Neuro-oncol 2013; 15: 1429–1437

242
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

29 Stereotactic Radiosurgery for the Management of Five


or More Brain Metastases
Christopher L. Tinkle, Steve Braunstein, Penny K. Sneed, Igor J. Barani, and David Larson

above, other important end points including neurocognitive


Key Points function, health-related quality of life, functional independ-
ence, and cost-effectiveness must be considered. In an attempt
● No prospective randomized trials have been reported to date to address many of these critical questions, the North American
comparing stereotactic radiosurgery versus whole-brain ra- Gamma Knife Consortium (NAGKC) is opening the NAGKC 12–
diotherapy in patients with five or more brain metastases, 01 study,7 a randomized controlled trial comparing SRS with
though at least one such trial is accruing patients. WBRT for patients with ≥ five brain metastases, with neurocog-
● Numerous retrospective studies suggest that patients with a nitive outcomes and tumor control as primary end points. Until
single brain metastasis live longer than those with multiple such studies mature, however, physicians and patients must
brain metastases, but that number of brain metastases is not rely on the limited available data. What follows is a review of
an important prognostic factor among patients with more many of the published studies on SRS or WBRT for multiple
than one brain metastasis. metastases, a summary of consensus statements that address
● In the well-designed, sufficiently powered, prospective Japa- management of patients with multiple metastases, and a
nese Leksell Gamma Knife Society 0901 trial, patients with 5 description of the toxicities of SRS and WBRT.
to 10 brain metastases fared no worse than those with 2 to 4
brain metastases in all end points analyzed, including overall
survival, intracranial tumor control, neurologic deterioration, 29.2 Treatment Options
and salvage therapy. Traditional cytotoxic chemotherapy has had limited success in
treatment of brain metastases of most histologies, either in
combination with WBRT8 or alone.9 Advances in molecularly
targeted systemic therapy, including tyrosine kinase inhibitors
29.1 Introduction and immune-modulating agents, have led to recent success in
Optimal management of patients with intracranial metastases systemic monotherapy for certain histologies of brain metasta-
remains an area of controversy and active investigation. Histor- ses, albeit most notably in patients with smaller asymptomatic
ically, based largely on limitations of treatment modalities at lesions.10,11 Observation of small asymptomatic brain metasta-
hand, including surgical resection and early applications of ster- ses is also an option.
eotactic radiosurgery (SRS), a patient’s total number of brain Therapeutic approaches for patients with brain metastases
metastases formed an important exclusion criterion for many most commonly include surgical resection, WBRT, and/or SRS.
clinical studies. With this in mind, several phase III randomized Surgical intervention is typically limited to single or, less com-
trials support the use of SRS for the treatment of patients with monly, two to three synchronous resectable brain metastases;
one to four brain metastases.1–5 Studies by Kondziolka et al1 thus, in this chapter we focus on SRS and WBRT, including
and Andrews et al2 demonstrated improved local control with recent strategies to lessen the risk of neurocognitive effects of
the addition of SRS to whole-brain radiotherapy (WBRT), WBRT.12,13
whereas studies by Aoyama et al,3 Chang et al,4 and Kocher et
al5 showed that withholding WBRT does not compromise over- 29.2.1 Stereotactic Radiosurgery for
all survival (OS). However, each of these studies has also shown
significant improvement in both local and distant intracranial
Five or More Brain Metastases
tumor control when SRS is combined with planned WBRT. One of the earlier studies of SRS for multiple metastases was
Nevertheless, the addition of WBRT to SRS has been associated reported by Serizawa et al.14 This was a single-institution retro-
with inferior neurocognitive outcomes in some patients.4 Ulti- spective review of patients treated with Gamma Knife stereo-
mately, results of these seminal studies have led to the general tactic radiosurgery (GKSRS) alone (62 patients) or WBRT alone
acceptance of the use of SRS alone as a primary therapeutic op- (34 patients) from 1990 to 1999 for 1 to 10 brain metastases
tion in the management of selected patients with one to four from non–small cell lung cancer (NSCLC). Surgical resection in
brain metastases. select patients preceded irradiation in both groups. Repeat SRS
Unfortunately, whereas relatively robust class I data exist in to new lesions rather than WBRT was performed in the SRS
patients with one to four brain metastases, management deci- group, whereas WBRT patients received subsequent SRS. Base-
sions for patients with ≥ five brain metastases have largely line characteristics, including number of brain metastases, did
relied on speculation and retrospective data, with the exception not significantly differ. On multivariable analysis, only uncon-
of a recent Japanese multi-institutional prospective observatio- trolled systemic disease, WBRT, and low Karnofsky Perform-
nal study by Yamamoto et al.6 Still lacking are prospective ance Score (KPS) were significantly associated with poor
randomized controlled trials comparing SRS with WBRT for outcome. Interestingly, mean survival time (377 vs. 199 days,
patients with five or more metastases. Importantly, given the p = 0.0158), neurologic survival (p = 0.0237), and OS (p = 0.158)
comparable OS for SRS ± WBRT borne out of the trials described favored the SRS-alone group.

243
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

In a subsequent study, Serizawa et al15 analyzed outcomes of In an initial report by Yamamoto et al20 of 456 patients
GKSRS among 2,390 patients at two institutions in Japan treated from 1991 to 2004 with GKSRS for 1 to 55 brain meta-
treated from 1998 to 2005 with either surgery followed by SRS stases (mean 6; median 2) arising from non–lung cancer pri-
or SRS alone. Prophylactic WBRT was not administered and dis- mary tumors, increasing tumor number was significantly
tant intracranial disease was managed with repeat SRS if the associated with inferior OS on univariable analysis (p = 0.001).
patient’s condition permitted. Roughly 40% of patients at each Up-front surgical resection occurred in 23% of patients, prior
facility had more than four brain metastases, with a mean of 6.2 radiotherapy in 6.4%, and prior surgery and radiotherapy in
and 6.5, respectively. Factors significantly associated with inferi- 3.3%. When median OS times were compared based on number
or OS included active extracranial disease, male gender, and of brain metastases, lesion number appeared to have a signifi-
low initial KPS score (all, p = 0.0001), whereas total number of cant impact on duration of survival: 1 to 4 lesions (7.4 months),
brain metastases (≤ 4 vs. > 4) was not significantly associated 5 to 9 lesions (4.0 months), 10 to 14 lesions (7.4 months), 15 to
with survival. A critical selection factor in this study was the 19 lesions (3.4 months), 20 to 29 lesions (4.9 months), and 30
limitation of the total skull integral dose to 10 to 12 joules (J), a or more lesions (4.6 months); p = 0.0002. However, an impor-
concept put forth to integrate tumor number, tumor size, and tant limitation to this study is the limited numbers of patients
SRS dose to maintain an equivalent threshold dose of a single within each group beyond five brain metastases.
3-Gy fraction of whole-brain irradiation. Below this threshold, An update on this study was reported by Yamamoto et al 21
this group had not observed acute cerebral swelling.15 with 1,676 patients with brain metastases from a variety of pri-
Nam et al16 reported a single institution’s experience with mary tumors treated with GKSRS from 1991 to 2006. The mean
GKSRS for 130 patients with brain metastases of various histol- and median lesion numbers were 7 and 3, respectively, with a
ogies treated from 2001 to 2004, including 84 patients with one range of 1 to 85. In this analysis, patients again were grouped
to three brain metastases and 46 with four or more. The major- by number of brain metastases, yet included comparisons of
ity of these patients also received WBRT (73%). Although median survival times between patients with 1 versus 2 lesions,
median OS was significantly longer for patients with one to 2 versus 3 lesions, and up to 14 versus 15 lesions. Among the
three metastases compared with those with four or more (48 14 comparison pairs, only one versus two lesions showed a sig-
vs. 26 weeks, p = 0.005), the number of metastases did not sig- nificant median survival difference (10.2 vs. 7.2 months,
nificantly predict for improved survival. Rather, recursive parti- p = 0.0002), whereas the remaining pairwise comparisons did
tioning analysis (RPA)17 class was the only variable found to be not differ significantly. That said, there was a significant differ-
significantly associated with survival on multivariable analysis, ence in median survival comparing patients with ≤ four versus ≥
leading to the authors’ recommendation that RPA, rather than five brain metastases (8.4 vs. 5.3 months, respectively,
lesion number, be used to restrict the use of SRS monotherapy. p < 0.0001). Additionally, female patients lived longer than male
A study from the University of Pittsburgh (Pittsburgh, PA) by patients (9.0 vs. 5.9 months, p < 0.0001).
Bhatnagar et al18 (▶ Table 29.1) reported outcomes for 205 A more recent single-institution retrospective case-matched
patients treated from 1995 to 2005 with SRS to 4 to 18 brain study by Yamamoto et al22 (▶ Table 29.1) was undertaken to
metastases of various histologies; 64% of patients had ≥ five meta- determine the outcomes of patients with ≤ four versus ≥ five
stases. The majority of patients also received WBRT, 46% concur- brain metastases treated with GKSRS from 1998 to 2011. Prior
rently with SRS and 38% after progression following SRS. Median surgical resection was performed in 17% of patients, and 4.2%
OS for patients meeting RPA class I, II, and III criteria were 18, 9, had prior WBRT. With 548 patients in each group, median sur-
and 3 months, respectively, comparing favorably with historical vival time was statistically longer in patients with ≤ four brain
results with WBRT alone (7, 4, and 2 months, respectively). Multi- metastases compared with those with ≥ five, 7.9 versus 7.0
variable analysis revealed total treatment volume as a significant months, respectively, p = 0.01. However, with overlapping 95%
variable associated with improved one-year local control confidence intervals of 7.0 to 8.9 months versus 6.2 to 7.8
(p = 0.002), whereas total treatment volume (p = 0.002), age months and a hazard ratio of 1.176, the significance is question-
(p = 0.005), and RPA class (p = 0.009) were significantly associated able. Additionally, as noted by the authors, the difference of 27
with improved survival. Total number of treated brain metastases days is likely not clinically meaningful. Furthermore, there was
was not significantly prognostic for local control nor OS (p = 0.91 no significant difference between the two groups in freedom
and 0.333, respectively). Thus, total treatment volume rather than from neurologic death, and through the use of competing risk
number of lesions was posited by this group as an important analysis, there were no significant differences in local control,
selection factor for the use of SRS alone. use of repeat SRS, or major SRS-related complications.
Park et al19 reported their institutional outcomes for 33 Another large retrospective study was conducted by Karlsson
patients with 2 to 20 NSCLC brain metastases < 3 cm diameter et al23 (▶ Table 29.1) of 1,855 patients from four institutions
treated from 2005 to 2006 with either GKSRS (14 patients) or treated with GKSRS from 1975 to 2007. Incomplete records lim-
WBRT (19 patients) based on physician and patient preference. ited analysis of prior treatment, but at least 13% received prior
Overall survival was significantly increased for patients treated WBRT and 22% underwent repeat SRS. Nearly half of the
with SRS compared with those treated with WBRT (p = 0.04), patients (46%) had one brain metastasis, 22% had two, 19% had
with 1-year survival rates of 47.7% versus 10.5%, respectively. three to four, 10% had five to eight, and 3% > eight brain meta-
Given the small sample size, analysis of prognostic factors was stases. There was significantly longer median survival time in
limited to univariable analysis, which revealed control of pri- patients with single versus multiple lesions, 7.5 versus 6.1
mary tumor site (p = 0.03) and use of SRS (p = 0.04) to be signifi- months, p < 0.0001, as well as for patients with controlled ver-
cantly associated with superior OS, whereas number of brain sus uncontrolled primary disease, 9.8 versus 5.2 months,
metastases (< 10 vs. ≥ 10) was not (p = 0.33). p < 0.0001, and patients < 60 versus ≥ 60 years of age, 8.3 months

244
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

Table 29.1 Selected studies of outcomes of stereotactic radiosurgery for multiple brain metastases
Study Period N Outcome(s) No. metastases Key findings
[PubMed ID]
Study type

Bhatnagar et al, 200618 1995–2005 205 OS 4–18 Median OS 8.0 months; 71% 1-year LC
[16338097] Treatment volume, age, RPA class,
Retrospective marginal dose associated with OS

Yamamoto et al, 201322 1998–2011 1,096 OS 1–4 vs. ≥ 5 Median OS 7.0 months for ≥ 5 mets vs.
[23600938] LC 7.9 months for 1–4 mets, p = 0.01
Case-matched ND No difference in LC, ND, toxicity

Karlsson et al, 200923 1975–2007 1,885 OS 1–8 + Median OS 6.1 months for ≥ 2 mets
[19199505] Age and primary tumor control asso-
Retrospective ciated with OS

Chang et al, 201024 2005–2008 323 OS 1–5 vs. 6–10 vs. 11–15 vs. > 15 Median OS 10 months for 6–10 mets
[21121789] LC No differences in OS, LC; increased
Retrospective distant progression for > 15 mets

Hunter et al, 201225 1995–2008 64 OS ≥5 Median OS 7.5 months


[22209150] KPS associated with OS
Retrospective

Raldow et al, 201226 2000–2010 103 OS 5–9 (84 patients) vs. ≥ 10 Median OS 7.6 months for 5–9 mets
[22706180] RFS (19 patients) Higher KPS associated with better OS
Retrospective

Serizawa et al, 201033 1998–2009 778 OS 1–10 (5–10 in 215 patients) Median OS 7.3 months for 5–10 mets
[20411300] NS Active systemic disease, KPS < 70, and
Retrospective NLFS male gender associated with poorer
QS OS

Yamamoto et al, 20146 2009–2012 1,194 OS 2–4 vs. 5–10 Median OS 10.8 months for 5–10 mets
[24621620] Toxicity Risk of AE 3% for 5–10 mets
Prospective trial JLGK0901 NCF Noninferior OS and toxicity for 5–10
vs. 2–4 mets; NCF analysis pending

Abbreviations: AE, adverse events; KPS, Karnofsky Performance Status; LC, local control; mets, metastases; NCF, neurocognitive function; ND, neurologic
deterioration; NLFS, new-lesion-free survival; NS, neurologic survival (survival with KPS ≥ 70); OS, overall survival; QS, qualitative survival; RFS,
recurrence-free survival; RPA, recursive partitioning analysis.

versus 5.3 months, p < 0.0001. However, primary disease con- However, median progression-free survival and time to
trol appeared to be the dominant prognostic factor, as signifi- distant brain progression were significantly shorter for
cant differences in OS between single and multiple brain patients with > 15 brain metastases, p = 0.03 and p = 0.014,
metastases disappeared when patients with controlled or respectively.
uncontrolled primary disease were analyzed separately. Addi- Hunter et al25 (▶ Table 29.1) studied 64 patients with 5 to 10
tionally, there were no significant differences in median surviv- brain metastases (median 6) treated with SRS from 1995 to
al between groups of patients with two versus three to four 2008; 63% had received prior WBRT, 14% had concurrent WBRT,
versus five to eight versus > eight lesions, or between patients and 23% had SRS alone. The median survival after SRS was 7.5
with < four versus ≥ five lesions. The authors concluded that pa- months overall, 6.6 months in patients with ≤ eight brain meta-
tient age and primary disease status should be used as limiting stases, and 9.9 months in those with > eight metastases (not sig-
parameters for treatment with SRS, rather than total number of nificant). However, higher KPS (≥ 80, p = 0.02) and prior as
brain metastases. opposed to concurrent radiotherapy (p = 0.03) were found to be
Chang et al24 (▶ Table 29.1) evaluated 323 patients treated significantly associated with improved OS on multivariable
from 2005 to 2008 at a single institution with SRS. Fourteen analysis.
patients with ≥ 15 brain metastases received prior WBRT. A similar analysis was done by Raldow et al26 (▶ Table 29.1)
Patients were retrospectively grouped based on number of for 103 patients with ≥ five brain metastases treated from 2000
treated lesions: 1 to 5 (67%), 6 to 10 (18%), 11 to 15 (5%), to 2010 by SRS. Eighty-two percent of patients had 5 to 9 brain
and > 15 (10%). Overall survival times after SRS were 10, 10, metastases, whereas 18% had 10 or more; 59% had prior treat-
13, and 8 months, respectively, and did not differ signifi- ment (WBRT, 33%; SRS, 12%; or both, 15%). The median survival
cantly among groups, p = 0.554. Additionally, local tumor con- times were 7.6 versus 8.3 months for patients with 5 to 9 ver-
trol also did not differ significantly among the groups of sus ≥ 10 brain metastases, p = 0.38. Karnofsky Performance
patients, nor did radiographic changes on interval imaging. Score was the only variable significantly associated with

245
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

survival time on multivariable analysis; number and total vol- Prior to completion of the prospective trial, Serizawa per-
ume of brain metastases were not significant. formed two retrospective studies incorporating the eligibility
In an analysis of prognostic factors after salvage SRS, Cabal- criteria of JLGK0901. One study32 examined results of SRS as
lero et al27 retrospectively reviewed 310 patients treated sole treatment for 1,508 patients with 1 to 10 brain metastases
with GKSRS for new, progressive, or recurrent brain metasta- treated from 1998 to 2007 with GKSRS at two institutions in
ses after WBRT. The median number of treated lesions was 4, Japan. The median survival times by number of metastases
with a range of 1 to 35, and with more than half of patients were 11.9 months for group A (1 tumor), 8.2 months for group
(53%) having 4 to 35 lesions. Median survival from SRS was B (2–4 tumors), and 7.4 months for group C (5–10 tumors).
8.2 months, significantly longer in patients with a single There were significant differences in median survival for group
brain metastasis compared with those with multiple meta- A versus B (p < 0.0001) and group B versus C (p = 0.0312). How-
stases, 12.0 versus 7.9 months, p = 0.001. Yet, there was no ever, on multivariable analysis no significant difference in OS
relationship between number of brain metastases and OS between groups B and C was found (p = 0.103; hazard ratio
among patients with multiple lesions. Favorable prognostic [HR] = 1.124, 95% confidence interval [CI] 0.999–1.265). The
factors on multivariable analysis varied by primary disease upper limit of the 95% CI of 1.265 was within the predeter-
site. The nearly 8-month median survival time led the mined threshold of 1.3 corresponding to a noninferiority mar-
authors to conclude that salvage SRS is a reasonable treat- gin of 7.5% employed in the JLGK0901 study. Prognostic factors
ment option, with no evidence for a threshold related to associated with inferior outcomes included male gender
brain metastases number. (p < 0.0001), lung primary (vs. breast, p = 0.0047), higher RPA
Several studies have restricted analysis to patients with ≥ 10 classification (p < 0.0001), and multiple brain metastases (group
brain metastases. Suzuki et al28 reported on 24 patients without A vs. B, p < 0.0001).
prior WBRT with 10 to 47 lesions (mean 20) treated from 1998 In a second study (▶ Table 29.1),33 again employing JLGK0901
to 2000 with GKSRS. With mean follow-up of only 13 weeks, eligibility criteria, 778 patients were divided into groups based
the Kaplan-Meier estimate of OS at 24 weeks was 49.3%, and on number of brain metastases: A (1), B (2), C (3–4), D (5–6),
the mean KPS score improved significantly after completion of and group E (7–10). Mean survival times ranged from 10.7 to
SRS (p < 0.05). In an effort to assess cumulative whole-brain 7.1 months, yet were not significantly different between any
dose in patients with a large number of brain metastases, two groups. Additional outcomes of freedom from neurologic
Yamamoto et al29 reported on 80 patients with a median of 17 death and survival with KPS ≥ 70 at year were also not signifi-
lesions (range 10–43) treated with GKSRS from 1998 to 2000. cantly different between any two groups; however, 1-year dis-
With a median tumor volume of 8.02 mL and median pre- tant intracranial control was significantly worse for group B
scribed dose of 20 Gy, the median cumulative dose to the whole versus A (p = 0.0003) and for group C versus B (p = 0.0047).
brain (excluding the tumor volume) was 4.71 Gy (range 2.16– In 2014, Yamamoto et al6 (▶ Table 29.1) reported the results
8.51 Gy), a dose the authors concluded was below the threshold pertaining to the primary end point of OS of the JLGK0901
level of normal brain tissue necrosis. study; assessment of secondary end points of SRS-induced
Several groups have performed retrospective reviews to elu- complications and neurocognitive function will continue into
cidate potential prognostic factors of OS in patients with 10 or 2014. From 2009 to 2012, 1,194 patients were enrolled. Median
more brain metastases treated with SRS. Kim et al30 reviewed OS time after SRS was 13.9 months in the 455 patients with one
outcomes for 26 patients with 10 to 37 lesions, half of whom tumor, 10.8 months in the 531 patients with 2 to 4 tumors, and
had received prior WBRT, and found NSCLC histology 10.8 months in the 208 patients with 5 to 10 tumors. Overall
(p = 0.007), KPS ≥ 80 (p = 0.029), and controlled primary disease survival did not significantly differ between patients with 2 to 4
(p = 0.02) to be associated with longer survival on univariable vs. 5 to 10 brain metastases, with a HR of 0.97, 95% CI 0.81–
analysis. In a similar analysis by Grandhi et al31 in 61 patients 1.18, p = 0.78, and pnoninferiority < 0.0001. Incidence of neurologic
with a mean of 13.2 lesions, 62% of whom had prior WBRT ± death, neurologic deterioration, local recurrence, development
SRS, prognostic factors on univariable analysis included < 14 of new lesions, and salvage SRS, WBRT, or surgery did not differ
brain metastases, nonmelanoma histology, controlled systemic significantly between the two groups of patients with multiple
disease, KPS ≥ 90, and lower RPA class. Among patients with the tumors. Multivariable analysis showed the presence of a single
favorable factors, median survival was 21 months. Prior WBRT tumor, female gender, age < 65, KPS ≥ 80, stable extracranial dis-
was significantly associated with the development of new ease, and absence of neurologic symptoms at the time of SRS to
adverse radiation effect. significantly impact OS. Preliminary analysis revealed no signif-
In 2009, the Japanese Leksell Gamma Knife (JLGK) Society ini- icant difference in adverse events of any grade comparing
tiated a prospective multi-institutional observational cohort patients with 2 to 4 versus 5 to 10 tumors. Thus, the authors
study, JLGK0901, to assess whether treatment outcomes were conclude that SRS alone for the management of patients with 5
different in patients with 5 to 10 versus 2 to 4 brain metastases to 10 brain metastases is noninferior to SRS alone in those with
treated with SRS, and to determine prognostic factors affecting 2 to 4 metastases.
inferiority or noninferiority of outcomes.6 Results from this
observational study were expected to provide support for a 29.2.2 WBRT for Five or More Brain
future randomized controlled trial testing SRS versus WBRT for
patients with 5 to 10 brain metastases. Eligibility criteria Metastases
included 1 to 10 newly diagnosed brain metastases, largest Despite the fact that WBRT is generally considered the de facto
tumor volume < 10 mL, total brain metastasis volume < 15 mL, treatment of choice for patients with a more extensive intracra-
no cerebrospinal fluid dissemination, and KPS ≥ 70. nial disease burden, there are scant data relating the number of

246
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

brain metastases to outcomes for patients with five or more agreeing that treatment of ≥ five lesions was “reasonable.” In
brain lesions treated with WBRT. One study that did address the 2009 survey in Sendai, Japan, the mean and median number
this topic was reported by Nieder et al34 who analyzed 113 of metastases deemed suitable for SRS alone had increased to
patients with a median of six brain metastases (range 4–50) 11 and 10, and 83% of participants felt that treating ≥ five
treated with WBRT without surgical resection or SRS. The num- lesions was “reasonable.”
ber of brain metastases had no significant influence on OS.
Additionally, although there were too few patients who met
RPA class I criteria, there was no significant difference in OS 29.3 Complications: Toxicities and
between RPA class II and III patients.
Neurocognitive Outcomes
Limited radiation tolerance of brain tissues is well recognized,
29.2.3 Consensus Guidelines with white matter and subcortical structures showing greatest
The majority of current consensus statements regarding ma- sensitivity in a dose-dependent fashion.41 Although the precise
nagement of brain metastases do not specifically address ma- mechanisms of radiation-induced brain injury remain unclear,
nagement of patients with ≥ five metastases, other than by alterations in microvasculature and glial cell–mediated inflam-
extrapolation from previously discussed randomized trials in mation are associated with late radiation toxicity.42 Moreover,
patients with one to four metastases. The European Federation evidence suggests that radiation-sensitive, active neurogenesis
of Neurological Societies produced an evidence-based guide- in adults is essential to hippocampal-dependent memory func-
lines document in 200635 in which, without reference, the tion.43 Recent attempts to improve functional outcomes have
authors claim, “In patients with more than three brain metasta- included hippocampal-sparing WBRT12 and adjuvant pharma-
ses WBRT with hypofractionated regimens is the treatment of cological therapy, such as memantine administration,13 with
choice.” In 2009 (updated in 2011), the American College of promising preliminary results.
Radiology published appropriateness criteria using the formal A strong rationale for use of SRS over WBRT for brain meta-
Delphi consensus process for the management of multiple brain stases is the potential sparing of neurocognitive toxicities such
metastases.36 The report includes five clinical case descriptions, as memory impairment, dementia, and loss of functional inde-
but only one case representing ≥ five metastases. A theoretical pendence. An early study on neurocognition following WBRT
50-year-old patient with NSCLC, KPS 90, controlled primary estimated an 11% incidence of severe late clinical dementia,44
disease, no systemic disease, and six newly diagnosed asympto- and a retrospective series of patients receiving WBRT for vari-
matic brain metastases received a score of 1 (least appropriate) ous intracranial malignancies suggested an incidence of 32%
for SRS alone and a score of 8 (9 most appropriate) for WBRT grade 1 to 3 toxicity at one-year post-WBRT at a median dose of
alone. Although clinical evidence was not presented, the num- 30 Gy in 10 fractions.41 A more recent meta-analysis highlights
ber of lesions and lack of evidence for SRS was used to justify the emergence of early neurocognitive impairment, approach-
these scores. ing 50% as early as 3 months following WBRT.45 Notably, a
The American Association of Neurological Surgeons and Con- challenge to accurate assessment of radiation-related neurocog-
gress of Neurosurgeons reviewed 11 observational studies that nitive decline is the confounding effects of intracranial disease
support the notion that SRS alone may provide equivalent sur- burden and progression46 and systemic therapy. Yet, imaging
vival rates compared with WBRT in combination with SRS.37 approaches reveal incidence of leukoencephalopathy ranging
The report, published in 2011, acknowledges the risk of poten- between 34 and 83% at ≥ 6 months after WBRT for brain meta-
tial neurocognitive deficits with the use of WBRT; however, ma- stases.47,48
nagement of multiple metastases is not specifically addressed. The limited treatment volumes of SRS as compared with
The American Society for Radiation Oncology (ASTRO) evi- WBRT hold the potential to reduce late neurocognitive
dence-based guidelines on the role of radiation for newly diag- impairment. Early trials including SRS-only brain metastasis
nosed brain metastases were published in 2012,38 and although treatment arms showed no overt evidence of neurologic toxic-
not directly evaluating management of multiple metastases, the ity.1 Aoyama et al3 compared WBRT + SRS versus SRS alone for
document does state, “It is unknown if there is a cutoff for the one to four brain metastases and performed pre- and post-
maximum number of targets appropriate for SRS. Total target treatment Mini-Mental Status Examination (MMSE). Neuro-
volume as well as number of targets may be important for cognitive outcomes were comparable at 1 year, but there was
safety and efficacy.” Finally, the current National Comprehen- a trend toward improved preservation of MMSE score over
sive Cancer Network (NCCN) guidelines suggest that WBRT extended follow-up (22.5% vs. 42.6%, at 3 years, respectively).
alone is the only appropriate therapeutic intervention for The more recent randomized controlled study by Chang et al 4
patients with > three newly diagnosed brain metastases. 39 comparing WBRT + SRS versus SRS alone was designed with
Although there appears to be reluctance to recommend SRS neurocognitive function as the primary end point, and
for > three to four brain metastases in the consensus guidelines employed a wide battery of neurocognitive assessments. The
described above, this does not appear to be the case among study was closed early due to significantly diminished neuro-
practitioners of SRS.40 Physician surveys at two International cognitive function after WBRT + SRS versus SRS alone 4-month
Stereotactic Radiosurgery Society meetings were conducted to posttreatment (52% vs. 24% decline in immediate verbal recall,
assess acceptance of treating various numbers of brain metasta- respectively). Furthermore, newer series have also reported
ses. In the 2007 survey in San Francisco, the mean and median significantly decreased incidence of leukoencephalopathy on
numbers of metastases physicians would consider treating with imaging after SRS alone vs. WBRT + SRS for one to two
SRS alone were 6.7 and 5, respectively, with 55% of respondents lesions.49

247
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

The first Radiation Therapy Oncology Group (RTOG) SRS trial, precision than the standard MMSE and Hopkins Verbal Learn-
90–05, was a dose-escalation study in recurrent brain tumors 50; ing Test.
64% of patients had brain metastases, of which 22% had multi-
ple brain lesions, although only the dominant lesion received
SRS. There was no evidence of dose-limiting toxicity up to 29.4 Summary
24 Gy for lesions ≤ 20 mm, but treatment of larger lesions (21–
30 and 31–40 mm) caused unacceptable toxicity above 18 Gy Physicians who have traditionally favored WBRT for patients
and 15 Gy, respectively, largely related to radionecrosis and with multiple brain metastases often argue that comprehensive
brain edema at a median of 4.5 to 5.0 months. intracranial radiation is necessary to treat micrometastases,
Of concern, SRS of multiple brain metastases can result in thereby reducing the development of new intracranial metasta-
larger brain volume exposed to both high dose and peripheral ses that could result in high rates of clinical neurologic
dose51 with consequent increased risk of brain radiation decline.55 Supporters of SRS alone often counterargue that the
injury.52 Several reports of SRS for large numbers of metastases neurocognitive effects of WBRT are very real, and that salvage
have shown low cumulative whole-brain radiation dose. An 80- SRS, although more frequently needed in patients initially
patient series of ≥ 10 SRS-treated brain metastases showed treated with SRS alone, is safe, efficient, and effective.27 Addi-
median cumulative whole-brain irradiation dose of 4.71 Gy tional arguments favoring up-front SRS, often invoked by refer-
(range 2.16–8.51 Gy), with no acute or late radiation-related ring physicians and patients alike, is the convenient single-day
toxicities reported. Moreover, a subset of these patients who treatment with minimal interruption in systemic therapy.
had subsequent SRS likewise did not experience any ensuing As outlined in this chapter, despite the vast heterogeneity of
toxicity.29 the retrospective studies described, the majority of reports do
In the large series reported by Serizawa et al, 15 of 2,533 not identify the total number of brain metastases beyond a sin-
patients treated at two major Japanese centers for an average of gle metastasis to be significantly associated with either local
6.2 ± 7.6 and 6.5 ± 9.3 brain metastases, respectively, there were control or OS. Rather, young age; higher KPS; controlled pri-
no severe acute toxicities with total skull integrated dose of less mary, absent, or controlled systemic disease; and smaller total
than 10 to 12 J, equivalent to approximately 3 Gy of WBRT or a volume of brain metastases appear to be important prognostic
15-mL treatment volume. However, multiple studies of SRS factors. Additionally, in the well-designed, sufficiently powered,
treatment of intracranial brain lesions, including metastases, prospective JLGK 0901 trial, patients with 5 to 10 brain meta-
have shown rates of radionecrosis up to 50% depending upon stases with total volume < 15 mL fared no worse than patients
dose, volume, and tumor location.53,54 As reported by Minniti et with 2 to 4 in all end points analyzed, including OS, intracranial
al,54 10-Gy volume > 12.6 mL 12-Gy volume > 10.9 mL was asso- tumor control, neurologic deterioration, and salvage therapy.
ciated with a risk of radionecrosis of 47%. However, only a small Thus, although we await class I evidence on survival, neurocog-
fraction of patients with evidence of radionecrosis on interval nitive function, health-related quality of life, functional inde-
imaging were symptomatic and severe neurologic complica- pendence, and cost-effectiveness of SRS versus WBRT to help
tions were rare. guide treatment decision making for patients with five or more
There are limited data with regard to neurocognitive out- brain metastases, it appears that, when coupled with close
comes following SRS treatment of patients specifically with > active surveillance measures, SRS may be considered a viable
four brain metastases. Recent reports comparing SRS treatment therapeutic option for this important and growing patient
of ≤ four versus > four lesions indicate similar limited treatment- population.56
related toxicities. For example, the case-matched retrospective
study by Yamamoto et al22 comparing outcomes of SRS treat-
ment of patients with one to four versus ≥ five brain metastases
References
revealed equivalence in neurologic deterioration (13.1% vs. [1] Kondziolka D, Patel A, Lunsford LD, Kassam A, Flickinger JC. Stereotactic ra-
9.7%) and treatment-related complications (2.7% vs. 2.0%). diosurgery plus whole brain radiotherapy versus radiotherapy alone for pa-
tients with multiple brain metastases. Int J Radiat Oncol Biol Phys 1999; 45:
The JLGK0901 prospective multi-institutional observational
427–434
study also stratified SRS outcomes by number of brain metasta- [2] Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy
ses.6 Overall rate of SRS-related adverse events was 9% (101/ with or without stereotactic radiosurgery boost for patients with one to three
1,194 patients). Analysis demonstrated no relationship between brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet
2004; 363: 1665–1672
the number of tumors treated and any-grade toxicity. More-
[3] Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosurgery plus
over, high-grade toxicity (grade 3–4) was present at 3% for whole-brain radiation therapy vs stereotactic radiosurgery alone for
patients with 5 to 10 tumors, which was not significantly differ- treatment of brain metastases: a randomized controlled trial. JAMA
ent from those with one (2%) or two to four (2%) tumors. Main- 2006; 295: 2483–2491
tenance of neurocognitive function at up to 36 months post- [4] Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain
metastases treated with radiosurgery or radiosurgery plus whole-brain irra-
SRS was similar among those with 1, 2 to 4, and 5 to 10 tumors
diation: a randomised controlled trial. Lancet Oncol 2009; 10: 1037–1044
at 88 to 100%. [5] Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant whole-brain radiotherapy
Finally, the recently opened North American Gamma Knife versus observation after radiosurgery or surgical resection of one to three
Consortium randomized controlled trial (NAGKC 12–01),7 a cerebral metastases: results of the EORTC 22952–26001 study. J Clin Oncol
comparison of WBRT versus SRS, is specifically examining neu- 2011; 29: 134–141
[6] Yamamoto M, Serizawa T, Shuto T, et al. Stereotactic radiosurgery for patients
rocognitive outcomes in patients with 5 to 10 brain metastases
with multiple brain metastases (JLGK0901): a multi-institutional prospective
using a battery of neurocognitive assessment with greater observational study. Lancet Oncol 2014; 15: 387–395

248
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

[7] North American Gamma-knife Consortium. NAGKC Newsletter Fall. 2012. [29] Yamamoto M, Ide M, Nishio Si, Urakawa Y. Gamma Knife radiosurgery for nu-
Available at: http://www.nagkc.com/news/newsletter/2012–05-spring.pdf. merous brain metastases: is this a safe treatment? Int J Radiat Oncol Biol Phys
Accessed May 7, 2014 2002; 53: 1279–1283
[8] Neuhaus T, Ko Y, Muller RP, et al. A phase III trial of topotecan and whole [30] Kim CH, Im YS, Nam DH, Park K, Kim JH, Lee JI. Gamma knife radiosurgery for
brain radiation therapy for patients with CNS-metastases due to lung cancer. ten or more brain metastases. J Korean Neurosurg Soc 2008; 44: 358–363
Br J Cancer 2009; 100: 291–297 [31] Grandhi R, Kondziolka D, Panczykowski D, et al. Stereotactic radiosurgery us-
[9] Postmus PE, Haaxma-Reiche H, Smit EF, et al. Treatment of brain metastases ing the Leksell Gamma Knife Perfexion unit in the management of patients
of small-cell lung cancer: comparing teniposide and teniposide with whole- with 10 or more brain metastases. J Neurosurg 2012; 117: 237–245
brain radiotherapy—a phase III study of the European Organization for the [32] Serizawa T, Yamamoto M, Sato Y, et al. Gamma Knife surgery as sole treat-
Research and Treatment of Cancer Lung Cancer Cooperative Group. J Clin On- ment for multiple brain metastases: 2-center retrospective review of 1508
col 2000; 18: 3400–3408 cases meeting the inclusion criteria of the JLGK0901 multi-institutional pro-
[10] Margolin K, Ernstoff MS, Hamid O, et al. Ipilimumab in patients with melano- spective study. J Neurosurg 2010; 113 (Suppl): 48–52
ma and brain metastases: an open-label, phase 2 trial. Lancet Oncol 2012; 13: [33] Serizawa T, Hirai T, Nagano O, et al. Gamma knife surgery for 1–10 brain
459–465 metastases without prophylactic whole-brain radiation therapy: analysis of
[11] Long GV, Trefzer U, Davies MA, et al. Dabrafenib in patients with Val600- cases meeting the Japanese prospective multi-institute study (JLGK0901) in-
Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain clusion criteria. J Neurooncol 2010; 98: 163–167
(BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol 2012; [34] Nieder C, Andratschke N, Grosu AL, Molls M. Recursive partitioning analysis
13: 1087–1095 (RPA) class does not predict survival in patients with four or more brain
[12] Gondi V, Pugh SL, Tome WA, Caine C, Corn B, Kanner A, et al. Preservation metastases. Strahlenther Onkol 2003; 179: 16–20
of memory with conformal avoidance of the hippocampal neural stem-cell [35] Soffietti R, Cornu P, Delattre JY, et al. EFNS Guidelines on diagnosis and treat-
compartment during whole-brain radiotherapy for brain metastases ment of brain metastases: report of an EFNS Task Force. Eur J Neurol 2006;
(RTOG 0933): A phase II multi-institutional trial. J Clin Oncol 2014; 32: 13: 674–681
3810–3816 [36] Videtic GM, Gaspar LE, Aref AM, et al. Expert Panel on Radiation Oncology-
[13] Brown PD, Pugh S, Laack NN, et al. Radiation Therapy Oncology Group Brain Metastases. American College of Radiology appropriateness criteria on
(RTOG). Memantine for the prevention of cognitive dysfunction in patients multiple brain metastases. Int J Radiat Oncol Biol Phys 2009; 75: 961–965
receiving whole-brain radiotherapy: a randomized, double-blind, placebo- [37] Bhangoo SS, Linskey ME, Kalkanis SN, American Association of Neurologic
controlled trial. Neuro-oncol 2013; 15: 1429–1437 Surgeons (AANS). Congress of Neurologic Surgeons (CNS). Evidence-based
[14] Serizawa T, Iuchi T, Ono J, et al. Gamma knife treatment for multiple meta- guidelines for the management of brain metastases. Neurosurg Clin N Am
static brain tumors compared with whole-brain radiation therapy. J Neuro- 2011; 22: 97–104, viii
surg 2000; 93 (Suppl 3): 32–36 [38] Tsao MN, Rades D, Wirth A, et al. Radiotherapeutic and surgical management
[15] Serizawa T, Yamamoto M, Nagano O, et al. Gamma Knife surgery for meta- for newly diagnosed brain metastasis(es): an American Society for Radiation
static brain tumors. J Neurosurg 2008; 109 (Suppl): 118–121 Oncology evidence-based guideline. Pract Radiat Oncol. 2012; 2: 210–225
[16] Nam TK, Lee JI, Jung YJ, et al. Gamma knife surgery for brain metastases in [39] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines
patients harboring four or more lesions: survival and prognostic factors. J in Oncology. Central Nervous Systems Cancer. Version 1.2014. Available at:
Neurosurg 2005; 102 (Suppl): 147–150 http://www.nccn.org/professionals/physician_gls/pdf/cns.pdf. Accessed May
[17] Gaspar L, Scott C, Rotman M, et al. Recursive partitioning analysis 7, 2014
(RPA) of prognostic factors in three Radiation Therapy Oncology Group [40] Knisely JP, Yamamoto M, Gross CP, Castrucci WA, Jokura H, Chiang VL. Radio-
(RTOG) brain metastases trials. Int J Radiat Oncol Biol Phys 1997; 37: surgery alone for 5 or more brain metastases: expert opinion survey. J Neuro-
745–751 surg 2010; 113 (Suppl): 84–89
[18] Bhatnagar AK, Flickinger JC, Kondziolka D, Lunsford LD. Stereotactic radiosur- [41] Nieder C, Leicht A, Motaref B, Nestle U, Niewald M, Schnabel K. Late radiation
gery for four or more intracranial metastases. Int J Radiat Oncol Biol Phys toxicity after whole brain radiotherapy: the influence of antiepileptic drugs.
2006; 64: 898–903 Am J Clin Oncol 1999; 22: 573–579
[19] Park SH, Hwang SK, Kang DH, et al. Gamma knife radiosurgery for multiple [42] Greene-Schloesser D, Moore E, Robbins ME. Molecular pathways: radiation-
brain metastases from lung cancer. J Clin Neurosci 2009; 16: 626–629 induced cognitive impairment. Clin Cancer Res 2013; 19: 2294–2300
[20] Yamamoto M, Barfod BE, Urakawa Y. Gamma knife radiosurgery for brain [43] Shors TJ, Miesegaes G, Beylin A, Zhao M, Rydel T, Gould E. Neurogenesis in
metastases of non-lung cancer origin: focusing on multiple brain lesions. Prog the adult is involved in the formation of trace memories. Nature 2001; 410:
Neurol Surg 2009; 22: 154–169 372–376
[21] Yamamoto M, Kawabe T, Barfod BE. How many metastases can be treated [44] DeAngelis LM, Delattre JY, Posner JB. Radiation-induced dementia in patients
with radiosurgery? Prog Neurol Surg 2012; 25: 261–272 cured of brain metastases. Neurology 1989; 39: 789–796
[22] Yamamoto M, Kawabe T, Sato Y, et al. A case-matched study of stereotactic [45] Tallet AV, Azria D, Barlesi F, et al. Neurocognitive function impairment after
radiosurgery for patients with multiple brain metastases: comparing treat- whole brain radiotherapy for brain metastases: actual assessment. Radiat On-
ment results for 1–4 vs ≥ 5 tumors: clinical article. J Neurosurg 2013; 118: col 2012; 7: 77
1258–1268 [46] Corn BW, Moughan J, Knisely JP, et al. Prospective evaluation of quality of life
[23] Karlsson B, Hanssens P, Wolff R, Söderman M, Lindquist C, Beute G. Thirty and neurocognitive effects in patients with multiple brain metastases receiv-
years’ experience with Gamma Knife surgery for metastases to the brain. J ing whole-brain radiotherapy with or without thalidomide on Radiation
Neurosurg 2009; 111: 449–457 Therapy Oncology Group (RTOG) trial 0118. Int J Radiat Oncol Biol Phys 2008;
[24] Chang WS, Kim HY, Chang JW, Park YG, Chang JH. Analysis of radiosurgical 71: 71–78
results in patients with brain metastases according to the number of brain [47] Ebi J, Sato H, Nakajima M, Shishido F. Incidence of leukoencephalopathy after
lesions: is stereotactic radiosurgery effective for multiple brain metastases? J whole-brain radiation therapy for brain metastases. Int J Radiat Oncol Biol
Neurosurg 2010; 113 (Suppl): 73–78 Phys 2013; 85: 1212–1217
[25] Hunter GK, Suh JH, Reuther AM, et al. Treatment of five or more brain meta- [48] Fujii O, Tsujino K, Soejima T, Yoden E, Ichimiya Y, Sugimura K. White matter
stases with stereotactic radiosurgery. Int J Radiat Oncol Biol Phys 2012; 83: changes on magnetic resonance imaging following whole-brain radiotherapy
1394–1398 for brain metastases. Radiat Med 2006; 24: 345–350
[26] Raldow AC, Chiang VL, Knisely JP, Yu JB. Survival and intracranial control of [49] Monaco EA III, Faraji AH, Berkowitz O, et al. Leukoencephalopathy after
patients with 5 or more brain metastases treated with gamma knife stereo- whole-brain radiation therapy plus radiosurgery versus radiosurgery alone
tactic radiosurgery. Am J Clin Oncol 2013; 36: 486–490 for metastatic lung cancer. Cancer 2013; 119: 226–232
[27] Caballero JA, Sneed PK, Lamborn KR, et al. Prognostic factors for survival in [50] Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treatment of recur-
patients treated with stereotactic radiosurgery for recurrent brain metastases rent previously irradiated primary brain tumors and brain metastases: final
after prior whole brain radiotherapy. Int J Radiat Oncol Biol Phys 2012; 83: report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys 2000; 47: 291–
303–309 298
[28] Suzuki S, Omagari J, Nishio S, Nishiye E, Fukui M. Gamma knife radiosurgery [51] Sahgal A, Barani IJ, Novotny J Jr, et al. Prescription dose guideline based on
for simultaneous multiple metastatic brain tumors. J Neurosurg 2000; 93 physical criterion for multiple metastatic brain tumors treated with stereo-
(Suppl 3): 30–31 tactic radiosurgery. Int J Radiat Oncol Biol Phys 2010; 78: 605–608

249
Stereotactic Radiosurgery for the Management of Five or More Brain Metastases

[52] Korytko T, Radivoyevitch T, Colussi V, et al. 12 Gy gamma knife radiosurgical [55] Regine WF, Huhn JL, Patchell RA, et al. Risk of symptomatic brain tumor re-
volume is a predictor for radiation necrosis in non-AVM intracranial tumors. currence and neurologic deficit after radiosurgery alone in patients with
Int J Radiat Oncol Biol Phys 2006; 64: 419–424 newly diagnosed brain metastases: results and implications. Int J Radiat On-
[53] Monaco EA III, Niranjan A, Kano H, Flickinger JC, Kondziolka D, Lunsford LD. col Biol Phys 2002; 52: 333–338
Management of adverse radiation effects after radiosurgery for arteriovenous [56] Jairam V, Chiang VL, Yu JB, Knisely JP. Role of stereotactic radiosurgery in pa-
malformations. Prog Neurol Surg 2013; 27: 107–118 tients with more than four brain metastases. CNS Oncol 2013; 2: 181–193
[54] Minniti G, Clarke E, Lanzetta G, et al. Stereotactic radiosurgery for brain meta-
stases: analysis of outcome and risk of brain radionecrosis. Radiat Oncol
2011; 6: 48

250
Repeat Radiosurgery for Brain Metastases

30 Repeat Radiosurgery for Brain Metastases


Aditya Halthore, Ami B. Raval, Michael Schulder, and Jonathan Knisely

benefits and drawbacks in the context of a review of the exist-


Key Points ing literature.

● Diagnosis of local progression after stereotactic radiosurgery


for brain metastases is often difficult. Specialized imaging 30.2 Decision to Treat
may help when evaluation of T1/T2 mismatch is not disposi-
The very diagnosis of progression or recurrence of brain meta-
tive.
stases previously treated with whole-brain irradiation or radio-
● The gold standard for determining local progression remains
surgery can prove challenging. Distinguishing between
biopsy or resection histopathologically documenting persis-
recurrence and radiation necrosis, for example, is not always
tent/recurrent tumor.
clear despite high-resolution, thin-slice structural magnetic res-
● Repeat radiosurgery for brain metastases is a viable treat-
onance imaging (MRI). As such, specialized imaging such as
ment modality.
proton magnetic resonance spectroscopy, perfusion-weighted
● Level I evidence evaluating repeat stereotactic radiosurgery
MRI, and 11C-methyl-L-methionine positron emission tomogra-
for brain metastases is needed.
phy (PET) or even a biopsy of the lesion is helpful in many cases
to confirm recurrence. More recently, literature has shown that
concordant enhancement on T1 postcontrast and T2 sequences
is more likely to represent recurrence rather than radiation
30.1 Introduction effect. Similarly, diffuse T2 signal enhancement that is discord-
The management of newly diagnosed brain metastases by ster- ant from rim enhancement on T1 sequencing may suggest radi-
eotactic radiosurgery (SRS) has been shown to provide excel- ation change, a concept known as “mismatch.”7,8,9,10,11
lent local control, with treated metastases showing local When considering delivery of repeat SRS for patients with
control rates at 1 year approaching 90%.1,2,3 In patients who previously treated brain metastases, evaluation of prognosis is
continue to demonstrate low overall disease burden (i.e., good important. Yamamoto et al (unpublished data) most recently
performance status and few visceral metastases), maintaining analyzed the recursive partitioning analysis (RPA) classification
good control of brain metastases remains paramount to extend- first described by Gaspar et al (1997) along with four other
ing survival and preserving quality of life. Randomized control prognostic schema to identify which index provides the best
trials comparing SRS to SRS combined with whole-brain radio- stratification of patients undergoing repeat radiosurgery for
therapy (WBRT) have shown that radiosurgery alone is not infe- brain metastases.12 The index with the best discrimination,
rior to combination therapy with regards to overall survival.1,2,3 known as the “modified” RPA classification, was based on a
As such, SRS alone has increasingly become a viable initial ma- cohort that included 804 patients who underwent repeat SRS
nagement strategy for patients with brain metastases. As radia- (local or remote). Of these patients, 122 (15%) received repeat
tion delivery technologies and systemic therapies have SRS for local recurrence. Within the entire cohort, patients in
improved, patients with brain metastases are also living long modified RPA classes I, II, and III had median survival times of
enough to experience intracranial recurrence—for example, one 14.5, 7.2, and 4.5 months, respectively.10 A combination of clin-
retrospective review of 1,300 patients from the Cleveland Clinic ical judgment and stratification based on such prognostic tools
(Cleveland, OH) treated for brain metastases showed that over is useful in deciding who may benefit from repeat radiosurgery
2.5% of patients survived at least 5 years. 4,5 As more patients and in whom such therapy may be futile.
begin to receive SRS alone for initial brain metastasis, the ques-
tion of how to manage local recurrence of metastases previ-
ously treated with radiosurgery has become increasingly 30.3 Efficacy and Toxicity of
relevant.
As in the case of newly diagnosed metastases, the options for
Repeat Radiosurgery
recurrent brain metastases include but are not limited to surgi- As with radiosurgery for the initial treatment of brain metasta-
cal extirpation, WBRT, and repeat radiosurgery. Emerging ses, the advantages of repeat SRS are many. Repeat radiosurgery
modalities such as magnetic resonance–guided focused laser minimizes the dose that normal brain tissue receives and pre-
interstitial thermal therapy may be considered in patients serves the capability to deliver WBRT at a later date. By delaying
whose metastases measure less than 3 cm or show significant or avoiding WBRT altogether, the patient may be spared poten-
failure to prior radiosurgery.6 For many patients, the risk of tially debilitating neurocognitive sequelae. This is particularly
morbidity from surgery outweighs its benefits, and as such, a relevant in patients who have a range of effective systemic ther-
radiotherapeutic approach is often preferred. In patients who apy options or those who have slow-growing primary histolo-
have previously received SRS for metastatic intracranial disease, gies such as certain breast cancers, in whom life expectancy can
repeat radiosurgery is a viable approach in the appropriate pa- be measured on the order of several months to years. Further-
tient, although there is no level I evidence to support this claim. more, deferring WBRT in the setting of small intraparenchymal
In this chapter, we discuss the feasibility of repeat radiosurgery metastases allows it to be utilized more effectively when the
for local metastatic recurrence in the brain, including potential number or size of metastases has progressed beyond the

251
Repeat Radiosurgery for Brain Metastases

capability of SRS or when the location of new metastases war- The evidence base for salvage radiosurgery following initial
rants a fractionated partial or whole-brain approach (e.g., for whole-brain therapy is somewhat larger and may shed light on
disease involving the leptomeninges or optic apparatus). Repeat the ability of repeat irradiation to provide effective local control
stereotactic radiosurgery, due to its inherently high dose per while avoiding normal tissue toxicity.17,18,19,20 In RTOG 9005,
fraction, may also afford better local control in radioresistant Shaw and colleagues demonstrated low rates of necrosis and
histologies such as melanoma, renal cell carcinoma, and sarco- local progression in the context of dose escalation SRS after
mas.13 Finally, patients who have a good performance status whole-brain irradiation for primary brain tumors and metasta-
may be candidates for further systemic therapy, which can be ses.18 In this study, the incidence of radionecrosis was 11% at 24
initiated more quickly with a single-fraction approach to brain months and local tumor progression occurred in 48% of all
metastasis treatment than with protracted fractionation. patients. In a retrospective series of 111 patients by Chao et al,
The potential downside of repeat radiosurgery pertains to treatment with salvage radiosurgery following initial WBRT
the composite dose delivered over multiple sessions, which if yielded a median survival of 10.4 months for patients with
high enough, can lead to symptomatic edema and radiation local-only recurrence, and two patients developed pathologi-
necrosis.14,15 Increased volume of metastasis treated as well as cally confirmed radiation necrosis within 1.5 years after SRS. 17
decreased interval between first and second treatments both
contribute to the risk of brain toxicity. Location may also play a
role in the clinical evaluation for possible repeat SRS—repeated 30.4 Clinical Vignette
doses delivered to a metastasis near the optic chiasm, for exam-
A 49-year-old woman with a history of triple-negative invasive
ple, may cause permanent visual impairment, whereas the
ductal carcinoma of the left breast was treated with mastec-
same total dose delivered to a peripheral lesion in noneloquent
tomy, systemic chemotherapy, and adjuvant radiotherapy. Four-
cortex may tend to be more innocuous.
teen months after definitive management, she developed gait
With regard to the outcome and toxicity profiles of repeat
instability and dizziness, for which diagnostic workup demon-
radiosurgery for local recurrence of brain metastases, there is
strated two large brain metastases. She underwent resection of
no level I evidence. However, two case series have retrospec-
a left frontal metastasis and subsequently received 24 Gy in
tively examined the outcome of patients with recurrent brain
three fractions to the resection bed. A smaller left parietal le-
metastases who received repeat local SRS.14,16 In a retrospective
sion was also treated in a single fraction of 20 Gy (▶ Fig. 30.1).
analysis by Kwon et al, 43 patients underwent repeat SRS, of
Surveillance imaging 5 months thereafter demonstrated stable
whom 30 were treated with repeat local SRS (16 patients had
disease within the original resection bed and left parietal meta-
local retreatment only and 14 had both repeat local and new
stasis; however, a new left frontal dural-based lesion inferior to
remote treatments).16 Median survival among the 43 re-treated
the resection bed was noted and treated with 20 Gy in one
patients was 68 weeks after the first SRS procedure and 32
weeks after the second procedure. Of the 30 patients who died
of known causes, 16 died of systemic progression of disease
whereas 14 died of neurologic compromise. The actuarial sur-
vival rate after second SRS was 58% and 28% at 6 months and 1
year, respectively. The intracranial progression-free rate was 91
and 86% at 6 months and 1 year, respectively. Nineteen percent
of the 43 patients developed symptomatic radiation necrosis.
Multivariate analysis of prognostic factors identified only RPA
class (I and II vs. III) as statistically significant, with median sur-
vival difference of 42 and 36 versus 12 weeks, respectively.
Number of lesions, tumor volume, and time interval between
treatments did not statistically impact survival. Aside from the
typical drawbacks of a retrospective study with limited cohort
size, conclusions from this study are confounded by the fact
that patients receiving remote SRS treatment were included in
the cohort—these patients may have presumably had better
survival and lower radiation necrosis rates than patients receiv-
ing local-only treatment.
Yamanaka et al published the results of a series of 41 patients
who received repeat Gamma Knife radiosurgery for brain meta-
stases.14 Only four patients (with eight targets) included in the
study received repeat SRS after local tumor recurrence. Local
control rate was on average 7.5 months after the first treatment
and 4.7 months after the second stereotactic procedure. Three
of these patients developed local recurrence and one developed Fig. 30.1 T1-weighted magnetic resonance imaging of brain with
histologically proven radiation necrosis in the unreported fol- gadolinium and superimposed dosimetry demonstrating left frontal
low-up period, a fact that the authors speculated was due to a resection cavity treated with 24 Gy in three fractions (target volume
theoretical resistance to high dose-per-fraction radiation and 30.7 cc) and left parietal metastasis treated with 20 Gy in one fraction
(target volume 10.4 cc). Isodose lines for 12, 20, and 24 Gy are shown.
large volume targets at the time of second SRS.

252
Repeat Radiosurgery for Brain Metastases

Fig. 30.2 T1-weighted magnetic resonance imaging with gadolinium performed 3 months after initial stereotactic radiosurgery (SRS) treatment.
Superimposed SRS dosimetry for new left frontal dural-based lesion treated with 20 Gy in one fraction is shown (target volume 6.5 cc). Planning was
constrained due to proximity to previous SRS treatment. Isodose lines for 12, 19, and 20 Gy are shown.

fraction (▶ Fig. 30.2). Follow-up imaging of the patient per- this guideline acknowledge a potential selection bias in the
formed 6 months after her initial SRS demonstrated enlarge- existing literature, as it is not clear whether favorable patient
ment and increased enhancement of the left parietal lesion; prognostic characteristics or radiosurgery is responsible for the
however, MR perfusion and single-photon emission computed apparent improvement in outcomes. In 2012, guidelines for
tomography (SPECT) imaging demonstrated the lesions to be appropriate care published by an expert panel of the American
consistent with radiation necrosis. The patient was then fol- College of Radiology (ACR) recommended considering SRS as a
lowed with short-interval surveillance imaging, which showed viable option for salvage therapy after recurrent remote or local
stable disease until 10 months after her original SRS, when she metastases following initial whole-brain irradiation, surgery, or
developed worsening neurologic symptoms, including head- even radiosurgery.22 They wrote that if recurrence can be
ache and dizziness. Both the left frontal and left parietal lesions detected prior to development of neurologic symptoms, SRS
demonstrated significant enlargement with increased edema, may be the most effective option for management. Importantly,
contrast enhancement, and mass effect. She underwent gross however, neither the ACR nor ASTRO guidelines described
total resection of the left frontal lesion and subtotal resection of above distinguish between local and remote intracranial
the left parietal lesion, with pathological findings consistent recurrence.
with tumor recurrence at both sites. Both the left frontal lesion
and left parietal resection beds were subsequently treated with
Gamma Knife radiosurgery (▶ Fig. 30.3a,b). Magnetic resonance 30.6 Summary
imaging performed approximately 1 month after her Gamma
When considering repeat SRS for locally recurrent brain meta-
Knife radiosurgery demonstrated all three resection beds—left
stases, a diagnosis of recurrence must first be confirmed, as
superior frontal, left inferior frontal, and left parietal—to have
radiation-induced changes may be present following initial
decreased size, stable-to-decreased contrast enhancement, and
radiotherapy and can mimic tumor on postcontrast MRI. Once a
decreased mass effect (▶ Fig. 30.4).
diagnosis has been established, the existing literature provides
level II and III evidence that may support the use of SRS for sal-
vage, particularly in patients with good performance status and
30.5 National Consensus prognosis in whom systemic options may still remain. Repeat
Guidelines radiosurgery at typical doses has a reasonable chance of local
control, and the majority of patients do not appear to develop
The American Society for Therapeutic Radiology and Oncology radionecrosis. We propose a framework here that outlines
(ASTRO) published a set of guidelines in 2005 that reviews the potential clinical pathways for patients with local recurrence
role of radiosurgery in the management of brain metastases. 21 previously treated with radiosurgery (▶ Fig. 30.5). At this time,
In particular, the review primarily cites level II-2 and II-3 evi- the precise role of radiosurgery in the management of recurrent
dence that shows improved local response and 1-year survival brain metastases treated previously by radiosurgery or radio-
for SRS salvage after whole-brain irradiation. The authors of therapy is not known. As the number of patients treated with

253
Repeat Radiosurgery for Brain Metastases

Fig. 30.3 T1-weighted magnetic resonance


imaging with gadolinium demonstrating recur-
rence of left frontal and left parietal lesions 10
months after initial treatment and superimposed
Gamma Knife dosimetry. (a) The left frontal
lesion recurred after initial resection and adjuvant
stereotactic radiosurgery (SRS); it was resected
and treated with a single fraction of 16 Gy to a
target volume of 3.4 cc. (b) The left parietal
lesion recurred after initial SRS alone; it was
resected and treated with a single fraction of
16 Gy to a target volume of 20.1 cc. Isodose lines
for 12, 16, and 30 Gy are shown.

Fig. 30.4 T1-weighted magnetic resonance imaging with gadolinium demonstrating three sites of previous stereotactic radiosurgery (SRS). Both left
frontal lesion (asterisk) and left parietal lesion (red arrow) were treated with repeat SRS. Left dural-based metastasis is also shown (white arrow). All
three lesions demonstrate decreased size, stable-to-decreased contrast enhancement, and decreased mass effect compared with prior imaging.

254
Repeat Radiosurgery for Brain Metastases

Fig. 30.5 Proposed algorithm for patients with local recurrence of brain metastases previously treated by stereotactic radiosurgery. MR, magnetic
resonance; PET, positron emission tomography; SRS, stereotactic radiosurgery; WBRT, whole-brain radiation therapy.

SRS alone for an initial presentation of brain metastases least one year after radiosurgery for brain metastases. Int J Radiat Oncol Biol
Phys 2003; 57: 452–464
increases, the need for further studies that provide level I evi-
[6] Hawasli AH, Bagade S, Shimony JS, Miller-Thomas M, Leuthardt EC. Magnetic
dence will be critical to evaluate the potential use of repeat resonance imaging-guided focused laser interstitial thermal therapy for in-
radiosurgery in recurrent brain metastases. tracranial lesions: single-institution series. Neurosurgery 2013; 73: 1007–
1017
[7] Plowman PN. Stereotactic radiosurgery. VIII. The classification of postradia-
References [8]
tion reactions. Br J Neurosurg 1999; 13: 256–264
Chernov M, Hayashi M, Izawa M, et al. Differentiation of the radiation-in-
[1] Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosurgery plus whole- duced necrosis and tumor recurrence after gamma knife radiosurgery for
brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: importance of multi-voxel proton MRS. Minim Invasive
brain metastases: a randomized controlled trial. JAMA 2006; 295: 2483– Neurosurg 2005; 48: 228–234
2491 [9] Kickingereder P, Dorn F, Blau T, et al. Differentiation of local tumor recurrence
[2] Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain from radiation-induced changes after stereotactic radiosurgery for treatment
metastases treated with radiosurgery or radiosurgery plus whole-brain irra- of brain metastasis: case report and review of the literature. Radiat Oncol
diation: a randomised controlled trial. Lancet Oncol 2009; 10: 1037–1044 2013; 8: 52
[3] Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant whole-brain radiotherapy [10] Yamamoto M, Sato Y, Serizawa T, et al. Subclassification of recursive parti-
versus observation after radiosurgery or surgical resection of one to three tioning analysis Class II patients with brain metastases treated radiosurgi-
cerebral metastases: results of the EORTC 22952–26001 study. J Clin Oncol cally. Int J Radiat Oncol Biol Phys 2012; 83: 1399–1405
2011; 29: 134–141 [11] Kano H, Kondziolka D, Lobato-Polo J, Zorro O, Flickinger JC, Lunsford LD. T1/
[4] Chao ST, Barnett GH, Liu SW, et al. Five-year survivors of brain metastases: a T2 matching to differentiate tumor growth from radiation effects after ster-
single-institution report of 32 patients. Int J Radiat Oncol Biol Phys 2006; 66: eotactic radiosurgery. Neurosurgery 2010; 66: 486–491, discussion 491–492
801–809 [12] Gaspar L, Scott C, Rotman M, et al. Recursive partitioning analysis (RPA) of
[5] Varlotto JM, Flickinger JC, Niranjan A, Bhatnagar AK, Kondziolka D, Lunsford prognostic factors in three Radiation Therapy Oncology Group (RTOG) brain
LD. Analysis of tumor control and toxicity in patients who have survived at metastases trials. Int J Radiat Oncol Biol Phys 1997; 37: 745–751

255
Repeat Radiosurgery for Brain Metastases

[13] Manon R, O’Neill A, Knisely J, et al. Eastern Cooperative Oncology Group. [18] Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treatment of recur-
Phase II trial of radiosurgery for one to three newly diagnosed brain metasta- rent previously irradiated primary brain tumors and brain metastases: final
ses from renal cell carcinoma, melanoma, and sarcoma: an Eastern Coopera- report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys 2000; 47: 291–
tive Oncology Group study (E 6397). J Clin Oncol 2005; 23: 8870–8876 298
[14] Yamanaka K, Iwai Y, Yasui T, et al. Gamma Knife radiosurgery for metastatic [19] Loeffler JS, Kooy HM, Wen PY, et al. The treatment of recurrent brain metasta-
brain tumor: the usefulness of repeated Gamma Knife radiosurgery for recur- ses with stereotactic radiosurgery. J Clin Oncol 1990; 8: 576–582
rent cases. Stereotact Funct Neurosurg 1999; 72 (Suppl 1): 73–80 [20] Davey P, O’Brien PF, Schwartz ML, Cooper PW. A phase I/II study of salvage
[15] Johnson M, Baschnagel AM, Chen PY, et al. Analysis of risk factors for develop- radiosurgery in the treatment of recurrent brain metastases. Br J Neurosurg
ment of radiation necrosis following gamma knife radiosurgery for brain 1994; 8: 717–723
metastases. Intl J Rad Oncol 2013; 87: 279–280 [21] Mehta MP, Tsao MN, Whelan TJ, et al. The American Society for Therapeutic
[16] Kwon KY, Kong DS, Lee JI, Nam DH, Park K, Kim JH. Outcome of repeated ra- Radiology and Oncology (ASTRO) evidence-based review of the role of radio-
diosurgery for recurrent metastatic brain tumors. Clin Neurol Neurosurg surgery for brain metastases. Int J Radiat Oncol Biol Phys 2005; 63: 37–46
2007; 109: 132–137 [22] Patel SH, Robbins JR, Gore EM, et al. Expert Panel on Radiation Oncology–
[17] Chao ST, Barnett GH, Vogelbaum MA, et al. Salvage stereotactic radiosurgery Brain Metastases. ACR Appropriateness Criteria® follow-up and retreatment
effectively treats recurrences from whole-brain radiation therapy. Cancer of brain metastases. Am J Clin Oncol 2012; 35: 302–306
2008; 113: 2198–2204

256
31 Promising Advances in Radiosurgery:
Part VIII Where Are the Frontiers? 258

Radiosurgical Horizon

VIII
Promising Advances in Radiosurgery: Where Are the Frontiers?

31 Promising Advances in Radiosurgery: Where Are the


Frontiers?
Brian D. Kavanagh

has prompted renewed interest in proton SRS. Recent reports


Key Points document the safety and efficacy of proton SRS in a variety of
clinical settings traditionally approached with photon-based
● Big data may help to refine the quality and outcomes associ- SRS,3,4,5,6 and further investigations are ongoing to explore oth-
ated with radiosurgery. er applications for proton SRS that might combine properties of
● Charged particles may play an expanded role in stereotactic charged particles that are not currently exploited to their fullest
radiosurgery and stereotactic body radiotherapy. potential. For example, as discussed by Bert and Durante in an
● Molecularly targeted agents may augment the efficacy and overview of workshop on particle radiosurgery held in Austria
safety of stereotactic radiosurgery. in 2013, there are opportunities to perform proton imaging and
even “proton microscopy” to reveal tumor and normal tissue
properties not presently captured by X-ray imaging, and their
incorporation into so-called theranostic strategies for a variety
31.1 Introduction of conditions is an intriguing possibility.7 Much work remains
to improve our understanding of all particle interactions with
The notable successes achieved in the past half century in the
human tissues, but likely there will be forthcoming insights that
clinical implementation of stereotactic radiosurgery (SRS) are
yield progress for a variety of benign and malignant disease.
well chronicled throughout the preceding chapters of this text.
A high level of understanding has been achieved in the area of
technical refinements, from image guidance to beam optimiza-
tion and modulation to safety and quality measures. There are
31.3 Prediction 2: Big Data Will
also abundant published data concerning the use of SRS in a Teach Us about Radiosurgery
wide range of clinical indications, from the treatment of func-
The advent of the electronic medical record has ushered in a
tional neurologic disorders to the ablation of primary and
new era in medicine where enormous amounts of data are
metastatic tumor deposits. It is, therefore, tempting to conclude
potentially retrievable. With proper attention to methodologi-
that there might be few if any remaining unrecognized frontiers
cal detail, large-scale observational studies are fertile ground
for investigation. Have all problems been solved, all questions
for harvesting new insights about how we can improve the
answered? Where might we or should we go from here? Will
quality of medical care rendered to the population. One bias to
SRS look or feel any different 50 years from now?
overcome, ironically, was pointed out in 2010 in an editorial in
One or another variation of the expression, “It is tough to
the Journal of the American Medical Association, namely, that “a
make predictions, especially about the future” has been attrib-
generation of clinicians has been taught that randomized con-
uted to pundits as far away from each other in the intellectual
trolled trials (RCTs) can find truth, whereas observational stud-
spectrum of human thinking as the Nobel Prize–winning physi-
ies are inherently flawed.”8 The reality is that although the RCT
cist, Niels Bohr, and the legendary baseball icon, Yogi Berra, 1
will remain an important tool for testing medical hypotheses,
and numerous others in between. This timeless caveat is often
the traditional methods of advancing medical knowledge via
invoked prior to speculation about what lies ahead and here
classic prospective clinical trials have become expensive and at
would serve similarly to acknowledge the uncertainty associ-
times unwieldy, limiting their feasibility to address all ques-
ated with trying to foresee upcoming trends. Nevertheless, it
tions of clinical importance, especially when the issue is a mat-
can be useful to consider the emerging opportunities for devel-
ter of trying to identify small differences in outcomes between
oping SRS that are suggested by current investigational efforts.
established treatments. For proper statistical power in a RCT to
In this piece, a handful of predictions is offered in the spirit of
detect the superiority of a novel approach that might be a few
knowing that they might all be incorrect and yet hoping that at
percent better than the accepted gold standard, thousands of
least by mentioning them, dialogue leading to better ideas is
patients would have to be enrolled.
stimulated.
In 2014, the American Association of Neurological Surgeons
(AANS) and the American Society for Radiation Oncology
(ASTRO) entered into a partnership to launch a prospective SRS
31.2 Prediction 1: The Particle registry. The AANS had already managed a very large and very
Strikes Back successful spine surgery registry that has yielded a number of
published papers offering insights into matters of quality assur-
The first modality used for SRS in the 1950s was a proton ance, patient selection, cost-effectiveness, and patient-reported
beam,2 although limitations in the capacity for image guidance outcomes.9,10,11 There is good reason to expect that the AANS
and other technical challenges dampened enthusiasm for this and ASTRO SRS Registry will provide equally valuable knowl-
modality in all but a few centers as cobalt-60 and linear acceler- edge of the type that can only be gleaned from the study of
ator–based devices gained traction. In recent years, however, large numbers of patients (thousands and thousands) and that
the proliferation of clinical proton and carbon therapy facilities is unobtainable from other sources.

258
Promising Advances in Radiosurgery: Where Are the Frontiers?

observe radiation effects and learn lessons quickly. It would be


31.4 Prediction 3: SRS Will Be wise for neurosurgeons and radiation oncologists to follow
Combined with Novel Molecular developments in the field of veterinary SRS in the coming years
because likely there will be insights gained that have transla-
Agents and Nanoparticles for the tional value to the human patient population.
Purpose of Reducing Side Effects
and Enhancing Therapeutic
Efficacy References
Stereotactic radiosurgery is understood to achieve beneficial [1] Cf. multiple discussions, e.g., http://en.wikiquote.org/wiki/Niels_Bohr#Dis-
effects through a variety of mechanisms that include impair- puted and http://quoteinvestigator.com/2013/10/20/no-predict/, both ac-
ment of the reproductive machinery of neoplastic cells and cessed August 25, 2014, and Hill B, Proceedings of the Meeting, J R Stat Soc
Ser A Gen 1956; 119: 147
apoptotic cell death in endothelial cells, among other effects.
[2] Larsson B, Leksell L, Rexed B, Sourander P, Mair W, Andersson B. The high-en-
But we are only beginning to accumulate enough layers of ergy proton beam as a neurosurgical tool. Nature 1958; 182: 1222–1223
understanding of the complex cellular signaling events that are [3] Walcott BP, Hattangadi-Gluth JA, Stapleton CJ, Ogilvy CS, Chapman PH,
triggered by SRS to allow us to exploit new molecular agents Loeffler JS. Proton beam stereotactic radiosurgery for pediatric cerebral
that selectively modulate pathways leading to normal brain arteriovenous malformations. Neurosurgery 2014; 74: 367–373, discus-
sion 374
injury or tumor response. Likely in the coming decades, we will
[4] Halasz LM, Bussière MR, Dennis ER, et al. Proton stereotactic radiosurgery for
begin to incorporate agents that might, for example, inhibit gly- the treatment of benign meningiomas. Int J Radiat Oncol Biol Phys 2011; 81:
cogen synthase kinase (GSK)-3β or block Kv1.3 channels, there- 1428–1435
by affording normal tissue protection that could allow for dose [5] Petit JH, Biller BM, Yock TI, et al. Proton stereotactic radiotherapy for persis-
escalation.12,13 Likewise, the burgeoning field of nanomedicine tent adrenocorticotropin-producing adenomas. J Clin Endocrinol Metab
2008; 93: 393–399
will almost certainly find its way into an interface with SRS,
[6] Lettmaier S. Radiosurgery in trigeminal neuralgia. Phys Med 2014; 30:
either in the context of the use of gold nanoparticles as radio- 592–595
sensitizers14,15 or perhaps in the setting of designing targeted [7] Bert C, Durante M. Particle radiosurgery: a new frontier of physics in medi-
nanoparticles that are specifically drawn to irradiated tissue to cine. Phys Med 2014; 30: 535–538
[8] Concato J. Is it time for medicine-based evidence? JAMA 2012; 307: 1641–
facilitate drug delivery to tumors or regions of functional disor-
1643
der in the brain.16 [9] Godil SS, Parker SL, Zuckerman SL, et al. Determining the quality and effec-
tiveness of surgical spine care: patient satisfaction is not a valid proxy. Spine J
2013; 13: 1006–1012
31.5 Prediction 4: SRS as Applied [10] Ghogawala Z, Shaffrey CI, Asher AL, et al. The efficacy of lumbar discectomy
and single-level fusion for spondylolisthesis: results from the NeuroPoint-SD
for Human Patients Will Be registry: clinical article. J Neurosurg Spine 2013; 19: 555–563
[11] Mummaneni PV, Whitmore RG, Curran JN, et al. Cost-effectiveness of lumbar
Refined by Lessons Learned from discectomy and single-level fusion for spondylolisthesis: experience with the
NeuroPoint-SD registry. Neurosurg Focus 2014; 36: E3
Man’s Best Friend [12] Jiang X, Perez-Torres CJ, Thotala D, et al. A GSK-3β inhibitor protects
against radiation necrosis in mouse brain. Int J Radiat Oncol Biol Phys
Techniques in cancer management for the veterinary popula- 2014; 89: 714–721
tion have become very sophisticated in recent years, paralleling [13] Peng Y, Lu K, Li Z, et al. Blockade of Kv1.3 channels ameliorates radiation-in-
progress made in the human population. For veterinary radia- duced brain injury. Neuro-oncol 2014; 16: 528–539
[14] Hainfeld JF, Smilowitz HM, O’Connor MJ, Dilmanian FA, Slatkin DN. Gold
tion oncology specifically, there is a dissemination of high tech-
nanoparticle imaging and radiotherapy of brain tumors in mice. Nanomedi-
nology that might be surprising to those unfamiliar with the cine (Lond) 2013; 8: 1601–1609
field, and thousands of domestic animals are treated with ther- [15] Joh DY, Sun L, Stangl M, et al. Selective targeting of brain tumors with gold
apeutic irradiation annually.17 There is frequently an opportu- nanoparticle-induced radiosensitization. PLoS ONE 2013; 8: e62425
nity for mutually beneficial learning from the act of comparing [16] Passarella RJ, Spratt DE, van der Ende AE, et al. Targeted nanoparticles that
deliver a sustained, specific release of Paclitaxel to irradiated tumors. Cancer
and contrasting how treatments are administered and tolerated
Res 2010; 70: 4550–4559
in conditions of sometimes widely different anatomical config- [17] Farrelly J, McEntee MC. A survey of veterinary radiation facilities in 2010. Vet
urations. In the area of SRS, technology of a quality and com- Radiol Ultrasound 2014
plexity identical to that used in human patients is commonly [18] Mariani CL, Schubert TA, House RA, et al. Frameless stereotactic radiosurgery
for the treatment of primary intracranial tumours in dogs. Vet Comp Oncol
implemented.18,19 One of the interesting features about canine
2013
oncology in particular is that given the shorter life span of dogs, [19] Glasser SA, Charney S, Dervisis NG, et al. Use of an image-guided robotic ra-
often events happen in a more compressed time frame, with diosurgery system for the treatment of canine nonlymphomatous nasal tu-
differing tissue growth kinetics, providing an opportunity to mors. J Am Anim Hosp Assoc 2014; 50: 96–104

259
Index
1 Agent combinations 259 Brain metastases, see Cancer and cancer –– angiographically occult vascular
American Association of Physicists in classifications malformations (AOVMs) 81, 83
1 to 4 metastases 235 Medicine (AAPM) protocols 32 – five or more 243 –– arteriovenous malformations
– See also Metastases American Society for Therapeutic Radi- – one to four 235 (AVMs) 81
ology and Oncology (ASTRO) 247, – repeat radiosurgery for 251 –– classification 82
253 Brain Tumor Cooperative Group –– de novo formations 82
5 Angioarchitecture 91 (BTCG) 226 – overviews of 81, 89
5 or more metastases 243 Angiographically occult vascular mal- Brain Tumor Study Group (BTSG) 226 – references for 89
– See also Metastases formations (AOVMs) 81, 83 Brainstem, dose tolerance of 53 – stereotactic radiosurgery (SRS)
Angiomas, cavernous 81 Brown-Séquard syndrome 54 for 83
– See also Cerebral cavernomas (CCMs) BRW frames 31 –– critical considerations for 85, 86
A Antisecreting medications 116 BTCG, see Brain Tumor Cooperative –– epilepsy effects of 84
AOVMs, see Angiographically occult Group (BTCG) ––– See also Epilepsy
AAPM protocols, see American Associa-
vascular malformations (AOVMs) BTSG, see Brain Tumor Study Group –– hemorrage risks 88
tion of Physicists in Medicine
ARMD, see Age-related macular degen- (BTSG) –– hemorrhage rate effects of 84
(AAPM) protocols
eration (ARMD) Bunge, Hernan 8 –– morbidity after 88
Ablation, see Radiofrequency (RF) abla-
Arteriovenous fistulas, see Dural arte- Burchiel classification 160, 160 –– rationales for 84
tion
riovenous fistulas (DAVFs) –– reviews of 85, 87
Acromegaly 111, 113–114
Arteriovenous malformations – treatment options for 83, 84
Activities of Daily Living (ADLs) 169
(AVMs) 68 C Chadwick, James 11
Acute radiation injuries 112
– decision-tree algorithm for 68, 69 Callosotomy, corpus 197 Charged particle equilibrium (CPE) 32
– See also Radiation injuries
– dural arteriovenous fistulas Cancers and cancer classifications, see Charged particle therapy 121
Adenomas, pituitary 107
(DAVFs) 78, 79 Metastases Chescotta, Roberto 8
– overviews of 107, 117
– future directions for 78–79 – adenomas, pituitary 107 Chondrosarcomas 128
– prognoses for 115
– historical perspectives of 68 – cerebral cavernomas (CCMs) 81 – classification of 128
– references for 117
– large 76, 77–78 – chondrosarcomas 128 – definition of 128
– special considerations for 115
– lesional epilepsy and 194 – chordomas 120 – North American Gamma Knife Con-
– sterotactic radiosurgery (SRS)
–– See also Lesional epilepsy – epileptogenic tumors 194 sortium (NAGKC) reports on 129,
for 108
– overviews of 68 – glial tumors 226 129, 130
–– clinical outcomes, acromegaly 111,
– references for 79 – glomus tumors 226 – overviews of 128–129
113–114
– stereotactic radiosurgery (SRS) – hamartomas, hypothalamic – references for 130
–– clinical outcomes, Cushing dis-
for 68 (HHs) 182 – sterotactic radiosurgery (SRS)
ease 111, 111–112
–– adverse effects of 75 – hemangioblastomas 146 for 128
–– clinical outcomes, nonfunctioning
–– bleeding risks of 74, 74 – meningiomas 102 – treatment options for 128
pituitary adenomas 110
–– outcomes for 73, 74 – pediatric brain tumors 210 –– radiotherapy (RT) 128
–– complications of 112
–– post-surgical obliteration 75 – schwannomas, see Schwannomas –– surgical resections 128
–– cranial neuropathy and 114
–– preradiosurgical embolization 77 –– nonvestibular cranial nerve Chordomas 120
–– critical structures, dose limitations
–– procedures for 72, 73 (NVCNS) 137 – complications of 125, 126
to 109
–– repeat 76 –– vestibular (VSs) 150 – definition of 120
–– dose selection criteria 108
––– See also Repeat radiosurgery Cavernomas 81 – epidemiology of 120
–– efficacy of 116
– University of Pittsburgh experien- – See also Cerebral cavernomas (CCMs) – histopathology of 120
–– frame placement 108
ces 69, 70 Cavernous angiomas 81 – overviews of 120, 126
–– histological entities and 116
ASTRO, see American Society for Thera- – See also Cerebral cavernomas (CCMs) – pathogenesis of 120
–– historical perspectives of 107
peutic Radiology and Oncology (AS- Cavernous hemangiomas 81 – radiographic features of 120
–– hypopituitarism and 114
TRO) – See also Cerebral cavernomas (CCMs) – references for 126
–– hypothalamic dysfunction and 114
Astrocytomas, pilocytic (PAs) 226 Cavernous malformations (CMs) 81, – stereotactic radiosurgery (SRS)
–– optic neuritis and 114
Auditory apparatus, dose tolerance 194 for 121, 123
–– patient selection criteria 107
of 54 – See also Cerebral cavernomas (CCMs) – treatment options for 121
–– pre-surgical evaluations 107
AVMs, see Arteriovenous malforma- CBCT, see Cone-beam computed to- –– charged particle therapy 121
–– principles of 108
tions (AVMs) mography (CBCT) –– conventional radiation therapy 121
–– radiation injuries, acute 112
CCMs, see Cerebral cavernomas (CCMs) –– surgical management 121
–– radiation injuries, early de-
Central nervous system (CNS) toler- Choroidal neovascular membranes
layed 114 B ance 52 (CNVMs) 207
–– radiation injuries, late delayed 115
Backlund, Erik-Olaf 5–6, 7–8 – critical structures 52 Choroidal neovascularization
–– radiation necrosis and 115
Bainbridge, Kenneth 12 –– auditory apparatus 54 (CNV) 207
–– radiographic images 108
Barrow Neurologic Institute (BNI) Pain –– brain 52 Circular collimators/cones 49, 49, 50
–– techniques 108
Intensty Scale 160, 160 –– brainstem 53 – See also Collimators
–– treatment planning 108, 109
Barrow’s classification 91 –– optic pathway 54 Clarke, Robert H. 3, 12
–– whole-sellar 116
Beam shaping and delivery 33 –– spinal cord 54 Clinical hemorrhage, see Hemorrhage,
– treatment options for 115
Betti, Osvaldo 8 – overviews of 52, 55 clinical
–– antisecreting medications, radiore-
Bilateral pallidotomy 172 – radiation necrosis treatments 55 Clinical target volumes (CTVs) 35
sistant effects of 116
– See also Pallidotomy – references for 56 CMs, see Cavernous malformations
–– external beam radiation therapy
Bilateral trigeminal neuralgia 164 Cerebral cavernomas (CCMs) 81 (CMs)
(EBRT) 115
– See also Trigeminal neuralgia – clinical hemorrhage and 81 CNS tolerance, see Central nervous sys-
–– up-front radiosurgery 116
Bite plates 47, 48–49 –– definition of 82 tem (CNS) tolerance
Adjustable Nomogrip TALON devi-
BNT Pain Intensity Scale, see Barrow –– morbidity of 82 CNV, see Choroidal neovascularization
ces 31
Neurologic Institute (BNI) Pain In- –– risk of 82 (CNV)
ADLs, see Activities of Daily Living
tensty Scale – hemispheric/superficial 81 CNVMs, see Choroidal neovascular
(ADLs)
Boost therapy 211 – natural history of 81, 83 membranes (CNVMs)
Age-related macular degeneration
Borden-Shuicart system 91 Cognard system 91
(ARMD) 207

260
Index

Collimators 49 DAVFs, see Dural arteriovenous fistulas – machine-specific reference (msr) 32


– See also Devices (DAVFs)
E – nonstandard 31
– circular/cones 49, 49, 50 DBS, see Deep brain stimulatin (DBS) Ear canal immobilization 47 – plan-class-specific reference
– multileaf (MLCs) 49, 49 De novo formations 82 – See also Immobilization (pcsr) 32
Colombo, Federico 8, 13 – See also Cerebral cavernomas (CCMs) Early delayed/subacute radiation inju- – problems of 31
Complications, see Radiation injuries Deep brain stimulation (DBS) 168–169, ries 52, 114 – small 30–32
– chordomas 125, 126 172, 174, 174 – See also Radiation injuries Fistulas, see Dural arteriovenous fistu-
– dural arteriovenous fistulas Delayed radiation injuries, see Radia- EBRT, see External beam radiation ther- las (DAVFs)
(DAVFs) 96, 98 tion injuries apy (EBRT) Five or more metastases 243
– glial tumors 231 Derechinsky, Victor 13 Eclipse plans 36, 36, 43 – See also Metastases
– metastases, five or more 247 Detectors 32 Edlefsen, Niels 11 Fixation, eye 202
– metastases, one to four 240 Devices 39 Electronic data and medical re- Floor-mounted/floor-stand positioning
– nonvestibular cranial nerve schwan- – collimators 49 cords 258 systems 44, 45
nomas (NVCNS) 143 –– circular/cones 49, 49, 50 Embolization, preradiosurgical 77 – See also Positioning systems
– pediatric brain tumors 221 –– multileaf (MLCs) 49, 49 End-to-end tests 38 Forster, David 7
– pituitary adenomas 112 – CyberKnife 16 Ependymomas 211, 212–213 Forward vs. inverse techniques 33
– radiation necrosis 55, 115 – Gamma Knife 2 Epilepsy 193 Fractionation 20
– trigeminal neuralgia 163 – overviews of 42, 42, 43, 50 – lesional 194 – effects estimate calculations 22, 22
– vestibular schwannomas (VSs) 154 – positioning systems, comparisons –– arteriovenous malformations – high-dose single fractions 20
Cone-beam computed tomography of 43 (AVMs) 194 – limitations of 23
(CBCT) 37 – positioning systems, minimally-inva- ––– See also Arteriovenous malforma- – multiple small-dose fractions 20
Cones, see Circular collimators/cones sive 43 tions (AVMS) Future directions 226
Conformity vs. inhomogeneity 35 –– comparisons of 43 –– cavernous malformations – electronic data and medical re-
Conventional radiation therapy 121 –– couch-mounted 45 (CMs) 194 cords 258
Cooper, Irving 3 –– floor-mounted 44, 45 –– cerebral cavernomas (CCMs) – human-veterinary radiation oncol-
Corpus callosotomy 197 –– floor-stand 44, 45 and 84 ogy parallels 259
Couch-mounted positioning sys- –– Gamma Knife 44 ––– See also Cerebral cavernomas – nanoparticles 259
tems 45 –– imaging-dependent 46 (CCMs) – novel molecular agent combina-
– See also Positioning systems –– linear accelerator (linac) frame sys- –– epileptogenic tumors 194 tions 259
CPE, see Charged particle equilibrium tems 44 –– hypothalamic hamartomas (HHs) – of linear accelerator (linac) radiosur-
(CPE) – positioning systems, noninvasive 46 and 194 gery 14
Cranial nerve (CN) schwanommas, see –– comparisons of 46 ––– See also Hypothalamic hamarto- – overviews 255, 258
Nonvestibular cranial nerve schwan- –– immobilization 47 mas (HHs) – proton-based techniques 258
nomas (NVCNS) ––– See also Immobilization – other nonlesional 197 – references 259
Cranial neuropathy 114 –– localization 48 –– corpus callosotomy 197 – side effect reductions 259
Craniopharyngiomas 217 ––– See also Localization –– neocortical foci 197 – therapeutic efficacy enhance-
Creutz, Edward 11 – references for 50 – overviews of 193, 198 ments 259
Critical structures 52 Diagnostic-specific Graded Prognostic – physiological 195
– Central nervous system (CNS) toler- Assessment (DS-GPA) 235 –– clinical outcomes for 195
ance, dose limitations 52, 55, 109 Digital reconstructed radiographs –– medial temporal lobe epilepsy G
– dose tolerance 52, 55, 109 (RDDs) 37 (MTLE) 195 G-Frames, Leskell 31
–– auditory apparatus 54 Dosimetry 203 –– morbidities of 196
Gamma Knife 2
–– brain 52, 52 DS-GPA, see Diagnostic-specific Graded –– neurocognition and 197
– clinical experiences with 3–4, 4, 5–7
–– brainstem 53 Prognostic Assessment (DS-GPA) –– secondary outcomes of 197
– clinical outcomes with 3, 3
–– dose-volume histogram (DVH) anal- Dural arteriovenous fistulas –– seizure remission for 195, 195, 196
– current configurations of 7
yses 52 (DAVFs) 78, 79, 91 –– treatment protocols for 196
– first designs of 5
–– limitation overviews 109 – angioarchitecture of 91 – references for 198
– future directions for 8
–– optic pathway 54 – classification of 91 – stereotactic radiosurgery (SRS) – historical perpsectives of 2
–– organs-at-risk (OARs) data 55, 56 –– Barrow’s 91 for 193
– overviews of 2, 2, 8, 44
–– Quantitative Analysis of Normal Tis- –– Borden-Shuicart system 91 –– experimental models of 193 – prototypes of 7
sue Effects in the CLinic (QUANTEC) –– Cognard system 91 –– radiation mechanisms of 193
– references for 9
report 52 – clinical manifestations of 92, 92 Epileptogenic tumors 194 – term derivation of 5
–– spinal cord 54 – natural history of 96 Essential tremor (ET) 174
GBMs, see Glioblastomas (GBMs)
– overivews of 52, 55 – overviews of 91, 98 – See also See Movement disorders
Gill-Thomas-Cosman (GTC) frames 31
– radiation necrosis treatments 55 – references for 98 ET, see Essential tremor (ET)
Glaucoma 206, 206
– references for 56 – special considerations for 96 Exophthalmos refratory to connserva-
Glial tumors 226
CTVs, see Clinical target volumes (CTVs) – steroeotactic radiosurgery (SRS) tive treatments 207 – glioblastomas (GBMs) 226, 229, 230,
Cushing disease 111, 111–112 for 93 Experimental models 193
232
CyberKnife 16 –– clinical outcomes 94, 94, 95 External beam radiation therapy – gliomas 226
– current configuations of 17 –– complications of 95, 96 (EBRT) 115
–– high-grade 228, 229
– extracranial radiosurgery applica- –– dose prescriptions 94 Extracranial radiosurgery applica- –– low-grade 228
tions of 17 –– follow-up programs 94 tions 17
–– malignant, newly-diagnosed 230,
– frame vs. frameless techniques 17 –– frame placements 93 Eye fixation 202
232
– historical perspectives of 16 –– principles 93 Eye-related disorders, see Ocular disor-
–– malignant, recurrent 229, 229, 230
– image guidance and 16 –– radiographic images 93 ders
– optic nerve tumors 226
– linear accelerator (linac) technology –– treatment planning 94 – overviews of 226, 232
and 16, 17 – treatment options for 93 – pilocytic astrocytomas (PAs) 226,
– overviews of 16 –– management strategies 97, 98 F 228
– references for 18 –– selection criteria 97 Factor-VIII immune reactions 64 – references for 232
–– sinus recanalization roles 97 Fields 30 – stereotactic radiosurgery (SRS)
Dystonia 168 – charged particle equilibrium for 226
D – See also Movement disorders (CPE) 32 –– Brain Tumor Cooperative Group
Davenport, Lee 12 – effects of 32 (BTCG) studies 226

261
Index

–– Brain Tumor Study Group (BTSG) –– multicenter retrospective trials 148 Imaging-dependent positioning sys- – Stereotaxic Method and Radiosur-
studies 226 –– techniques 146 tems 46 gery of the Brain 11
–– complications of 231 – treatment options for 146 – See also Positioning systems Linac radiosurgery, see Linear accelera-
–– Radiation Therapy Oncology Group –– fractionated radio therapy (RT) 146 Immobilization 47 tor (linac) radiosurgery
(RTOG) studies 229 –– surgical resections 146 – See also Noninvasive positioning sys- Lindquist, Christer 7
– treatment options for 226 – von Hippel-Lindau (VHL) disease tems Linear accelerator (linac) radiosur-
Glioblastomas (GBMs) 226, 229, 230, and 146 – bite plates 47, 48–49 gery 11
232 Hemangioblastomas, ocular 207 – ear canal 47 – frame systems 44
Gliomas 226 Hemangiomas, cavernous 81 – eye fixation 202 –– See also Minimally-invasive posi-
– See also Glial tumors – See also Cerebral cavernomas (CCMs) – thermoplastic masks 47, 47 tioning systems
– high-grade 228, 229 Hemispheric/superficial cerebral caver- Infrared guidance 37 – future directions for 13, 14
– low-grade 228 nomas (CCMs) 81 Inhomogeneity vs. conformity 35 – historical perspectives of 11
– malignant 229 – See also Cerebral cavernomas (CCMs) International Atomic Energy Agency – overviews of 11
–– newly-diagnosed 230, 232 Hemorrhage, clinical 81 (IAEA) protocols 32 – physics of 30
–– recurrent 229, 229, 230 – rate effects of 84 Inverse vs. forward techniques 33 – references for 14
– pediatric 215 – risks 88 Ionization 193 – stereotactic principles of 12
Glomus tumors 226 HHs, see Hypothalamic hamartomas – definition of 193 Linear-quadratic (LQ) models 22
– glomus jugular tumors 134 (HHs) – overviews of 193, 198 Localization 48
– overviews of 132, 136 Hickman, Roger W. 12 – references for 198 – manual target 6, 6
– references for 136 Hidden target tests 38 Isocentric vs. nonisocentric techni- – multiplanar X-ray imaging 48
– stereotactic radiosurgery (SRS) High-grade gliomas 228, 229 ques 34 – optical tracking 48
for 132 – See also Gliomas – surface monitoring 48
–– clinical outcomes of 134, 135 Historical perspectives 16 Low-grade gliomas 228
–– dose planning 132 – Gamma Knife 2 J – See also Gliomas
–– dose prescription 133 – of linear accelerator (linac) radiosur- Jernberg, Bengt 6 LQ models, see Linear-quadratic (LQ)
–– evaluations 134 gery 11 models
–– meta-analyses of 134 – of proton beam radiosurgery 11 Lunsford, L. Dade 5, 8
–– patient selection criteria for 135 Hopkins Verbal Learning Test K Lutz, Wendell 13
–– postoperative care 134 (HVLT) 236, 240
Karnosky Performace Score (KPS) 240
–– stereotactic imaging 132, 133 Horsley, Victor A. H. 3, 12
– University of Pittsburgh experience Hot spots 35
Kilovoltage-megavoltage (kV-MV) im- M
age pairings 37
with 132 HRST, see Hypofractionated stereotactic Machine-specific reference (msr) 32
Kjellberg, Raymond 8, 12
GPA, see Graded Prognostic Assessment radiosurgery (HRST) Manhattan Project 12
KPS, see Karnosky Performace Score
(GPA) Human-veterinary radiation oncology Manual target localization 6, 6
(KPS)
Graded Prognostic Assessment parallels 259 Margins 35
(GPA) 235 HVLT, see Hopkins Verbal Learning Test – dose conformity vs. inhomogene-
Granit, Ragnar 2 (HVLT) L ity 35
Grantry-mounted vs. room-mounted Hypofractionated stereotactic radiosur- – eclipse plans 35, 36, 36
systems 37 gery (HRST) 239 Large arteriovenous malformations
– hot spots 35
Greuter, Matthias 7, 7 Hypopituitarism 114 (AVMs) 76, 77–78
– margin assumption 35
Gross tumor volumes (GTVs) 35 Hypothalamic dysfunction 114 – See also Arteriovenous malforma-
– multimodality imaging registra-
GTC frames, see Gill-Thomas-Cosman Hypothalamic hamartomas (HHs) 182 tions
tion 36
(GTC) frames – classification of 182 Late delayed radiation injuries 115
– planning target volumes (PTVs) 35
GTVs, see Gross tumor volumes (GTVs) – definition of 182 – See also Radiation injuries
– prescription isodose spillover 35
– histology of 182, 183 Late Effects Normal Tissues, subjective,
Masks, thermoplastic 47, 47
– imaging findings for 182 objective, management, and analytic
Medial temporal lobe epilepsy
H – lesional epilepsy and 194 (LENT SOMA) scoring system 204
(MTLE) 195
–– See also Epilepsy Late stage 58
Hamartomas, hypothalamic (HHs) 182 – See also Physiological epilepsy
– natural history of 183, 184–185 Lawrence, Ernest Orlando 11
– classification of 182 Medical records, electronic 258
– overviews of 182, 190 Lawrence, John H. 8, 11
– definition of 182 Meningiomas 102
– physiology of 182 LENT SOMA scoring system, see Late Ef-
– histology of 182, 183 – classification of 103
– references for 191 fects Normal Tissues, subjective, ob-
– imaging findings for 182 –– location-specific series 103, 104–
– stereotactic radiosurgery (SRS) jective, management, and analytic
– lesional epilepsy and 194 105
for 184 (LENT SOMA) scoring system
–– See also Epilepsy –– major series 103
–– action mechanisms of 189 Lesional epilepsy 194
– natural history of 183, 184–185 – overviews of 102, 105
–– complications of 188 – See also Epilepsy
– overviews of 182, 190 – pediatric 216, 216
–– Gamma Knife techniques 189, 189, – arteriovenous malformations
– physiology of 182 – references for 105
190 (AVMs) 194
– references for 191 – stereotactic radiogurgery (SRS)
–– reviews of 187, 188 – cavernous malformations (CMs) 194
– stereotactic radiosurgery (SRS) for 102
–– technique comparisons 186 – epileptogenic tumors 194
for 184 –– historical perspectives of 102
–– vs. open surgical procedures 186 – hypothalamic hamartomas (HHs)
–– action mechanisms of 189 –– techniques 102
and 194
–– Gamma Knife techniques 189, 189, – sterotactic radiosurgery (SRS)
–– See also Hypothalamic hamartomas
190 for 105
–– reviews of 187, 188
I (HHs)
Metastases, see Cancers and cancer
Leskell frames 3–4, 31
–– technique comparisons 186 IAEA protocols, see International classifications
Leskell Gamma Knife 2
–– vs. open surgical procedures 186 Atomic Energy Agency (IAEA) proto- – carcinomas, orbital/uveal 207
– See also Gamma Knife
Harvard Cyclotron Laboratory 12, 12 cols – five or more 243
Leskell, Lars 6
Hearing toxicity 54 Image guidance techniques 36 –– American Society for Therapeutic
– Gamma Knife and 2
Hemangioblastomas 146 – comparisons of 36 Radiology and Oncology (ASTRO)
– linear acceleration radiotherapy
– overviews of 146, 148 – cone-beam computed tomography National Concensus Guidelines
and 11
– references for 149 (CBCT) 37 for 247
– proton beam radiosurgery and 11
– stereotactic radiosurgery (SRS) – digital reconstructed radiographs –– clinical outcomes 245
for 146, 147–148 (RDDs) and 37 –– complications 247

262
Index

–– Mini-Mental Status Examination – couch-mounted 45 – classification of 160, 160 – See also Physiological epilepsy
(MMSE) 247 – floor-mounted 44, 45 –– Barrow Neurologic Institute (BNI) – corpus callosotomy 197
–– National Comprehensive Cancer – floor-stand 44, 45 Pain Intensty Scale 160, 160 – neocortical foci 197
Network (NCCN) concensus guide- – Gamma Knife 44 –– Burchiel 160, 160 Nonstandard fields 31
lines 247 – imaging-dependent 46 – clinical scenarios for 165 – See also Fields
–– neurocognitive outcomes 247 – linear accelerator (linac) frame sys- –– computed tomography (CT)-based Nonvestibular cranial nerve schwanno-
–– overviews of 243, 248 tems 44 treatment planning 165 mas (NVCNS) 137
–– references for 248 MLCs, see Multileaf collimators (MLCs) –– linear accelerator (linac)-based ap- – overviews of 137, 143
–– stereotactic radiosurgery (SRS) MMSE, see Mini-Mental Status Exami- proaches 165 – references for 144
for 243 nation (MMSE) –– repeat surgeries 165, 165 – stereotactic radiosurgery (SRS)
–– toxicities 247 Models 27 – overviews of 160, 165 for 138
–– treatment options for 243 – experimental 193 – pathophysiology of 160 –– complications of 143
–– whole-brain radiotherapy (WBRT) – linear-quadratic (LQ) 22 – population-specific considerations –– techniques 138, 140–142
for 246 – neuropathy 24 for 164 – treatment options for 138
– one to four 235 Modulation, dose 164 –– bilateral trigeminal neuralgia 164 –– complications of 142
–– complications of 240 Molecular agent combinations 259 –– multiple sclerosis (MS)-related tri- –– fractionated radiotherapy 141
–– diagnostic-specific Graded Prognos- Morphological appearance 61, 61 geminal neuralgia 164 –– microsurgery 137, 138, 139
tic Assessment (DS-GPA) and 235 Movement disorders 168 –– type II trigeminal neuralgia 164 Noren, George 7
–– dose prescriptions 239 – definition of 168 – references for 166 North American Gamma Knife Consor-
–– Graded Prognostic Assessment – future directions of 174 – stereotactic radiosurgery (SRS) tium (NAGKC) reports 129, 129, 130
(GPA) and 235 – overviews of 168, 174 for 162 Novel molecular agent combina-
–– Hopkins Verbal Learning Test – references for 175 –– clinical outcomes for 162 tions 259
(HVLT) 236, 240 – stereotactic radiosurgery (SRS) –– complications of 163 NVCNS, see Nonvestibular cranial nerve
–– hypofractionated stereotactic radio- for 168 –– dose modulation 164 schwannomas (NVCNS)
surgery (HRST) 235, 239 –– indications for 168 –– dose rate effects 164
–– immobilization systems 237 –– indirect targeting 169 –– isocenter location 163
–– Karnosky Performace Score (KPS) –– patient selection criteria 168 –– pain outcomes 162 O
and 240, 241 –– radiosurgical pallidotomy 172, 173 –– quality-of-life (QOL) and 162 OARs data, see Organs-at-risk (OARs)
–– Mini-Mental Status Examination ––– See also Pallidotomy –– response factors 162 data
(MMSE) and 236 –– radiosurgical subthalamotomy 174 –– targeting 163 Obsessive-compulsive disorder
–– neurocognitive dysfunction –– radiosurgical thalamotomy 169, –– treatment planning 163 (OCD) 177
and 240 170–172 – treatment options for 161 – comorbidities of 177
–– overviews of 235, 241 ––– See also Thalamotomy –– medical management 161, 161 – definition of 177
–– postoperative management 235 – treatment options for 168 –– percutaneous rhizotomy 162 – neuroanatomy of 177
–– prophylactic cranial whole-brain ra- –– deep brain stimulation (DBS) 169, Neuritis, optic 114 – overviews of 177, 179
diotherapy (PC-WBRT) and 240 172, 174, 174 Neuroanatomy 177 – pathophysiology of 177
–– Quality of Life Questionnaire –– radiofrequency (RF) ablation 168– Neurocognition 58, 197 – references for 180
(QLQ) 240 169 Neurocognitive dysfunction 240 – stereotactic radiosurgery (SRS) for
–– radiation necrosis and 240 – Tremor Rating Scale and 169 Neuropathology fundamentals 58 –– clinical outcomes 178
–– Radiation Therapy Oncology Group – Unified Parkinson’s Disease Rating – analyses 62 –– deep brain stimulatin (DBS)
(RTOG) recursive partitioning anal- Scale (UPDRS) and 168, 171, 174 – effectiveness 62 and 179
yses (RPA) and 235 MS-related tirgeminal neuralgia, see – historical perspectives of 58, 59 –– patient selection criteria 178
–– references for 241 Multiple sclerosis (MS)-related trige- – materials 59, 60 –– reviews of 180
–– reviews of 237 minal neuralgia – overviews of 58, 65 –– target selection criteria 105, 177
–– stereotactic radiosurgery (SRS) msr, see Machine-specific reference – references for 66 –– techniques 178
for 236 (msr) – results 60, 61 – treatment options for 177
–– surgical resections 235 MTLE, see Medial temporal lobe epi- –– acute-type reactions 60–61, 62 –– neurosurgery 177
–– treatment delivery systems 238 lepsy (MTLE) –– chronic-type/end-stage reac- –– pharmacotherapy 177
–– treatment planning 237 Multileaf collimators (MLCs) 49, 49 tions 61, 63 OCD, see Obsessive-compulsive disor-
–– whole-brain radiotherapy (WBRT) – See also Collimators –– enhanced apoptotic activities 62, der (OCD)
for 236 Multiplanar X-ray imaging 48 64 Ocular disorders 202
– pediatric 211 Multiple sclerosis (MS)-related trigemi- –– Factor-VIII immune reactions 64 – age-related macular degeneration
– repeat radiosurgery for 251 nal neuralgia 164 –– morphological appearance 11, 61 (ARMD) 207
–– American Society for Therapeutic – See also Trigeminal neuralgia –– subacute-type reactions 61, 63 –– choroidal neovascular membranes
Radiology and Oncology (ASTRO) Myelopathy, radiation 54 Neuropathy 24, 114 (CNVMs) and 207
National Concensus Guidelines Myokemia 54 Nomogrip TALON devices 31 –– choroidal neovascularization (CNV)
for 253 Nonfunctioning pituitary adeno- and 207
–– clinical experiences 252, 252, 253 mas 110 – exophthalmos refratory to connser-
–– decision-making criteria for 251, N – See also Pituitary adenomas vative treatments 207
255 NAGKC reports, see North American Noninvasive positioning systems 46 – glaucoma 206, 206
–– efficacy of 251 Gamma Knife Consortium (NAGKC) – See also Positioning systems – hemangioblastomas, ocular 207
–– overviews of 251, 253 reports – comparisons of 46 – overviews of 202, 208
–– references for 255 Nanoparticles 259 – immobilization 205 – rare 207
–– toxicity of 251 National Comprehensive Cancer Net- –– bite plates 47, 48–49 –– carcinomas, orbital/uveal metasta-
Micro-multileaf collimators (MLCs) 30, work (NCCN) 247 –– ear canal 47 ses 207
49 NCCN, see National Comprehensive –– thermoplastic masks 47, 47 –– rentinoblastomas 207
– See also Collimators Cancer Network (NCCN) – localization 48 – references for 208
Mini-Mental Status Examination Necrosis, radiation 55, 115 –– multiplanar X-ray imaging 48 – stereotactic radiosurgery (SRS)
(MMSE) 236, 247 – See also Radiation injuries –– optical tracking 48 for 202
Minimally-invasive positioning sys- Necrotic stage 58 –– surface monitoring 48 –– dosimetry 203
tems 43 Neocortical foci 197 Nonisocentric vs. isocentric techni- –– eye fixation 202
– See also Positioning systems Neuralgia, trigeminal 160 ques 34 –– technical issues 202
– comparisons of 43 – as suicide disease 160 Nonlesional epilepsy 197 –– treatment planning 203

263
Index

– uveal melanomas 203 Pharmacotherapy 177 –– features of 33 – minimally-invasive 43


–– clinical manifestations of 203, 205 Physics fundamentals 30 –– forward vs. inverse techniques 33 –– comparisons of 43
–– complications of 204 – beam shaping and delivery 33 –– gross tumor volumes (GTVs) 35 –– couch-mounted 45
–– Late Effects Normal Tissues, subjec- – clinical implementations and 39 –– isocentric vs. nonisocentric techni- –– floor-mounted 44, 45
tive, objective, management, and – detectors 32 ques 33, 34 –– floor-stand 44, 45
analytic (LENT SOMA) scoring sys- –– American Association of Physicists –– planning target volumes (PTVs) 33, –– Gamma Knife 44
tem 204, 204 in Medicine (AAPM) protocols 32 35 –– imaging-dependent 46
–– natural history of 203 –– International Atomic Energy Agency –– steep dose falloffs 33 –– linac frame systems 44
–– prognoses for 205 (IAEA) protocols 32 Physiological epilepsy 195 – noninvasive 46
–– reviews of 204, 204 – fields 30 – See also Epilepsy –– comparisons of 46
–– treatment decision criteria 205 –– charged particle equilibrium – clinical outcomes for 195 –– immobilization 47
– von Hippel-Lindau disease 207 (CPE) 32 – medial temporal lobe epilepsy ––– See also Immobilization
Olivecrona, Herbet 2, 2, 3 –– effects of 32 (MTLE) 195 Pre-surgical evaluations 107
One to four metastases 235 –– machine-specific reference – morbidities of 196 Preradiosurgical embolization 77
– See also Metastases (msr) 32 – neurocognition and 197 Prophylactic cranial whole-brain radio-
Open surgical procedures 186 –– nonstandard 31–32 – other nonlesional 197 therapy (PC-WBRT) 240
Opthalmic-related disorders, see Ocular –– plan-class-specific reference –– corpus callosotomy 197 PTVs, see Planning target volumes
disorders (pcsr) 32 –– neocortical foci 197 (PTVs)
Optic nerve tumors 226 –– problems of 31 – secondary outcomes of 197
Optic neuritis 114 –– small 30–32 – seizure remission for 195, 195, 196
Optic pathway, dose tolerance of 54 – high-dose gradients 30 – treatment protocols for 196 Q
Optical guidance 37 – image guidance techniques 36 Pilocytic astrocytomas (PAs) 226, 228 QA, see Quality assurance (QA) and
Optical tracking 48 –– comparisons of 36 Pineal tumors 220 safety
Orbital carcinomas 207 –– cone-beam computed tomography Pituitary adenomas 107 QLQ, see Quality of Life Questionnaire
Organs-at-risk (OARs) data 55, 55 (CBCT) 37 – overviews of 107, 117 (QLQ)
– See also Critical structures –– digital reconstructed radiographs – pediatric 219 QOL considerations, see Quality-of-life
(RDDs) and 37 – prognoses for 115 (QOL) consideratons
–– grantry-mounted vs. room- – references for 117 Quality assurance (QA) and safety 38
P mounted systems 37 – special considerations for 115 – end-to-end tests 38
Pain scales 160, 160 –– infrared 37 – sterotactic radiosurgery (SRS) – hidden target tests 38
Pallidotomy 172 –– kilovoltage (kV) S-ray tubes 37 for 108 – importance of 38
– complications of 173 –– magnetic-resonance (MR)-based 37 –– clinical outcomes, acromegaly 111, – Winston-Lutz tests 38
– for movement disorders 172 –– optical 37 113–114 Quality of Life Questionnaire
–– See also Movement disorders –– stereoscopic 2d X-ray imaging 37 –– clinical outcomes, Cushing dis- (QLQ) 240
– outcomes of 173 –– volumetric 3D X-ray imaging 37 ease 111, 111–112 Quality-of-life (QOL) consideratons 162
– overviews of 174 – immobilization techniques 31 –– clinical outcomes, functioning pitui-
– reviews of 173 –– frameless 31 tary adenomas 110, 111–114 QUANTEC report, see Quantitative
– unilateral vs. bilateral 172 –– frames 31 –– clinical outcomes, nonfunctioning Analysis of Normal Tissue Effects in
– vs. radiofrequency (RF) pallidoto- –– frames, relocatable 31 pituitary adenomas 110 the Clinic (QUANTEC) report
my 173 –– masks 31 –– complications of 112 Quantitative Analysis of Normal Tissue
Parkinson disease (PD) 168 – localization, precision/accuracy –– cranial neuropathy and 114 Effects in the Clinic (QUANTEC) re-
– See also Movement disorders of 30 –– critical structures, dose limitations port 52
PAs, see See Pilocytic astrocytomas – margins 35 to 109
(PAs) –– dose conformity vs. inhomogene- –– dose selection criteria 108
PC-WBRT, see Prophylactic cranial ity 35, 35, 36 –– efficacy of 116 R
whole-brain radiotherapy (PC- –– eclipse plans 35, 36, 36 –– frame placement 108
Radiation injuries, see Complications
WBRT) –– hot spots 35 –– histological entities and 116
– acute 112
pcsr, see Plan-class-specifc reference –– margin assumption 35 –– historical perspectives of 107
– early delayed/subacute 52, 114
(pcsr) –– multimodality imaging registra- –– hypopituitarism and 114
– late delayed 115
PD, see Parkinson disease (PD) tion 36 –– hypothalamic dysfunction and 114
– myelopathy 54
Pediatric brain tumors 210 –– planning target volumes (PTVs) 35 –– optic neuritis and 114
– necrosis 115
– overviews of 210, 221 –– prescription isodose spillover 35 –– patient selection criteria 107
Radiation mechanisms 193
– references for 222 – overviews of 30 –– pre-surgical evaluations 107
Radiation Therapy Oncology Group
– technical issues 210, 210 – physical principles 30 –– principles of 108
(RTOG) 229, 235
– treatment options 211 – quality assurance (QA) and safety 38 –– radiation injuries, acute 112
Radiobiology 20
–– boost therapy 211 –– end-to-end tests 38 –– radiation injuries, early de-
– fractionation 20
–– complications of 221 –– hidden target tests 38 layed 114
–– effects estimate calculations 22, 22
–– for astrocytomas, low-grade 213, –– importance of 38 –– radiation injuries, late delayed 115
–– high-dose single fractions 20
214–215 –– Winston-Lutz tests 38 –– radiation necrosis and 115
–– limitations of 23
–– for craniopharyngiomas 217, 219 – references for 39 –– radiographic images 108
–– multiple small-dose fractions 20
–– for ependymomas 211, 212–213 – small fields 30 –– techniques 108
– future directions of 27
–– for gliomas, high-grade 215 – stereotactic radiosurgery/stereotac- –– treatment planning 108, 109
– models 27
–– for medulloblastomas 215 tic radiotherapy (SRS/SRT) pro- –– whole-sellar 116
–– linear-quadratic (LQ) 22
–– for meningiomas 216, 216 grams 32, 39 – treatment options for 115
–– neuropathy 24
–– for pineal tumors 220 –– –– antisecreting medications, radiore-
– optic nerve tolerance doses 23
–– for pituitary adenomas 219 –– clinial implementation of 39 sistant effects of 116
– overviews of 20
–– for recurrent disease, salvage ther- –– comparisons of 30 –– external beam radiation therapy
– parameter assessments 24, 24
apy 211 –– proton 32 (EBRT) 115
– radiation injury reactions 26, 26
–– for schwannomas, vestibular 216, –– training for 39 –– up-front radiosurgery 116
– Radiation Therapy Oncology Group
217 – technical challenges 31 Plan-class-specific reference (pcsr) 32
(RTOG) Dose-Escalation Study 27, 27
–– stereotactic radiosurgery (SRS) as – treatment planning 33 Planning target volumes (PTVs) 35
– references for 28
sole postoperative treatment 213 –– challenges of 33 Positioning systems 43
– tissue radiation mechanisms 21, 21
Percutaneous rhizotomy 162 –– clinical target volumes (CTVs) 35 – See also Devices

264
Index

–– dose-response curves 21, 21 – efficacy of 251 – for brain metastases, see Brain meta- Superficial/hemispheric cerebral caver-
–– therapeutic ratio 21 – for arteriovenous malformations stases nomas (CCMs) 81
–– therapeutic window 21 (AVMs) 76 –– five or more metastases 243 – See also Cerebral cavernomas (CCMs)
– tumor hypoxia 27, 28 –– See also Arteriovenous malforma- –– one to four metastases 235 Surface monitoring 48
Radiofrequency (RF) ablation 168–169 tion (AVMs) –– repeat radiosurgery 251 Surgical resections 128, 146, 235
Radiofrequency (RF) pallidotomy 173 – overviews of 251, 253 – for cerebral cavernomas (CCMs) 81 Sweet, William 12
Radioresistant vs. radiosensitive – references for 255 – for chondrosarcomas 128 Syndrome of inappropriate antidiuretic
cells 193 – toxicity of 251 – for chordomas 120 secretion (SIADH) 186
Radiosurgery, see Stereotactic radiosur- Resections, surgical 128, 146, 235 – for dural arteriovenous fistulas
gery (SRS) Resorption stage 58 (DAVFs) 91
– radiosurgical pallidotomy 172 RF ablation, see Radiofrequency (RF) – for epilepsy 193 T
–– complications of 173 ablation – for glial tumors 226 Tallairach, Jean 3
–– for movement disorders 172 Rhizotomy, percutaneous 162 – for glomus tumors 132 TALON frames 31
––– See also Movement disorders RPA, see Recursive partitioning analyses – for hemangioblastomas 146 Thalamotomy, radiosurgical 169
–– outcomes of 173 (RPA) – for hypolthalamic hamartomas Therapeutic ratio 21
–– overviews of 174 RT, see Radiotherapy (RT) (HHs) 182 Therapeutic window 21
–– reviews of 173 RTOG, see Radiation Therapy Oncology – for meningiomas 102 Thermoplastic masks 47, 47
–– techniques 172 Group (RTOG) – for movement disorders 168 Tic douloureux 160
–– unilateral vs. bilateral 172 Rutherford, Ernest 11 – for obsessive-compulsive disorder – See also Trigeminal neuralgia
–– vs. radiofrequency (RF) 173 (OCD) 177 Tissue radiation mechanisms 21, 21
– radiosurgical subthalamotomy 174 – for ocular disorders 202 Tobias, Cornelius A. 11
–– complications of 174 S – for pediatric brain tumors 210 Tremor Rating Scale 169
–– for movement disorders 174 Salorio, Juan Barcia 8 – for pituitary adenomas 107 Tremor-dominant Parkinson disease
––– See also Movement disorders Salvage therapy 211 – for schwannomas 137, 150 (PD) 169
–– overviews of 174 Schwannomas 137, 150 –– nonvestibular cranial nerve – See also Movement disorders
–– subthalamic nucleus (STN) tar- – nonvestibular cranial nerve (NVCNs) 137 Trigeminal neuralgia 160
gets 174 (NVCNS) 137 –– vestibular (VSs) 150 – as suicide disease 160
– radiosurgical thalamotomy 169 –– complications of 142–143 – for trigeminal neuralgia 160 – classification of 160, 160
–– complications of 171 –– fractionated radiotherapy 141 – future directions of 258 –– Barrow Neurologic Institute (BNI)
–– efficacy of 171 –– microsurgery 137, 138, 139 – historical perspectives of 58 Pain Intensty Scale 160, 160
–– for movement disorders 169 –– overviews of 137, 143 –– See also Historical perspectives –– Burchiel 160, 160
––– See also Movement disorders –– references for 144 –– CyberKnife 16 – clinical scenarios for 165
–– outcomes of 170 –– stereotactic radiosurgery (SRS) –– Gamma Knife 2 –– computed tomography (CT)-based
–– overviews of 169, 172 for 138 –– linear accelerator (linac) radiosur- treatment planning 165
–– reviews of 171 –– treatment options for 138 gery 11 –– linear accelerator (linac)-based ap-
–– targeting 169 – vestibular (VSs) 150 –– proton beam radiosurgery 11 proaches 165
––– See also indirect, ventralis inter- –– historical perspectives of 150 – neuropathology of 58 –– repeat surgeries 165, 165
medius (VIM) targets –– overviews of 150, 155 – physics of 20, 30 – overviews of 160, 165
–– techniques 169 –– references for 156 – radiobiology of 20 – pathophysiology of 160
–– timing of 171 –– stereotactic radiosurgery (SRS) – schwannomas 137 – population-specific considerations
– repeat 251 for 151 – tolerance, central nervous system for 164
–– American Society for Therapeutic –– treatment options for 150 (CNS) 52 –– bilateral trigeminal neuralgia 164
Radiology and Oncology (ASTRO) Seizure remission 195, 195, 196 Stereotactic radiosurgery/stereotactic –– multiple sclerosis (MS)-related tri-
National Concensus Guidelines Sensorineural hearing loss (SNHL) 54 radiotherapy (SRS/SRT) pro- geminal neuralgia 164
for 253 Shrinking field methods 229 grams 32, 39 –– type II trigeminal neuralgia 164
–– decision-making criteria for 251 SIADH, see Syndrome of inappropriate Stereotactic, definition of 43 – references for 166
–– efficacy of 251 antidiuretic secretion (SIADH) STN targets, see Subthalamic nucleus – stereotactic radiosurgery (SRS)
–– overviews of 251, 253 Sinus recanalization 97 (STN) targets for 162
–– references for 255 Small fields 30 Stone, Robert 11 –– clinical outcomes for 162
–– toxicity of 251 SNHL, see Sensorineural hearing loss Street Jabez C. 12 –– complications of 163
– repeat radiosurgery (SNHL) Structures, critical 52 –– dose modulation 164
–– clinical experiences 252, 252, 253 Spinal cord, dose tolerance of 54 – See also Critical structures –– dose rate effects 164
–– decision-making criteria for 255 SRS, see Stereotactic radiosurgery (SRS) Sturm, Volker 8 –– isocenter location 163
– up-front 116 SRS/SRT programs, see Stereotactic ra- Subacute radiation injuries, see Early –– pain outcomes 162
Radiotherapy (RT) 128 diosurgery/stereotactic radiotherapy delayed/subacute radiation injuries –– quality-of-life (QOL) and 162
Rähn, Tiit 7 (SRS/SRT) programs Subthalamic nucleus (STN) targets 174 –– response factors 162
Rand, Robert 7 STAR system, see Stereotactic Align- Subthalamotomy, radiosurgical 174 –– targeting 163
RDDs, see Digital reconstructed radio- ment for Radiosurgery (STAR) sys- – complications of 171, 174 –– treatment planning 163
graphs (RDDs) tem – efficacy of 171 – treatment options for 161
Recanalization, sinus 97 Steep dose falloffs 33 – for movement disorders 169, 174 –– medical management 161, 161
Recursive partitioning analyses Steiner, Ladislau 7 –– See also Movement disorders –– percutaneous rhizotomy 162
(RPA) 235 Stereotactic Alignment for Radiosur- – outcomes of 170
Reichert, Traugott 3 gery (STAR) system 12, 13 – overviews of 169, 172, 174
Relocatable frames 31 Stereotactic radiosurgery (SRS) 20, 30, – reviews of 171 U
Rentinoblastomas 207 42, 58, 58 – subthalamic nucleus (STN) tar-
Unified Parkinson’s Disease Rating
Repeat radiosurgery 251 – critical structures and 52 gets 174
Scale (UPDRS) 168, 169, 174
– See also See also Metastases – definition of 42, 58 – targeting 169
Unilateral vs. bilateral pallidotomy 172
– American Society for Therapeutic Ra- – devices for 42 –– indirect 169
Up-front radiosurgery 116
diology and Oncology (ASTRO) Na- –– CyberKnife 16 –– ventralis intermedius (VIM) tar-
UPDRS, see Unified Parkinson's Disease
tional Concensus Guidelines for 253 –– GammaKnife 2 gets 169–170, 170, 172
Rating Scale (UPDRS)
– clinical experiences 252, 252, 253 – for arteriovenous malformations – techniques 169
Uveal carcinomas 207
– decision-making criteria for 251, (AVMs) 68 – timing of 171
255 Suicide diseases 160

265
Index

–– efficacy of 154 Veterinary-human radiation oncology


V –– large Koos stage IV manage- parallels 259
W
Ventralis intermedius (VIM) tar- ment 154, 156 VIM targets, see Ventralis intermedius Wait-and-see strategies 150
gets 169–170, 170, 172 –– reviews of 152 (VIM) targets WBRT, see Whole-brain radiotherapy
Vestibular schwannomas (VSs) 150 –– Timone Hospital prospective co- VMAT, see Volume-modulated arc ther- (WBRT)
– See also Schwannomas hort 143, 153 apy (VMAT) Whole-brain radiotherapy
– historical perspectives of 150 – treatment options for 150 Volume-modulated arc therapy (WBRT) 236, 246, 251
– overviews of 150, 155 –– clinical interviews 150 (VMAT) 30 Whole-sellar sterotactic radiosurgery
– references for 156 –– follow-ups 150 Volumetric 3D X-ray imaging 37 (SRS) 116
– stereotactic radiosurgery (SRS) –– wait-and-see strategies 150 von Hippel-Lindau disease 207 Wilson, Robert A. 11, 12
for 151 Winston-Lutz tests 38
–– complications of 154 Wycis, Henry 3

266

You might also like